1
|
Tsuruoka M, Tokizaki H, Yamasu K. Definition of the characteristic neurogenesis pattern in the neural plate by the Hes1 orthologue gene, her6, during early zebrafish development. Cells Dev 2025:204026. [PMID: 40228713 DOI: 10.1016/j.cdev.2025.204026] [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: 12/12/2024] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
During vertebrate embryonic development, a distinctive, spotty neurogenesis pattern emerges in the early neural plate, which represents proneural clusters. The determination of this pattern depends on the interaction between proneural genes and bHLH-O-type transcription factor (TF) genes, Hes/her, which suppress neurogenesis. In this study, we focused on the mouse Hes1 orthologue, her6, to understand the mechanism that controls neurodevelopmental patterns in the developing brain in zebrafish (Danio rerio). We first assessed the expression pattern of her6 in the neural plate, observing that it is consistently expressed in the entire forebrain throughout somitogenesis, including her9 expression within it. Meanwhile, the expression patterns of her6 changed dynamically in the hindbrain, in contrast to the Notch-independent her genes. The expression pattern was not significantly affected by forced NICD expression and DAPT treatment at the bud stage, showing that her6 expression is Notch-independent in the neural plate at this stage. To analyze the roles of her6, we disrupted her6 using the CRISPR/Cas9 method. The mutants thus obtained showed a deformed midbrain-hindbrain region and failed to grow to adulthood. At the bud stage, ectopic expression of neurogenesis-related genes was observed in her6 mutants in specific regions of the neural plate, where neurogenesis does not occur and which are considered neural progenitor pools (NPPs) in wild-type embryos. Of note, no other Notch-independent her genes are known to be expressed in these NPP regions. In contrast, the expression of regionalization genes in the forebrain and hindbrain was not affected in her6 mutants. These findings suggest that her6 defines the primary neurogenesis pattern in the neural plate, together with other Notch-independent her genes.
Collapse
Affiliation(s)
- Momo Tsuruoka
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Hiroki Tokizaki
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan.
| |
Collapse
|
2
|
Erofeeva N, Galstyan DS, Yang L, Strekalova T, Lim LW, de Abreu MS, Golushko NI, Stewart AM, Kalueff AV. Developing zebrafish models of Notch-related CNS pathologies. Neurosci Biobehav Rev 2025; 170:106059. [PMID: 39929383 DOI: 10.1016/j.neubiorev.2025.106059] [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: 09/10/2024] [Revised: 02/01/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Notch signaling is an evolutionarily conserved cellular pathway that regulates various stem cell functions, including fate determination, differentiation, proliferation, and apoptosis. This crucial signaling mechanism also plays an important role in the brain, regulating neurogenesis, cell differentiation, and homeostasis, whereas disrupted Notch signaling is linked to various neurodegenerative diseases and brain cancers. Here, we review the central nervous system (CNS) pathologies associated with aberrant Notch signaling, and summarize the available experimental (animal) models used to study these pathologies, with a special focus on zebrafish (Danio rerio). As genetic, pharmacological, and behavioral models in zebrafish have significantly advanced our understanding of Notch-related CNS disorders, future research is expected to further link Notch signaling to brain disorders and, eventually, lead to their more specific and targeted therapeuties.
Collapse
Affiliation(s)
- Natalia Erofeeva
- St. Petersburg State University, St Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia
| | - Longen Yang
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Tatiana Strekalova
- Peoples Friendship University of Russia (RUDN University) and Department of Normal Physiology, Sechenov University, Moscow, Russia
| | - Lee Wei Lim
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Municipal Key Laboratory on Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Western Caspian University, Baku, Azerbaijan.
| | - Nikita I Golushko
- Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia
| | - Adam Michael Stewart
- The International Zebrafish Neuroscience Research Consortium (ZNRC), New Orleans, USA
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St Petersburg, Russia; Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Municipal Key Laboratory on Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Neurobiology Program, Sirius University of Science and Technology, Sirius, Russia.
| |
Collapse
|
3
|
Gu C, Wang Z, Luo W, Ling H, Cui X, Deng T, Li K, Huang W, Xie Q, Tao B, Qi X, Peng X, Ding J, Qiu P. Impaired olfactory bulb neurogenesis mediated by Notch1 contributes to olfactory dysfunction in mice chronically exposed to methamphetamine. Cell Biol Toxicol 2025; 41:46. [PMID: 39976779 PMCID: PMC11842540 DOI: 10.1007/s10565-025-10004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
Methamphetamine (Meth) is a potent central nervous system stimulant with high addictive potential and neurotoxic effects. Chronic use results in significant damage in various brain functions, including cognition, memory, and sensory perception. Olfactory dysfunction is a notable yet often overlooked consequence of Meth abuse, and its underlying mechanisms are not fully understood. This study investigates the mechanisms of Meth-induced olfactory impairment through a thorough examination of olfactory bulb (OB) neurogenesis. We found that chronic Meth abuse impaired olfactory function in mice by not only reducing the self-renewal of subventricular zone (SVZ) neural stem cells (NSCs) but also altering their differentiation potential, leading their differentiation into astrocytes at the expense of neurons. Mechanistically, Meth inhibits autophagosome-lysosome fusion by downregulating Syntaxin 17 (Stx17), which reduces autophagic flux. In NSCs, autophagy tightly regulates Notch1 levels, and impaired autophagic degradation of Notch1 leads to its abnormal activation. This alters NSCs fate determination, ultimately affecting OB neurogenesis. Our study reveals that Meth impairs olfactory function through autophagic dysfunction and aberrant Notch1 signaling. Understanding these mechanisms not only provides new insights into Meth-induced olfactory dysfunction but also offers potential targets for developing therapies to alleviate Meth-induced neurotoxicity and sensory damage in the future.
Collapse
Affiliation(s)
- Cihang Gu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhuo Wang
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wenyu Luo
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Haosen Ling
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xilie Cui
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Tongtong Deng
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Kuan Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Wei Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qiqian Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Bowen Tao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, 550004, China
| | - Xiaojia Peng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jiuyang Ding
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, 550004, China.
| | - Pingming Qiu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China.
- Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
4
|
Azargoonjahromi A, Abutalebian F, Hoseinpour F. The role of resveratrol in neurogenesis: a systematic review. Nutr Rev 2025; 83:e257-e272. [PMID: 38511504 DOI: 10.1093/nutrit/nuae025] [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: 03/22/2024] Open
Abstract
CONTEXT Resveratrol (RV) is a natural compound found in grapes, wine, berries, and peanuts and has potential health benefits-namely, neurogenesis improvement. Neurogenesis, which is the process through which new neurons or nerve cells are generated in the brain, occurs in the subventricular zone and hippocampus and is influenced by various factors. RV has been shown to increase neural stem cell proliferation and survival, improving cognitive function in hippocampus-dependent tasks. Thus, to provide a convergent and unbiased conclusion of the available evidence on the correlation between the RV and neurogenesis, a systematic review needs to be undertaken meticulously and with appropriate attention. OBJECTIVE This study aimed to systematically review any potential connection between the RV and neurogenesis in animal models. DATA SOURCES AND EXTRACTION Based on the particular selection criteria, 8 original animal studies that investigated the relationship between RV and neurogenesis were included. Studies written in English and published in peer-reviewed journals with no restrictions on the starting date of publication on August 17, 2023, were searched in the Google Scholar and PubMed databases. Furthermore, data were extracted and analyzed independently by 2 researchers and then reviewed by a third researcher, and discrepancies were resolved by consensus. This project followed PRISMA reporting standards. DATA ANALYSIS In the studies analyzed in this review, there is a definite correlation between RV and neurogenesis, meaning that RV intake, irrespective of the mechanisms thereof, can boost neurogenesis in both the subventricular zone and hippocampus. CONCLUSION This finding, albeit with some limitations, provides a plausible indication of RV's beneficial function in neurogenesis. Indeed, RV intake may result in neurogenesis benefits-namely, cognitive function, mood regulation, stress resilience, and neuroprotection, potentially preventing cognitive decline.
Collapse
Affiliation(s)
| | - Fatemeh Abutalebian
- Department of Biotechnology and Medicine, Islamic Azad University of Tehran Central Branch, Tehran, Iran
| | - Fatemeh Hoseinpour
- Department of Occupational Therapy, Semnan University of Medical Sciences and Health Services, Semnan, Iran
| |
Collapse
|
5
|
Farahani RM. Neural differentiation in perspective: mitochondria as early programmers. Front Neurosci 2025; 18:1529855. [PMID: 39844856 PMCID: PMC11751005 DOI: 10.3389/fnins.2024.1529855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Neural differentiation during development of the nervous system has been extensively studied for decades. These efforts have culminated in the generation of a detailed map of developmental events that appear to be associated with emergence of committed cells in the nervous system. In this review the landscape of neural differentiation is revisited by focusing on abiotic signals that play a role in induction of neural differentiation. Evidence is presented regarding a chimeric landscape whereby abiotic signals generated by mitochondria orchestrate early events during neural differentiation. This early stage, characterised by mitochondrial hyperactivity, in turn triggers a late stage of differentiation by reprogramming the activity of biotic signals.
Collapse
Affiliation(s)
- Ramin M. Farahani
- IDR/Research and Education Network, Westmead, NSW, Australia
- Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
6
|
He L, Bhat K, Ioannidis A, Pajonk F. Effects of dopamine receptor antagonists and radiation on mouse neural stem/progenitor cells. Radiother Oncol 2024; 201:110562. [PMID: 39341503 PMCID: PMC11987595 DOI: 10.1016/j.radonc.2024.110562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Dopamine receptor antagonists have recently been identified as potential anti-cancer agents in combination with radiation, and a first drug of this class is in clinical trials against pediatric glioma. Radiotherapy causes cognitive impairment primarily by eliminating neural stem/progenitor cells and subsequent loss of neurogenesis, along with inducing inflammation, vascular damage, and synaptic alterations. Here, we tested the combined effects of dopamine receptor antagonists and radiation on neural stem/progenitor cells. METHODS Using transgenic mice that report the presence of neural stem/progenitor cells through Nestin promoter-driven expression of EGFP, the effects of dopamine receptor antagonists alone or in combination with radiation on neural stem/progenitor cells were assessed in sphere-formation assays, extreme limiting dilution assays, flow cytometry and real-time PCR in vitro and in vivo in both sexes. RESULTS We report that hydroxyzine and trifluoperazine exhibited sex-dependent effects on murine newborn neural stem/progenitor cells in vitro. In contrast, amisulpride, nemonapride, and quetiapine, when combined with radiation, significantly increased the number of neural stem/progenitor cells in both sexes. In vivo, trifluoperazine showed sex-dependent effects on adult neural stem/progenitor cells, while amisulpride demonstrated significant effects in both sexes. Further, amisulpride increased sphere forming capacity and stem cell frequency in both sexes when compared to controls. CONCLUSION We conclude that a therapeutic window for dopamine receptor antagonists in combination with radiation potentially exists, making it a novel combination therapy against glioblastoma. Normal tissue toxicity following this treatment scheme likely differs depending on age and sex and should be taken into consideration when designing clinical trials.
Collapse
Affiliation(s)
- Ling He
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States.
| | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States; Jonsson Comprehensive Cancer Center at UCLA, United States; Department of Neurosurgery, David Geffen School of Medicine at UCLA, United States
| |
Collapse
|
7
|
Weston NM, Green JC, Keoprasert TN, Sun D. Dendritic morphological development of traumatic brain injury-induced new neurons in the dentate gyrus is important for post-injury cognitive recovery and is regulated by Notch1. Exp Neurol 2024; 382:114963. [PMID: 39303845 PMCID: PMC11502241 DOI: 10.1016/j.expneurol.2024.114963] [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: 07/09/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Traumatic brain injury (TBI) is a prevalent problem with survivors suffering from chronic cognitive impairments. Following TBI there is a series of neuropathological changes including neurogenesis. It is well established that neurogenesis in the dentate gyrus (DG) of the hippocampus is important for hippocampal dependent learning and memory functions. Following TBI, injury-enhanced hippocampal neurogenesis is believed to contribute to post-injury cognitive recovery. Behavioral function is connected to synaptic plasticity and neuronal dendritic branching is critical for successful synapse formation. To ascertain the functional contribution of injury-induced DG new neurons in post-TBI cognitive recovery, it is necessary to study their dendritic morphological development and the molecular mechanisms controlling this process. Utilizing transgenic mice with tamoxifen-induced GFP expression and Notch1 knock-out in nestin+ neural stem cells, this study examined dendritic morphology, the role of Notch1 in regulating dendritic complexity of injury-induced DG new neurons, and their association to post-TBI cognitive recovery. We found that at 8 weeks after a moderate TBI, injury-induced DG new neurons in the injured control mice displayed a similar dendritic morphology as the cells in non-injured mice accompanied with cognitive recovery. In comparison, in Notch1 conditional knock-out mice, DG new neurons in the injured mice had a significant reduction in dendritic morphological development including dendritic arbors, volume span, and number of branches in comparison to the cells in non-injured mice concomitant with persistent cognitive dysfunction. The results of this study confirm the importance of post-injury generated new neurons in cognitive recovery following TBI and the role of Notch1 in regulating their maturation process.
Collapse
Affiliation(s)
- Nicole M Weston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States
| | - Jakob C Green
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States
| | - Timothy N Keoprasert
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States
| | - Dong Sun
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States.
| |
Collapse
|
8
|
Chouly M, Bally-Cuif L. Generating neurons in the embryonic and adult brain: compared principles and mechanisms. C R Biol 2024; 347:199-221. [PMID: 39535540 DOI: 10.5802/crbiol.167] [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: 08/31/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Neurogenesis is a lifelong process, generating neurons in the right amount, time and place and with the correct identity to permit the growth, function, plasticity and repair of the nervous system, notably the brain. Neurogenesis originates from neural progenitor cells (NPs), endowed with the capacity to divide, renew to maintain the progenitor population, or commit to engage in the neurogenesis process. In the adult brain, these progenitors are classically called neural stem cells (NSCs). We review here the commonalities and differences between NPs and NSCs, in their cellular and molecular attributes but also in their potential, regulators and lineage, in the embryonic and adult brains. Our comparison is based on the two most studied model systems, namely the telencephalon of the zebrafish and mouse. We also discuss how the population of embryonic NPs gives rise to adult NSCs, and outstanding questions pertaining to this transition.
Collapse
|
9
|
Man KH, Wu Y, Gao Z, Spreng AS, Keding J, Mangei J, Boskovic P, Mallm JP, Liu HK, Imbusch CD, Lichter P, Radlwimmer B. SOX10 mediates glioblastoma cell-state plasticity. EMBO Rep 2024; 25:5113-5140. [PMID: 39285246 PMCID: PMC11549307 DOI: 10.1038/s44319-024-00258-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 11/10/2024] Open
Abstract
Phenotypic plasticity is a cause of glioblastoma therapy failure. We previously showed that suppressing the oligodendrocyte-lineage regulator SOX10 promotes glioblastoma progression. Here, we analyze SOX10-mediated phenotypic plasticity and exploit it for glioblastoma therapy design. We show that low SOX10 expression is linked to neural stem-cell (NSC)-like glioblastoma cell states and is a consequence of temozolomide treatment in animal and cell line models. Single-cell transcriptome profiling of Sox10-KD tumors indicates that Sox10 suppression is sufficient to induce tumor progression to an aggressive NSC/developmental-like phenotype, including a quiescent NSC-like cell population. The quiescent NSC state is induced by temozolomide and Sox10-KD and reduced by Notch pathway inhibition in cell line models. Combination treatment using Notch and HDAC/PI3K inhibitors extends the survival of mice carrying Sox10-KD tumors, validating our experimental therapy approach. In summary, SOX10 suppression mediates glioblastoma progression through NSC/developmental cell-state transition, including the induction of a targetable quiescent NSC state. This work provides a rationale for the design of tumor therapies based on single-cell phenotypic plasticity analysis.
Collapse
Affiliation(s)
- Ka-Hou Man
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, 201210, Shanghai, China
| | - Zhenjiang Gao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Anna-Sophie Spreng
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Johanna Keding
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jasmin Mangei
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pavle Boskovic
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan-Philipp Mallm
- Single-Cell Open Lab, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hai-Kun Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, 201210, Shanghai, China
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Charles D Imbusch
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernhard Radlwimmer
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
10
|
Foley T, Thetiot M, Bally-Cuif L. Neural Stem Cell Regulation in Zebrafish. Annu Rev Genet 2024; 58:249-272. [PMID: 39121542 DOI: 10.1146/annurev-genet-111523-101949] [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: 08/12/2024]
Abstract
Neural stem cells (NSCs) are progenitor cell populations generating glial cells and neurons and endowed with long-lasting self-renewal and differentiation potential. While some neural progenitors (NPs) in the embryonic nervous system are also long-lived and match this definition, the term NSC classically refers to such progenitor types in the adult. With the discovery of extensive NSC populations in the adult brain of Danio rerio (zebrafish) and of their high neurogenic activity, including for neuronal regeneration, this model organism has become a powerful tool to characterize and mechanistically dissect NSC properties. On these bases, this article will consider NSCs in the adult zebrafish brain, with a focus on its most extensively characterized domain, the telencephalon (notably its dorsal part, the pallium). Whenever necessary, we will also refer to other brain subdivisions, embryonic processes, and the mouse adult brain, whether for comparative purposes or because more information is available in these other systems.
Collapse
Affiliation(s)
- Tanya Foley
- Zebrafish Neurogenetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Paris, France; , ,
| | - Melina Thetiot
- Zebrafish Neurogenetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Paris, France; , ,
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Paris, France; , ,
| |
Collapse
|
11
|
Todorov H, Weißbach S, Schlichtholz L, Mueller H, Hartwich D, Gerber S, Winter J. Stage-specific expression patterns and co-targeting relationships among miRNAs in the developing mouse cerebral cortex. Commun Biol 2024; 7:1366. [PMID: 39433948 PMCID: PMC11493953 DOI: 10.1038/s42003-024-07092-7] [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: 05/14/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
microRNAs are crucial regulators of brain development, however, miRNA regulatory networks are not sufficiently well characterized. By performing small RNA-seq of the mouse embryonic cortex at E14, E17, and P0 as well as in neural progenitor cells and neurons, here we detected clusters of miRNAs that were co-regulated at distinct developmental stages. miRNAs such as miR-92a/b acted as hubs during early, and miR-124 and miR-137 during late neurogenesis. Notably, validated targets of P0 hub miRNAs were enriched for downregulated genes related to stem cell proliferation, negative regulation of neuronal differentiation and RNA splicing, among others, suggesting that miRNAs are particularly important for modulating transcriptional programs of crucial factors that guide the switch to neuronal differentiation. As most genes contain binding sites for more than one miRNA, we furthermore constructed a co-targeting network where numerous miRNAs shared more targets than expected by chance. Using luciferase reporter assays, we demonstrated that simultaneous binding of miRNA pairs to neurodevelopmentally relevant genes exerted an enhanced transcriptional silencing effect compared to single miRNAs. Taken together, we provide a comprehensive resource of miRNA longitudinal expression changes during murine corticogenesis. Furthermore, we highlight several potential mechanisms through which miRNA regulatory networks can shape embryonic brain development.
Collapse
Affiliation(s)
- Hristo Todorov
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stephan Weißbach
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Developmental Biology and Neurobiology (iDN), Johannes Gutenberg University Mainz, Mainz, Germany
| | - Laura Schlichtholz
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Focus Program of Translational Neurosciences, University Medical Center Mainz, Mainz, Germany
| | - Hanna Mueller
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dewi Hartwich
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Jennifer Winter
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
12
|
Fogli M, Nato G, Greulich P, Pinto J, Ribodino M, Valsania G, Peretto P, Buffo A, Luzzati F. Dynamic spatiotemporal activation of a pervasive neurogenic competence in striatal astrocytes supports continuous neurogenesis following injury. Stem Cell Reports 2024; 19:1432-1450. [PMID: 39303706 PMCID: PMC11561465 DOI: 10.1016/j.stemcr.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Adult neural stem cells (NSCs) are conventionally regarded as rare cells restricted to two niches: the subventricular zone (SVZ) and the subgranular zone. Parenchymal astrocytes (ASs) can also contribute to neurogenesis after injury; however, the prevalence, distribution, and behavior of these latent NSCs remained elusive. To tackle these issues, we reconstructed the spatiotemporal pattern of striatal (STR) AS neurogenic activation after excitotoxic lesion in mice. Our results indicate that neurogenic potential is widespread among STR ASs but is focally activated at the lesion border, where it associates with different reactive AS subtypes. In this region, similarly to canonical niches, steady-state neurogenesis is ensured by the continuous stochastic activation of local ASs. Activated ASs quickly return to quiescence, while their progeny transiently expand following a stochastic behavior that features an acceleration in differentiation propensity. Notably, STR AS activation rate matches that of SVZ ASs indicating a comparable prevalence of NSC potential.
Collapse
Affiliation(s)
- Marco Fogli
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Giulia Nato
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Philip Greulich
- School of Mathematical Sciences, University of Southampton, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK
| | - Jacopo Pinto
- Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Marta Ribodino
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Gregorio Valsania
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Paolo Peretto
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Annalisa Buffo
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Federico Luzzati
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Turin), Italy; Department of Life Sciences and System Biology, University of Turin, Turin, Italy.
| |
Collapse
|
13
|
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
|
14
|
Friess D, Brauer S, Pöysti A, Choudhury C, Harris L. Tools to study neural and glioma stem cell quiescence. Trends Neurosci 2024; 47:736-748. [PMID: 39191628 DOI: 10.1016/j.tins.2024.07.004] [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: 05/13/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Quiescence is a prolonged but reversible state of cell-cycle arrest that is an adaptive feature of most adult stem cell populations. In the brain, quiescence helps to protect adult neural stem cells from stress and supports lifelong neurogenesis. Unfortunately however, entry into a quiescent or a slow-cycling state is also a malignant feature of brain cancer stem cells. In glioblastoma, where the process has been best characterised, quiescent glioma stem cells preferentially survive chemoradiation, and after therapy, reactivate to regrow the tumour and drive recurrence. In this Review, we discuss the in vitro and in vivo models that have been developed for studying neural stem cell quiescence and how these tools may be used to deepen biological understanding and to develop novel therapies targeting quiescent glioma stem cells.
Collapse
Affiliation(s)
- Dana Friess
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; The University of Queensland, Brisbane, School of Biomedical Sciences, QLD, 4067, Australia
| | - Stephanie Brauer
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; Queensland University of Technology, School of Biomedical Sciences, QLD, 4059, Australia
| | - Anni Pöysti
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, WC1E 6DD London, UK
| | - Chandra Choudhury
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; The University of Queensland, Brisbane, School of Biomedical Sciences, QLD, 4067, Australia
| | - Lachlan Harris
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia; The University of Queensland, Brisbane, School of Biomedical Sciences, QLD, 4067, Australia; Queensland University of Technology, School of Biomedical Sciences, QLD, 4059, Australia.
| |
Collapse
|
15
|
Zhao T, Hong Y, Yan B, Huang S, Ming GL, Song H. Epigenetic maintenance of adult neural stem cell quiescence in the mouse hippocampus via Setd1a. Nat Commun 2024; 15:5674. [PMID: 38971831 PMCID: PMC11227589 DOI: 10.1038/s41467-024-50010-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024] Open
Abstract
Quiescence, a hallmark of adult neural stem cells (NSCs), is required for maintaining the NSC pool to support life-long continuous neurogenesis in the adult dentate gyrus (DG). Whether long-lasting epigenetic modifications maintain NSC quiescence over the long term in the adult DG is not well-understood. Here we show that mice with haploinsufficiency of Setd1a, a schizophrenia risk gene encoding a histone H3K4 methyltransferase, develop an enlarged DG with more dentate granule cells after young adulthood. Deletion of Setd1a specifically in quiescent NSCs in the adult DG promotes their activation and neurogenesis, which is countered by inhibition of the histone demethylase LSD1. Mechanistically, RNA-sequencing and CUT & RUN analyses of cultured quiescent adult NSCs reveal Setd1a deletion-induced transcriptional changes and many Setd1a targets, among which down-regulation of Bhlhe40 promotes quiescent NSC activation in the adult DG in vivo. Together, our study reveals a Setd1a-dependent epigenetic mechanism that sustains NSC quiescence in the adult DG.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA
| | - Yan Hong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA
| | - Bowen Yan
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philedaphia, PA, 19104, USA.
- The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
16
|
Frye BM, Negrey JD, Johnson CSC, Kim J, Barcus RA, Lockhart SN, Whitlow CT, Chiou KL, Snyder-Mackler N, Montine TJ, Craft S, Shively CA, Register TC. Mediterranean diet protects against a neuroinflammatory cortical transcriptome: Associations with brain volumetrics, peripheral inflammation, social isolation, and anxiety in nonhuman primates (Macaca fascicularis). Brain Behav Immun 2024; 119:681-692. [PMID: 38636565 DOI: 10.1016/j.bbi.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/17/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Mediterranean diets may be neuroprotective and prevent cognitive decline relative to Western diets; however, the underlying biology is poorly understood. We assessed the effects of Western versus Mediterranean-like diets on RNAseq-generated transcriptional profiles in lateral temporal cortex and their relationships with longitudinal changes in neuroanatomy, circulating monocyte gene expression, and observations of social isolation and anxiety in 38 socially-housed, middle-aged female cynomolgus macaques (Macaca fascicularis). Diet resulted in differential expression of seven transcripts (FDR < 0.05). Cyclin dependent kinase 14 (CDK14), a proinflammatory regulator, was lower in the Mediterranean group. The remaining six transcripts [i.e., "lunatic fringe" (LFNG), mannose receptor C type 2 (MRC2), solute carrier family 3 member 2 (SLCA32), butyrophilin subfamily 2 member A1 (BTN2A1), katanin regulatory subunit B1 (KATNB1), and transmembrane protein 268 (TMEM268)] were higher in cortex of the Mediterranean group and generally associated with anti-inflammatory/neuroprotective pathways. KATNB1 encodes a subcomponent of katanin, important in maintaining microtubule homeostasis. BTN2A1 is involved in immunomodulation of γδ T-cells which have anti-neuroinflammatory and neuroprotective effects. CDK14, LFNG, MRC2, and SLCA32 are associated with inflammatory pathways. The latter four differentially expressed cortex transcripts were associated with peripheral monocyte transcript levels, neuroanatomical changes determined by MRI, and with social isolation and anxiety. These results provide important insights into the potential mechanistic processes linking diet, peripheral and central inflammation, and behavior. Collectively, our results provide evidence that, relative to Western diets, Mediterranean diets confer protection against peripheral and central inflammation which is reflected in preserved brain structure and socioemotional behavior. Ultimately, such protective effects may confer resilience to the development of neuropathology and associated disease.
Collapse
Affiliation(s)
- Brett M Frye
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Biology, Emory and Henry College, Emory, VA, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA
| | - Jacob D Negrey
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; School of Anthropology, University of Arizona, Tucson, AZ, USA
| | | | - Jeongchul Kim
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Richard A Barcus
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Samuel N Lockhart
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher T Whitlow
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kenneth L Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA; School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
| | | | - Suzanne Craft
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carol A Shively
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA.
| | - Thomas C Register
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA.
| |
Collapse
|
17
|
Lodewijk GA, de Geus M, Guimarães RLFP, Jacobs FMJ. Emergence of the ZNF675 Gene During Primate Evolution-Influenced Human Neurodevelopment Through Changing HES1 Autoregulation. J Comp Neurol 2024; 532:e25648. [PMID: 38958676 DOI: 10.1002/cne.25648] [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: 12/31/2023] [Revised: 04/27/2024] [Accepted: 05/30/2024] [Indexed: 07/04/2024]
Abstract
In this study, we investigated recurrent copy number variations (CNVs) in the 19p12 locus, which are associated with neurodevelopmental disorders. The two genes in this locus, ZNF675 and ZNF681, arose via gene duplication in primates, and their presence in several pathological CNVs in the human population suggests that either or both of these genes are required for normal human brain development. ZNF675 and ZNF681 are members of the Krüppel-associated box zinc finger (KZNF) protein family, a class of transcriptional repressors important for epigenetic silencing of specific genomic regions. About 170 primate-specific KZNFs are present in the human genome. Although KZNFs are primarily associated with repressing retrotransposon-derived DNA, evidence is emerging that they can be co-opted for other gene regulatory processes. We show that genetic deletion of ZNF675 causes developmental defects in cortical organoids, and our data suggest that part of the observed neurodevelopmental phenotype is mediated by a gene regulatory role of ZNF675 on the promoter of the neurodevelopmental gene Hes family BHLH transcription factor 1 (HES1). We also find evidence for the recently evolved regulation of genes involved in neurological disorders, microcephalin 1 and sestrin 3. We show that ZNF675 interferes with HES1 auto-inhibition, a process essential for the maintenance of neural progenitors. As a striking example of how some KZNFs have integrated into preexisting gene expression networks, these findings suggest the emergence of ZNF675 has caused a change in the balance of HES1 autoregulation. The association of ZNF675 CNV with human developmental disorders and ZNF675-mediated regulation of neurodevelopmental genes suggests that it evolved into an important factor for human brain development.
Collapse
Affiliation(s)
- Gerrald A Lodewijk
- Swammerdam Institute for Life Sciences, Evolutionary Neurogenomics, University of Amsterdam, Amsterdam, The Netherlands
| | - Matthijs de Geus
- Swammerdam Institute for Life Sciences, Evolutionary Neurogenomics, University of Amsterdam, Amsterdam, The Netherlands
| | - Rita L F P Guimarães
- Swammerdam Institute for Life Sciences, Evolutionary Neurogenomics, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank M J Jacobs
- Swammerdam Institute for Life Sciences, Evolutionary Neurogenomics, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Sachan N, Sharma V, Mutsuddi M, Mukherjee A. Notch signalling: multifaceted role in development and disease. FEBS J 2024; 291:3030-3059. [PMID: 37166442 DOI: 10.1111/febs.16815] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/08/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Notch pathway is an evolutionarily conserved signalling system that operates to influence an astonishing array of cell fate decisions in different developmental contexts. Notch signalling plays important roles in many developmental processes, making it difficult to name a tissue or a developing organ that does not depend on Notch function at one stage or another. Thus, dysregulation of Notch signalling is associated with many developmental defects and various pathological conditions, including cancer. Although many recent advances have been made to reveal different aspects of the Notch signalling mechanism and its intricate regulation, there are still many unanswered questions related to how the Notch signalling pathway functions in so many developmental events. The same pathway can be deployed in numerous cellular contexts to play varied and critical roles in an organism's development and this is only possible because of the complex regulatory mechanisms of the pathway. In this review, we provide an overview of the mechanism and regulation of the Notch signalling pathway along with its multifaceted functions in different aspects of development and disease.
Collapse
Affiliation(s)
- Nalani Sachan
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
19
|
Bou-Rouphael J, Doulazmi M, Eschstruth A, Abdou A, Durand BC. Cerebellar granular neuron progenitors exit their germinative niche via BarH-like1 activity mediated partly by inhibition of T-cell factor. Development 2024; 151:dev202234. [PMID: 38860486 DOI: 10.1242/dev.202234] [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: 12/26/2023] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Cerebellar granule neuron progenitors (GNPs) originate from the upper rhombic lip (URL), a germinative niche in which developmental defects produce human diseases. T-cell factor (TCF) responsiveness and Notch dependence are hallmarks of self-renewal in neural stem cells. TCF activity, together with transcripts encoding proneural gene repressors hairy and enhancer of split (Hes/Hey), are detected in the URL; however, their functions and regulatory modes are undeciphered. Here, we established amphibian as a pertinent model for studying vertebrate URL development. The amphibian long-lived URL is TCF active, whereas the external granular layer (EGL) is non-proliferative and expresses hes4 and hes5 genes. Using functional and transcriptomic approaches, we show that TCF activity is necessary for URL emergence and maintenance. We establish that the transcription factor Barhl1 controls GNP exit from the URL, acting partly through direct TCF inhibition. Identification of Barhl1 target genes suggests that, besides TCF, Barhl1 inhibits transcription of hes5 genes independently of Notch signaling. Observations in amniotes suggest a conserved role for Barhl in maintenance of the URL and/or EGL via co-regulation of TCF, Hes and Hey genes.
Collapse
Affiliation(s)
- Johnny Bou-Rouphael
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Mohamed Doulazmi
- Sorbonne Université, CNRS UMR8256, Institut de Biologie Paris-Seine (IBPS) - Laboratoire Adaptation Biologique et Vieillissement, 75005 Paris, France
| | - Alexis Eschstruth
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Asna Abdou
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Béatrice C Durand
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
- Sorbonne Université, CNRS UMR8256, Institut de Biologie Paris-Seine (IBPS) - Laboratoire Adaptation Biologique et Vieillissement, 75005 Paris, France
| |
Collapse
|
20
|
Khan AA, Langston HC, Walsh L, Roscoe R, Jayawardhana S, Francisco AF, Taylor MC, McCann CJ, Kelly JM, Lewis MD. Enteric nervous system regeneration and functional cure of experimental digestive Chagas disease with trypanocidal chemotherapy. Nat Commun 2024; 15:4400. [PMID: 38782898 PMCID: PMC11116530 DOI: 10.1038/s41467-024-48749-5] [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: 01/05/2023] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Digestive Chagas disease (DCD) is an enteric neuropathy caused by Trypanosoma cruzi infection. There is a lack of evidence on the mechanism of pathogenesis and rationales for treatment. We used a female C3H/HeN mouse model that recapitulates key clinical manifestations to study how infection dynamics shape DCD pathology and the impact of treatment with the front-line, anti-parasitic drug benznidazole. Curative treatment 6 weeks post-infection resulted in sustained recovery of gastrointestinal transit function, whereas treatment failure led to infection relapse and gradual return of DCD symptoms. Neuro/immune gene expression patterns shifted from chronic inflammation to a tissue repair profile after cure, accompanied by increased cellular proliferation, glial cell marker expression and recovery of neuronal density in the myenteric plexus. Delaying treatment until 24 weeks post-infection led to partial reversal of DCD, suggesting the accumulation of permanent tissue damage over the course of chronic infection. Our study shows that murine DCD pathogenesis is sustained by chronic T. cruzi infection and is not an inevitable consequence of acute stage denervation. The risk of irreversible enteric neuromuscular tissue damage and dysfunction developing highlights the importance of prompt diagnosis and treatment. These findings support the concept of treating asymptomatic, T. cruzi-infected individuals with benznidazole to prevent DCD development.
Collapse
Affiliation(s)
- Archie A Khan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Harry C Langston
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Louis Walsh
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Rebecca Roscoe
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Martin C Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, University College London, Great Ormond Street Institute of Child Health, London, UK
| | - John M Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | - Michael D Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK.
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CV4 7AJ, Coventry, UK.
| |
Collapse
|
21
|
Yao J, Dai S, Zhu R, Tan J, Zhao Q, Yin Y, Sun J, Du X, Ge L, Xu J, Hou C, Li N, Li J, Ji W, Zhu C, Zhang R, Li T. Deciphering molecular heterogeneity and dynamics of human hippocampal neural stem cells at different ages and injury states. eLife 2024; 12:RP89507. [PMID: 38607670 PMCID: PMC11014727 DOI: 10.7554/elife.89507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
While accumulated publications support the existence of neurogenesis in the adult human hippocampus, the homeostasis and developmental potentials of neural stem cells (NSCs) under different contexts remain unclear. Based on our generated single-nucleus atlas of the human hippocampus across neonatal, adult, aging, and injury, we dissected the molecular heterogeneity and transcriptional dynamics of human hippocampal NSCs under different contexts. We further identified new specific neurogenic lineage markers that overcome the lack of specificity found in some well-known markers. Based on developmental trajectory and molecular signatures, we found that a subset of NSCs exhibit quiescent properties after birth, and most NSCs become deep quiescence during aging. Furthermore, certain deep quiescent NSCs are reactivated following stroke injury. Together, our findings provide valuable insights into the development, aging, and reactivation of the human hippocampal NSCs, and help to explain why adult hippocampal neurogenesis is infrequently observed in humans.
Collapse
Affiliation(s)
- Junjun Yao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Shaoxing Dai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Ran Zhu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Ju Tan
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Qiancheng Zhao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Jiansen Sun
- Zhong-Zhi- Yi-Gu Research InstituteChongqingChina
| | - Xuewei Du
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Longjiao Ge
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Jianhua Xu
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Chunli Hou
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Nan Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Jun Li
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Chuhong Zhu
- Department of Anatomy, National and Regional Engineering Laboratory of Tissue Engineering, State Key Laboratory of Trauma, Burn and Combined Injury, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical UniversityChongqingChina
| | - Runrui Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| |
Collapse
|
22
|
Labusch M, Thetiot M, Than-Trong E, Morizet D, Coolen M, Varet H, Legendre R, Ortica S, Mancini L, Bally-Cuif L. Prosaposin maintains adult neural stem cells in a state associated with deep quiescence. Stem Cell Reports 2024; 19:515-528. [PMID: 38518783 PMCID: PMC11096431 DOI: 10.1016/j.stemcr.2024.02.007] [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: 08/12/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/24/2024] Open
Abstract
In most vertebrates, adult neural stem cells (NSCs) continuously give rise to neurons in discrete brain regions. A critical process for maintaining NSC pools over long periods of time in the adult brain is NSC quiescence, a reversible and tightly regulated state of cell-cycle arrest. Recently, lysosomes were identified to regulate the NSC quiescence-proliferation balance. However, it remains controversial whether lysosomal activity promotes NSC proliferation or quiescence, and a finer influence of lysosomal activity on NSC quiescence duration or depth remains unexplored. Using RNA sequencing and pharmacological manipulations, we show that lysosomes are necessary for NSC quiescence maintenance. In addition, we reveal that expression of psap, encoding the lysosomal regulator Prosaposin, is enriched in quiescent NSCs (qNSCs) that reside upstream in the NSC lineage and display a deep/long quiescence phase in the adult zebrafish telencephalon. We show that shRNA-mediated psap knockdown increases the proportion of activated NSCs (aNSCs) as well as NSCs that reside in shallower quiescence states (signed by ascl1a and deltaA expression). Collectively, our results identify the lysosomal protein Psap as a (direct or indirect) quiescence regulator and unfold the interplay between lysosomal function and NSC quiescence heterogeneities.
Collapse
Affiliation(s)
- Miriam Labusch
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Melina Thetiot
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - Emmanuel Than-Trong
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - David Morizet
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Marion Coolen
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - Hugo Varet
- Institut Pasteur, Université Paris Cité, Platform Biomics, 75015 Paris, France
| | - Rachel Legendre
- Institut Pasteur, Université Paris Cité, Platform Biomics, 75015 Paris, France
| | - Sara Ortica
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France
| | - Laure Mancini
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Laure Bally-Cuif
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, 75015 Paris, France.
| |
Collapse
|
23
|
Ciceri G, Baggiolini A, Cho HS, Kshirsagar M, Benito-Kwiecinski S, Walsh RM, Aromolaran KA, Gonzalez-Hernandez AJ, Munguba H, Koo SY, Xu N, Sevilla KJ, Goldstein PA, Levitz J, Leslie CS, Koche RP, Studer L. An epigenetic barrier sets the timing of human neuronal maturation. Nature 2024; 626:881-890. [PMID: 38297124 PMCID: PMC10881400 DOI: 10.1038/s41586-023-06984-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 12/15/2023] [Indexed: 02/02/2024]
Abstract
The pace of human brain development is highly protracted compared with most other species1-7. The maturation of cortical neurons is particularly slow, taking months to years to develop adult functions3-5. Remarkably, such protracted timing is retained in cortical neurons derived from human pluripotent stem cells (hPSCs) during in vitro differentiation or upon transplantation into the mouse brain4,8,9. Those findings suggest the presence of a cell-intrinsic clock setting the pace of neuronal maturation, although the molecular nature of this clock remains unknown. Here we identify an epigenetic developmental programme that sets the timing of human neuronal maturation. First, we developed a hPSC-based approach to synchronize the birth of cortical neurons in vitro which enabled us to define an atlas of morphological, functional and molecular maturation. We observed a slow unfolding of maturation programmes, limited by the retention of specific epigenetic factors. Loss of function of several of those factors in cortical neurons enables precocious maturation. Transient inhibition of EZH2, EHMT1 and EHMT2 or DOT1L, at progenitor stage primes newly born neurons to rapidly acquire mature properties upon differentiation. Thus our findings reveal that the rate at which human neurons mature is set well before neurogenesis through the establishment of an epigenetic barrier in progenitor cells. Mechanistically, this barrier holds transcriptional maturation programmes in a poised state that is gradually released to ensure the prolonged timeline of human cortical neuron maturation.
Collapse
Affiliation(s)
- Gabriele Ciceri
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Arianna Baggiolini
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Institute of Oncology Research (IOR), Bellinzona Institutes of Science (BIOS+), Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Hyein S Cho
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meghana Kshirsagar
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Microsoft AI for Good Research, Redmond, WA, USA
| | - Silvia Benito-Kwiecinski
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan M Walsh
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - So Yeon Koo
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Neuroscience PhD Program, New York, NY, USA
| | - Nan Xu
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kaylin J Sevilla
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter A Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Christina S Leslie
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
24
|
Wang G, Zhang Z, Li J, Han J, Lu C. The PTB and PRR domains of numb regulate neurite outgrowth by influencing voltage-gated calcium channel expression and kinetics. Brain Res Bull 2024; 207:110876. [PMID: 38215950 DOI: 10.1016/j.brainresbull.2024.110876] [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: 08/15/2023] [Revised: 12/17/2023] [Accepted: 01/08/2024] [Indexed: 01/14/2024]
Abstract
Numb is an evolutionarily conserved protein that regulates the differentiation of neuronal progenitor cells through unknown mechanisms. Numb has four alternative splice variants with different lengths of phosphotyrosine-binding (PTB) and proline-rich regions (PRR) domains. In this study, we demonstrated that Numb expression was increased in the primary cultures of rat cortical and hippocampal neurons over time in vitro, and Numb antisense inhibited neurite outgrowth. We verified that cells overexpressing short PTB (SPTB) or long PTB (LPTB) domains exhibited differentiation or proliferation, respectively. SPTB-mediated differentiation was related to the PRR domains, as cells expressing SPTB/LPRR had longer dendrites and more branched dendrites than cells expressing SPTB/SPRR. The differentiation of both cell types was completely blocked by the Ca2+ chelator. Western blot analysis revealed the increased total protein expression of voltage-gated calcium channel (VGCC) subunit α1C and α1D in cells expressing SPTB and LPTB Numb. The increased expression of the VGCC β3 subunit was only observed in cells expressing SPTB Numb. Immunocytochemistry further showed that SPTB-mediated cell differentiation was associated with increased membrane expression of VGCC subunits α1C, α1D and β3, which corresponded to the higher Ca2+ current (ICa) densities. Furthermore, we found that VGCC of cells transfected with SPTB/SPRR or SPTB/LPRR Numb isoforms exhibit steady-state inactivation (SSI) in both differentiated and undifferentiated phenotypes. A similar SSI of VGCC was observed in the differentiated cells transfected with SPTB/SPRR or SPTB/LPRR Numb isoforms, whereas a left shift SSI of VGCC in cells expressing SPTB/LPRR was detected in the undifferentiated cells. Collectively, these data indicate that SPTB domain is essential for neurite outgrowth involving in membrane expression of VGCC subunits, and LPRR plays a role in neuronal branching and the regulation of VGCC inactivation kinetics.
Collapse
Affiliation(s)
- Guodong Wang
- International-Joint Lab for Non-Invasive Neural Modulation of Henan Province, Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang 453003, China; School of Nursing, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhengyan Zhang
- International-Joint Lab for Non-Invasive Neural Modulation of Henan Province, Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang 453003, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Junmei Li
- International-Joint Lab for Non-Invasive Neural Modulation of Henan Province, Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang 453003, China
| | - Jinhong Han
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Chengbiao Lu
- International-Joint Lab for Non-Invasive Neural Modulation of Henan Province, Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
25
|
Zhang H, Rui M, Ma Z, Gong S, Zhang S, Zhou Q, Gan C, Gong W, Wang S. Golgi-to-ER retrograde transport prevents premature differentiation of Drosophila type II neuroblasts via Notch-signal-sending daughter cells. iScience 2024; 27:108545. [PMID: 38213621 PMCID: PMC10783626 DOI: 10.1016/j.isci.2023.108545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/18/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024] Open
Abstract
Stem cells are heterogeneous to generate diverse differentiated cell types required for organogenesis; however, the underlying mechanisms that differently maintain these heterogeneous stem cells are not well understood. In this study, we identify that Golgi-to-endoplasmic reticulum (ER) retrograde transport specifically maintains type II neuroblasts (NBs) through the Notch signaling. We reveal that intermediate neural progenitors (INPs), immediate daughter cells of type II NBs, provide Delta and function as the NB niche. The Delta used by INPs is mainly produced by NBs and asymmetrically distributed to INPs. Blocking retrograde transport leads to a decrease in INP number, which reduces Notch activity and results in the premature differentiation of type II NBs. Furthermore, the reduction of Delta could suppress tumor formation caused by type II NBs. Our results highlight the crosstalk between Golgi-to-ER retrograde transport, Notch signaling, stem cell niche, and fusion as an essential step in maintaining the self-renewal of type II NB lineage.
Collapse
Affiliation(s)
- Huanhuan Zhang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Menglong Rui
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Zhixin Ma
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Sifan Gong
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Shuliu Zhang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Qingxia Zhou
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Congfeng Gan
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Wenting Gong
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Su Wang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210096, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
| |
Collapse
|
26
|
Li Z, Li H, Yu X, Zhou J, Dong ZY, Meng X. bHLH transcription factors Hes1, Ascl1 and Oligo2 exhibit different expression patterns in the process of physiological electric fields-induced neuronal differentiation. Mol Biol Rep 2024; 51:115. [PMID: 38227267 DOI: 10.1007/s11033-023-09118-5] [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: 01/01/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Recent studies have shown that the expression of bHLH transcription factors Hes1, Ascl1, and Oligo2 has an oscillating balance in neural stem cells (NSCs) to maintain their self-proliferation and multi-directional differentiation potential. This balance can be disrupted by exogenous stimulation. Our previous work has identified that electrical stimulation could induce neuronal differentiation of mouse NSCs. METHODS To further evaluate if physiological electric fields (EFs)-induced neuronal differentiation is related to the expression patterns of bHLH transcription factors Hes1, Ascl1, and Oligo2, mouse embryonic brain NSCs were used to investigate the expression changes of Ascl1, Hes1 and Oligo2 in mRNA and protein levels during EF-induced neuronal differentiation. RESULTS Our results showed that NSCs expressed high level of Hes1, while expression of Ascl1 and Oligo2 stayed at very low levels. When NSCs exited proliferation, the expression of Hes1 in differentiated cells began to decrease and oscillated at the low expression level. Oligo2 showed irregular changes in low expression level. EF-stimulation significantly increased the expression of Ascl1 at mRNA and protein levels accompanied by an increased percentage of neuronal differentiation. What's more, over-expression of Hes1 inhibited the neuronal differentiation induced by EFs. CONCLUSION EF-stimulation directed neuronal differentiation of NSCs by promoting the continuous accumulation of Ascl1 expression and decreasing the expression of Hes1.
Collapse
Affiliation(s)
- Zhe Li
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Hai Li
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, People's Republic of China
- Department of Radiology, The First Hospital, Jilin University, Changchun, 130041, People's Republic of China
| | - Xiyao Yu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Jiaying Zhou
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Zhi Yong Dong
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Xiaoting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
27
|
Chen C, Song J, Pu Q, Liu X, Yan J, Wang X, Wang H, Qian Q. Azithromycin induces neurotoxicity in zebrafish by interfering with the VEGF/Notch signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 903:166505. [PMID: 37625730 DOI: 10.1016/j.scitotenv.2023.166505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Azithromycin (AZM) is a widely used antibiotic in both human and veterinary medicine, and its use has significantly increased during the COVID-19 pandemic. However, potential adverse effects of AZM on aquatic organisms have not been well studied. In this study, we explored the neurotoxicity of AZM in zebrafish and delved into its underlying mechanisms. Our results showed that AZM exposure resulted in a spectrum of detrimental effects in zebrafish, encompassing abnormal behaviors, damaged neuronal development, aberrant lateral line nervous system development, vascular malformations and perturbed expression of genes related to neural development. Moreover, we observed a concentration-dependent exacerbation of these neurotoxic manifestations with increasing AZM concentrations. Notably, AZM induced excessive cell apoptosis and oxidative stress damage. In addition, alterations in the expression levels of the genes involved in the VEGF/Notch signaling pathway were evident in AZM-exposed zebrafish. Consequently, we hypothesize that AZM may induce neurotoxicity by influencing the VEGF/Notch signaling pathway. To validate this hypothesis, we introduced a VEGF signaling inhibitor, axitinib, and a Notch signaling agonist, valproic acid, alongside AZM exposure. Remarkably, the administration of these rescue compounds significantly mitigated the neurotoxic effects induced by AZM. This dual verification provides compelling evidence that AZM indeed induces neurotoxicity during the early developmental stages of zebrafish, primarily through its interference with the VEGF/Notch pathway. Innovatively, our study reveals the molecular mechanism of AZM-induced neurotoxicity from the perspective of the close connection between blood vessels and nervous system. These findings provide new insights into the potential mechanisms underlying the neurotoxic effect of antibiotics and highlight the need for further investigation into the ecotoxicological effects of antibiotics on aquatic organisms and the potential risks to human health.
Collapse
Affiliation(s)
- Chen Chen
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jie Song
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Qian Pu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Xingcheng Liu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jin Yan
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Xuedong Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Huili Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Qiuhui Qian
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
28
|
Negrey JD, Frye BM, Johnson CSC, Kim J, Barcus RA, Lockhart SN, Whitlow CT, Sutphen C, Chiou KL, Snyder-Mackler N, Montine TJ, Craft S, Shively CA, Register TC. Mediterranean Diet Protects Against a Neuroinflammatory Cortical Transcriptome: Associations with Brain Volumetrics, Peripheral Inflammation, Social Isolation and Anxiety. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.565068. [PMID: 37961556 PMCID: PMC10635044 DOI: 10.1101/2023.11.01.565068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
INTRODUCTION Mediterranean diets may be neuroprotective and prevent cognitive decline relative to Western diets, however the underlying biology is poorly understood. METHODS We assessed the effects of Western vs. Mediterranean-like diets on RNAseq generated transcriptional profiles in temporal cortex and their relationships with changes in MRI neuroimaging phenotypes, circulating monocyte gene expression, and observations of social isolation and anxiety in 38 socially-housed, middle-aged female cynomolgus macaques. RESULTS Diet resulted in differential expression of seven transcripts (FDR<0.05). Cyclin dependent kinase 14 ( CDK14 ), a proinflammatory regulator, was lower in the Mediterranean group. The remaining six transcripts [i.e., "lunatic fringe" ( LFNG ), mannose receptor C type 2 ( MRC2 ), solute carrier family 3 member 2 ( SLCA32 ), butyrophilin subfamily 2 member A1 ( BTN2A1 ), katanin regulatory subunit B1 ( KATNB1 ), and transmembrane protein 268 ( TMEM268 )] were higher in cortex of the Mediterranean group and generally associated with anti-inflammatory/neuroprotective pathways. KATNB1 encodes a subcomponent of katanin, important in maintaining microtubule homeostasis. BTN2A1 is involved in immunomodulation of γδ T-cells which have anti-neuroinflammatory and neuroprotective effects. CDK14 , LFNG , MRC2, and SLCA32 are associated with inflammatory pathways. The latter four differentially expressed cortex transcripts were associated with monocyte transcript levels, changes in AD-relevant brain volumes determined by MRI over the course of the study, and social isolation and anxiety. CDK14 was positively correlated with monocyte inflammatory transcripts, changes in total brain, gray matter, cortical gray matter volumes, and time alone and anxious behavior, and negatively correlated with changes in total white matter and cerebrospinal fluid (CSF) volumes. In contrast, LFNG , MRC2 , and SLCA32 were negatively correlated with monocyte inflammatory transcripts and changes in total gray matter volume, and positively correlated with CSF volume changes, and SLCA32 was negatively correlated with time alone. DISCUSSION Collectively, our results suggest that relative to Western diets, Mediterranean diets confer protection against peripheral and central inflammation which is reflected in preserved brain structure and behavior.
Collapse
|
29
|
Murnan KM, Horbinski C, Stegh AH. Redox Homeostasis and Beyond: The Role of Wild-Type Isocitrate Dehydrogenases for the Pathogenesis of Glioblastoma. Antioxid Redox Signal 2023; 39:923-941. [PMID: 37132598 PMCID: PMC10654994 DOI: 10.1089/ars.2023.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 05/04/2023]
Abstract
Significance: Glioblastoma is an aggressive and devastating brain tumor characterized by a dismal prognosis and resistance to therapeutic intervention. To support catabolic processes critical for unabated cellular growth and defend against harmful reactive oxygen species, glioblastoma tumors upregulate the expression of wild-type isocitrate dehydrogenases (IDHs). IDH enzymes catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), NAD(P)H, and CO2. On molecular levels, IDHs epigenetically control gene expression through effects on α-KG-dependent dioxygenases, maintain redox balance, and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis. Recent Advances: While gain-of-function mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effects, recent studies identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down regulated, as contributing to glioblastoma progression. Critical Issues: Here, we will discuss molecular mechanisms of how wild-type IDHs control glioma pathogenesis, including the regulation of oxidative stress and de novo lipid biosynthesis, and provide an overview of current and future research directives that aim to fully characterize wild-type IDH-driven metabolic reprogramming and its contribution to the pathogenesis of glioblastoma. Future Directions: Future studies are required to further dissect mechanisms of metabolic and epigenomic reprogramming in tumors and the tumor microenvironment, and to develop pharmacological approaches to inhibit wild-type IDH function. Antioxid. Redox Signal. 39, 923-941.
Collapse
Affiliation(s)
- Kevin M. Murnan
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Malnati Brain Tumor Institute, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Alexander H. Stegh
- Department of Neurological Surgery, The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
30
|
Simorgh S, Mousavi SA, To SK, Pasque V, Wierda K, Vervliet T, Yeganeh M, Pooyan P, Chai YC, Verfaillie C, Baharvand H. A facile method to generate cerebral organoids from human pluripotent stem cells. EXCLI JOURNAL 2023; 22:1055-1076. [PMID: 37927348 PMCID: PMC10620858 DOI: 10.17179/excli2023-6299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
Human cerebral organoids (COs) are self-organizing three-dimensional (3D) neural structures that provide a human-specific platform to study the cellular and molecular processes that underlie different neurological events. The first step of CO generation from human pluripotent stem cells (hPSCs) is neural induction, which is an in vitro simulation of neural ectoderm development. Several signaling pathways cooperate during neural ectoderm development and in vitro differentiation of hPSCs toward neural cell lineages is also affected by them. In this study, we considered some of the known sources of these variable signaling cues arising from cell culture media components and sought to modulate their effects by applying a comprehensive combination of small molecules and growth factors for CO generation. Histological analysis demonstrated that these COs recapitulate the neural progenitor zone and early cortical layer organization, containing different types of neuronal and glial cells which was in accordance with single-nucleus transcriptome profiling results. Moreover, patch clamp and intracellular Ca2+ dynamic studies demonstrated that the COs behave as a functional neural network. Thus, this method serves as a facile protocol for generating hPSC-derived COs that faithfully mimic the features of their in vivo counterparts in the developing human brain. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Susan Simorgh
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Seyed Ahmad Mousavi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - San Kit To
- Department of Development and Regeneration, Lab for Epigenetic Reprogramming, Leuven Stem Cell Institute, Leuven Single-Cell Omics Institute and Leuven Cancer Institute, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Vincent Pasque
- Department of Development and Regeneration, Lab for Epigenetic Reprogramming, Leuven Stem Cell Institute, Leuven Single-Cell Omics Institute and Leuven Cancer Institute, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium
- Electrophysiology Unit, Leuven 3000, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Meghdad Yeganeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Paria Pooyan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Yoke Chin Chai
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Catherine Verfaillie
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Hossein Baharvand
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
31
|
Martin M, Gutierrez-Avino F, Shaikh MN, Tejedor FJ. A novel proneural function of Asense is integrated with the sequential actions of Delta-Notch, L'sc and Su(H) to promote the neuroepithelial to neuroblast transition. PLoS Genet 2023; 19:e1010991. [PMID: 37871020 PMCID: PMC10621995 DOI: 10.1371/journal.pgen.1010991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 11/02/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023] Open
Abstract
In order for neural progenitors (NPs) to generate distinct populations of neurons at the right time and place during CNS development, they must switch from undergoing purely proliferative, self-renewing divisions to neurogenic, asymmetric divisions in a tightly regulated manner. In the developing Drosophila optic lobe, neuroepithelial (NE) cells of the outer proliferation center (OPC) are progressively transformed into neurogenic NPs called neuroblasts (NBs) in a medial to lateral proneural wave. The cells undergoing this transition express Lethal of Scute (L'sc), a proneural transcription factor (TF) of the Acheate Scute Complex (AS-C). Here we show that there is also a peak of expression of Asense (Ase), another AS-C TF, in the cells neighboring those with transient L'sc expression. These peak of Ase cells help to identify a new transitional stage as they have lost NE markers and L'sc, they receive a strong Notch signal and barely exhibit NB markers. This expression of Ase is necessary and sufficient to promote the NE to NB transition in a more robust and rapid manner than that of l'sc gain of function or Notch loss of function. Thus, to our knowledge, these data provide the first direct evidence of a proneural role for Ase in CNS neurogenesis. Strikingly, we found that strong Delta-Notch signaling at the lateral border of the NE triggers l'sc expression, which in turn induces ase expression in the adjacent cells through the activation of Delta-Notch signaling. These results reveal two novel non-conventional actions of Notch signaling in driving the expression of proneural factors, in contrast to the repression that Notch signaling exerts on them during classical lateral inhibition. Finally, Suppressor of Hairless (Su(H)), which seems to be upregulated late in the transitioning cells and in NBs, represses l'sc and ase, ensuring their expression is transient. Thus, our data identify a key proneural role of Ase that is integrated with the sequential activities of Delta-Notch signaling, L'sc, and Su(H), driving the progressive transformation of NE cells into NBs.
Collapse
Affiliation(s)
- Mercedes Martin
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Francisco Gutierrez-Avino
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Mirja N. Shaikh
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Francisco J. Tejedor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| |
Collapse
|
32
|
Jiménez Peinado P, Urbach A. From Youthful Vigor to Aging Decline: Unravelling the Intrinsic and Extrinsic Determinants of Hippocampal Neural Stem Cell Aging. Cells 2023; 12:2086. [PMID: 37626896 PMCID: PMC10453598 DOI: 10.3390/cells12162086] [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: 06/22/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Since Joseph Altman published his pioneering work demonstrating neurogenesis in the hippocampus of adult rats, the number of publications in this field increased exponentially. Today, we know that the adult hippocampus harbors a pool of adult neural stem cells (NSCs) that are the source of life-long neurogenesis and plasticity. The functions of these NSCs are regulated by extrinsic cues arising from neighboring cells and the systemic environment. However, this tight regulation is subject to imbalance with age, resulting in a decline in adult NSCs and neurogenesis, which contributes to the progressive deterioration of hippocampus-related cognitive functions. Despite extensive investigation, the mechanisms underlying this age-related decline in neurogenesis are only incompletely understood, but appear to include an increase in NSC quiescence, changes in differentiation patterns, and NSC exhaustion. In this review, we summarize recent work that has improved our knowledge of hippocampal NSC aging, focusing on NSC-intrinsic mechanisms as well as cellular and molecular changes in the niche and systemic environment that might be involved in the age-related decline in NSC functions. Additionally, we identify future directions that may advance our understanding of NSC aging and the concomitant loss of hippocampal neurogenesis and plasticity.
Collapse
Affiliation(s)
| | - Anja Urbach
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
- Jena Center for Healthy Aging, Jena University Hospital, 07747 Jena, Germany
- Aging Research Center Jena, Leibniz Institute on Aging, 07745 Jena, Germany
| |
Collapse
|
33
|
Soto DC, Uribe-Salazar JM, Shew CJ, Sekar A, McGinty S, Dennis MY. Genomic structural variation: A complex but important driver of human evolution. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2023; 181 Suppl 76:118-144. [PMID: 36794631 PMCID: PMC10329998 DOI: 10.1002/ajpa.24713] [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: 10/02/2022] [Revised: 01/21/2023] [Accepted: 02/05/2023] [Indexed: 02/17/2023]
Abstract
Structural variants (SVs)-including duplications, deletions, and inversions of DNA-can have significant genomic and functional impacts but are technically difficult to identify and assay compared with single-nucleotide variants. With the aid of new genomic technologies, it has become clear that SVs account for significant differences across and within species. This phenomenon is particularly well-documented for humans and other primates due to the wealth of sequence data available. In great apes, SVs affect a larger number of nucleotides than single-nucleotide variants, with many identified SVs exhibiting population and species specificity. In this review, we highlight the importance of SVs in human evolution by (1) how they have shaped great ape genomes resulting in sensitized regions associated with traits and diseases, (2) their impact on gene functions and regulation, which subsequently has played a role in natural selection, and (3) the role of gene duplications in human brain evolution. We further discuss how to incorporate SVs in research, including the strengths and limitations of various genomic approaches. Finally, we propose future considerations in integrating existing data and biospecimens with the ever-expanding SV compendium propelled by biotechnology advancements.
Collapse
Affiliation(s)
- Daniela C. Soto
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, CA, USA
| | - José M. Uribe-Salazar
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, CA, USA
| | - Colin J. Shew
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, CA, USA
| | - Aarthi Sekar
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, CA, USA
| | - Sean McGinty
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, CA, USA
| | - Megan Y. Dennis
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, CA, USA
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, CA, USA
| |
Collapse
|
34
|
Silva-García CG. Devo-Aging: Intersections Between Development and Aging. GeroScience 2023; 45:2145-2159. [PMID: 37160658 PMCID: PMC10651630 DOI: 10.1007/s11357-023-00809-2] [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: 12/30/2022] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
There are two fundamental questions in developmental biology. How does a single fertilized cell give rise to a whole body? and how does this body later produce progeny? Synchronization of these embryonic and postembryonic developments ensures continuity of life from one generation to the next. An enormous amount of work has been done to unravel the molecular mechanisms behind these processes, but more recently, modern developmental biology has been expanded to study development in wider contexts, including regeneration, environment, disease, and even aging. However, we have just started to understand how the mechanisms that govern development also regulate aging. This review discusses examples of signaling pathways involved in development to elucidate how their regulation influences healthspan and lifespan. Therefore, a better knowledge of developmental signaling pathways stresses the possibility of using them as innovative biomarkers and targets for aging and age-related diseases.
Collapse
Affiliation(s)
- Carlos Giovanni Silva-García
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA.
- Center on the Biology of Aging, Brown University, Providence, RI, USA.
| |
Collapse
|
35
|
Lampada A, Taylor V. Notch signaling as a master regulator of adult neurogenesis. Front Neurosci 2023; 17:1179011. [PMID: 37457009 PMCID: PMC10339389 DOI: 10.3389/fnins.2023.1179011] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Neurogenesis ceases in most regions of the mammalian brain before or shortly after birth, however, in a few restricted brain regions, the production of new neurons proceeds into adulthood. Neural stem cells (NSCs) in these neurogenic zones are integrated into niches that control their activity and fate. Most stem cells in the adult brain are mitotically inactive and these cells can remain quiescent for months or even years. One of the key questions is what are the molecular mechanisms that regulate NSC maintenance and differentiation. Notch signaling has been shown to be a critical regulator of stem cell activity and maintenance in many tissues including in the nervous system. In this mini-review we discuss the roles of Notch signaling and the functions of the different Notch receptors and ligands in regulating neurogenesis in the adult murine brain. We review the functions of Notch signaling components in controlling NSC quiescence and entry into cell cycle and neurogenesis.
Collapse
|
36
|
Fang L, Kuniya T, Harada Y, Yasuda O, Maeda N, Suzuki Y, Kawaguchi D, Gotoh Y. TIMP3 promotes the maintenance of neural stem-progenitor cells in the mouse subventricular zone. Front Neurosci 2023; 17:1149603. [PMID: 37456993 PMCID: PMC10338847 DOI: 10.3389/fnins.2023.1149603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Adult neural stem cells (NSCs) in the mouse subventricular zone (SVZ) serve as a lifelong reservoir for newborn olfactory bulb neurons. Recent studies have identified a slowly dividing subpopulation of embryonic neural stem-progenitor cells (NPCs) as the embryonic origin of adult NSCs. Yet, little is known about how these slowly dividing embryonic NPCs are maintained until adulthood while other NPCs are extinguished by the completion of brain development. The extracellular matrix (ECM) is an essential component of stem cell niches and thus a key determinant of stem cell fate. Here we investigated tissue inhibitors of metalloproteinases (TIMPs)-regulators of ECM remodeling-for their potential roles in the establishment of adult NSCs. We found that Timp2, Timp3, and Timp4 were expressed at high levels in slowly dividing NPCs compared to rapidly dividing NPCs. Deletion of TIMP3 reduced the number of adult NSCs and neuroblasts in the lateral SVZ. In addition, overexpression of TIMP3 in the embryonic NPCs suppressed neuronal differentiation and upregulated the expression levels of Notch signaling relating genes. These results thus suggest that TIMP3 keeps the undifferentiated state of embryonic NPCs, leading to the establishment and maintenance of adult NSCs.
Collapse
Affiliation(s)
- Lingyan Fang
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takaaki Kuniya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yujin Harada
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Osamu Yasuda
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, Kanoya, Japan
| | - Nobuyo Maeda
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, Kanoya, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Daichi Kawaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
37
|
Mitsiadis TA, Pagella P, Capellini TD, Smith MM. The Notch-mediated circuitry in the evolution and generation of new cell lineages: the tooth model. Cell Mol Life Sci 2023; 80:182. [PMID: 37330998 DOI: 10.1007/s00018-023-04831-7] [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: 03/14/2023] [Revised: 05/19/2023] [Accepted: 06/09/2023] [Indexed: 06/20/2023]
Abstract
The Notch pathway is an ancient, evolutionary conserved intercellular signaling mechanism that is involved in cell fate specification and proper embryonic development. The Jagged2 gene, which encodes a ligand for the Notch family of receptors, is expressed from the earliest stages of odontogenesis in epithelial cells that will later generate the enamel-producing ameloblasts. Homozygous Jagged2 mutant mice exhibit abnormal tooth morphology and impaired enamel deposition. Enamel composition and structure in mammals are tightly linked to the enamel organ that represents an evolutionary unit formed by distinct dental epithelial cell types. The physical cooperativity between Notch ligands and receptors suggests that Jagged2 deletion could alter the expression profile of Notch receptors, thus modifying the whole Notch signaling cascade in cells within the enamel organ. Indeed, both Notch1 and Notch2 expression are severely disturbed in the enamel organ of Jagged2 mutant teeth. It appears that the deregulation of the Notch signaling cascade reverts the evolutionary path generating dental structures more reminiscent of the enameloid of fishes rather than of mammalian enamel. Loss of interactions between Notch and Jagged proteins may initiate the suppression of complementary dental epithelial cell fates acquired during evolution. We propose that the increased number of Notch homologues in metazoa enabled incipient sister cell types to form and maintain distinctive cell fates within organs and tissues along evolution.
Collapse
Affiliation(s)
- Thimios A Mitsiadis
- Institute of Oral Biology, Centre for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland.
| | - Pierfrancesco Pagella
- Institute of Oral Biology, Centre for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland
- Wallenberg Center for Molecular Medicine (WCMM) and Department of Biomedical and Clinical Sciences, Linköpings Universitet, 581 85, Linköping, Sweden
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Moya Meredith Smith
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, King's College London, London, UK
| |
Collapse
|
38
|
Han JS, Fishman-Williams E, Decker SC, Hino K, Reyes RV, Brown NL, Simó S, Torre AL. Notch directs telencephalic development and controls neocortical neuron fate determination by regulating microRNA levels. Development 2023; 150:dev201408. [PMID: 37272771 PMCID: PMC10309580 DOI: 10.1242/dev.201408] [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: 10/27/2022] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
The central nervous system contains a myriad of different cell types produced from multipotent neural progenitors. Neural progenitors acquire distinct cell identities depending on their spatial position, but they are also influenced by temporal cues to give rise to different cell populations over time. For instance, the progenitors of the cerebral neocortex generate different populations of excitatory projection neurons following a well-known sequence. The Notch signaling pathway plays crucial roles during this process, but the molecular mechanisms by which Notch impacts progenitor fate decisions have not been fully resolved. Here, we show that Notch signaling is essential for neocortical and hippocampal morphogenesis, and for the development of the corpus callosum and choroid plexus. Our data also indicate that, in the neocortex, Notch controls projection neuron fate determination through the regulation of two microRNA clusters that include let-7, miR-99a/100 and miR-125b. Our findings collectively suggest that balanced Notch signaling is crucial for telencephalic development and that the interplay between Notch and miRNAs is essential for the control of neocortical progenitor behaviors and neuron cell fate decisions.
Collapse
Affiliation(s)
- Jisoo S. Han
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | | | - Steven C. Decker
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Keiko Hino
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Raenier V. Reyes
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Nadean L. Brown
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
39
|
Maeda Y, Isomura A, Masaki T, Kageyama R. Differential cell-cycle control by oscillatory versus sustained Hes1 expression via p21. Cell Rep 2023; 42:112520. [PMID: 37200191 DOI: 10.1016/j.celrep.2023.112520] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/06/2023] [Accepted: 05/02/2023] [Indexed: 05/20/2023] Open
Abstract
Oscillatory Hes1 expression activates cell proliferation, while high and sustained Hes1 expression induces quiescence, but the mechanism by which Hes1 differentially controls cell proliferation depending on its expression dynamics is unclear. Here, we show that oscillatory Hes1 expression down-regulates the expression of the cyclin-dependent kinase inhibitor p21 (Cdkn1a), which delays cell-cycle progression, and thereby activates the proliferation of mouse neural stem cells (NSCs). By contrast, sustained Hes1 overexpression up-regulates p21 expression and inhibits NSC proliferation, although it initially down-regulates p21 expression. Compared with Hes1 oscillation, sustained Hes1 overexpression represses Dusp7, a phosphatase for phosphorylated Erk (p-Erk), and increases the levels of p-Erk, which can up-regulate p21 expression. These results indicate that p21 expression is directly repressed by oscillatory Hes1 expression, but indirectly up-regulated by sustained Hes1 overexpression, suggesting that depending on its expression dynamics, Hes1 differentially controls NSC proliferation via p21.
Collapse
Affiliation(s)
- Yuki Maeda
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Akihiro Isomura
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan; Japan Science and Technology Agency, PRESTO, Saitama 332-0012, Japan
| | - Taimu Masaki
- RIKEN Center for Brain Science, Wako 351-0198, Japan
| | - Ryoichiro Kageyama
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
40
|
Damle EB, Morrison VE, Cioma J, Volic M, Bix GJ. Co-administration of extracellular matrix-based biomaterials with neural stem cell transplantation for treatment of central nervous system injury. Front Neurosci 2023; 17:1177040. [PMID: 37255752 PMCID: PMC10225608 DOI: 10.3389/fnins.2023.1177040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/28/2023] [Indexed: 06/01/2023] Open
Abstract
Injuries and disorders of the central nervous system (CNS) present a particularly difficult challenge for modern medicine to address, given the complex nature of the tissues, obstacles in researching and implementing therapies, and barriers to translating efficacious treatments into human patients. Recent advancements in neural stem cell (NSC) transplantation, endogenous neurogenesis, and in vivo reprogramming of non-neural cells into the neuronal lineage represent multiple approaches to resolving CNS injury. However, we propose that one practice that must be incorporated universally in neuroregeneration studies is the use of extracellular matrix (ECM)-mimicking biomaterials to supply the architectural support and cellular microenvironment necessary for partial or complete restoration of function. Through consideration of developmental processes including neurogenesis, cellular migration, and establishment of functional connectivity, as well as evaluation of process-specific interactions between cells and ECM components, insights can be gained to harness and modulate native and induced neurobiological processes to promote CNS tissue repair. Further, evaluation of the current landscape of regenerative medicine and tissue engineering techniques external to the neurosciences provides key perspectives into the role of the ECM in the use of stem cell-based therapies, and the potential directions future neuroregenerative approaches may take. If the most successful of these approaches achieve wide-spread adoption, innovative paired NSC-ECM strategies for neuroregeneration may become prominent in the near future, and with the rapid advances these techniques are poised to herald, a new era of treatment for CNS injury may dawn.
Collapse
Affiliation(s)
- Eshan B. Damle
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Vivianne E. Morrison
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jozef Cioma
- Faculty of Biology, Medicine, and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Milla Volic
- Faculty of Biology, Medicine, and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Gregory J. Bix
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
41
|
Abstract
Traditional views of cellular metabolism imply that it is passively adapted to meet the demands of the cell. It is becoming increasingly clear, however, that metabolites do more than simply supply the substrates for biological processes; they also provide critical signals, either through effects on metabolic pathways or via modulation of other regulatory proteins. Recent investigation has also uncovered novel roles for several metabolites that expand their signalling influence to processes outside metabolism, including nutrient sensing and storage, embryonic development, cell survival and differentiation, and immune activation and cytokine secretion. Together, these studies suggest that, in contrast to the prevailing notion, the biochemistry of a cell is frequently governed by its underlying metabolism rather than vice versa. This important shift in perspective places common metabolites as key regulators of cell phenotype and behaviour. Yet the signalling metabolites, and the cognate targets and transducers through which they signal, are only beginning to be uncovered. In this Review, we discuss the emerging links between metabolism and cellular behaviour. We hope this will inspire further dissection of the mechanisms through which metabolic pathways and intermediates modulate cell function and will suggest possible drug targets for diseases linked to metabolic deregulation.
Collapse
Affiliation(s)
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, USA.
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
42
|
Loras A, Gonzalez-Bonet LG, Gutierrez-Arroyo JL, Martinez-Cadenas C, Marques-Torrejon MA. Neural Stem Cells as Potential Glioblastoma Cells of Origin. Life (Basel) 2023; 13:life13040905. [PMID: 37109434 PMCID: PMC10145968 DOI: 10.3390/life13040905] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor in adults and it remains incurable. These tumors are very heterogeneous, resistant to cytotoxic therapies, and they show high rates of invasiveness. Therefore, patients face poor prognosis, and the survival rates remain very low. Previous research states that GBM contains a cell population with stem cell characteristics called glioma stem cells (GSCs). These cells are able to self-renew and regenerate the tumor and, therefore, they are partly responsible for the observed resistance to therapies and tumor recurrence. Recent data indicate that neural stem cells (NSCs) in the subventricular zone (SVZ) are the cells of origin of GBM, that is, the cell type acquiring the initial tumorigenic mutation. The involvement of SVZ-NSCs is also associated with GBM progression and recurrence. Identifying the cellular origin of GBM is important for the development of early detection techniques and the discovery of early disease markers. In this review, we analyze the SVZ-NSC population as a potential GBM cell of origin, and its potential role for GBM therapies.
Collapse
Affiliation(s)
- Alba Loras
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon de la Plana, Spain
| | - Julia L. Gutierrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon de la Plana, Spain
| | | | - Maria Angeles Marques-Torrejon
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
- Correspondence: ; Tel.: +34-964-387-478
| |
Collapse
|
43
|
Pavlou MAS, Singh K, Ravichandran S, Halder R, Nicot N, Birck C, Grandbarbe L, Del Sol A, Michelucci A. Transcriptional and Chromatin Accessibility Profiling of Neural Stem Cells Differentiating into Astrocytes Reveal Dynamic Signatures Affected under Inflammatory Conditions. Cells 2023; 12:cells12060948. [PMID: 36980289 PMCID: PMC10047363 DOI: 10.3390/cells12060948] [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: 01/18/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Astrocytes arise from multipotent neural stem cells (NSCs) and represent the most abundant cell type of the central nervous system (CNS), playing key roles in the developing and adult brain. Since the differentiation of NSCs towards a gliogenic fate is a precisely timed and regulated process, its perturbation gives rise to dysfunctional astrocytic phenotypes. Inflammation, which often underlies neurological disorders, including neurodevelopmental disorders and brain tumors, disrupts the accurate developmental process of NSCs. However, the specific consequences of an inflammatory environment on the epigenetic and transcriptional programs underlying NSCs' differentiation into astrocytes is unexplored. Here, we address this gap by profiling in mice glial precursors from neural tissue derived from early embryonic stages along their astrocytic differentiation trajectory in the presence or absence of tumor necrosis factor (TNF), a master pro-inflammatory cytokine. By using a combination of RNA- and ATAC-sequencing approaches, together with footprint and integrated gene regulatory network analyses, we here identify key differences during the differentiation of NSCs into astrocytes under physiological and inflammatory settings. In agreement with its role to turn cells resistant to inflammatory challenges, we detect Nrf2 as a master transcription factor supporting the astrocytic differentiation under TNF exposure. Further, under these conditions, we unravel additional transcriptional regulatory hubs, including Stat3, Smad3, Cebpb, and Nfkb2, highlighting the interplay among pathways underlying physiological astrocytic developmental processes and those involved in inflammatory responses, resulting in discrete astrocytic phenotypes. Overall, our study reports key transcriptional and epigenetic changes leading to the identification of molecular regulators of astrocytic differentiation. Furthermore, our analyses provide a valuable resource for understanding inflammation-induced astrocytic phenotypes that might contribute to the development and progression of CNS disorders with an inflammatory component.
Collapse
Affiliation(s)
- Maria Angeliki S Pavlou
- Department of Life Sciences and Medicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1210 Luxembourg, Luxembourg
| | - Kartikeya Singh
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Srikanth Ravichandran
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Rashi Halder
- Scientific Central Services, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Nathalie Nicot
- Translational Medicine Operations Hub, Luxembourg Institute of Health, L-3555 Dudelange, Luxembourg
- LuxGen Genome Center, Luxembourg Institute of Health & Laboratoire National de Santé, L-3555 Dudelange, Luxembourg
| | - Cindy Birck
- Department of Life Sciences and Medicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Luc Grandbarbe
- Department of Life Sciences and Medicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
- Computational Biology Group, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), 48160 Derio, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, L-1210 Luxembourg, Luxembourg
- Scientific Central Services, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
44
|
Coronas V, Arnault P, Jégou JF, Cousin L, Rabeony H, Clarhaut S, Harnois T, Lecron JC, Morel F. IL-22 Promotes Neural Stem Cell Self-Renewal in the Adult Brain. Stem Cells 2023; 41:252-259. [PMID: 36635952 DOI: 10.1093/stmcls/sxad003] [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: 07/05/2022] [Accepted: 12/19/2022] [Indexed: 01/14/2023]
Abstract
Mainly known for its role in immune defense and inflammation, interleukin 22 (IL-22) has emerged over the past decade as a cytokine involved in the adaptation of stem/progenitor cell activity for tissue homeostasis and repair. IL-22 is present in the brain, which harbors neural stem cells (NSC) in specific niches of which the ventricular-subventricular zone (V-SVZ) is the most important. In this study, we examined a possible effect of IL-22 on NSC in the adult mouse brain. We demonstrate that the IL-22 receptor is expressed in the V-SVZ, mainly in NSC characterized by their SOX2 expression. Addition of IL-22 to V-VSZ cell cultures resulted in an increase in NSC self-renewal, associated with a shift in NSC division mode towards symmetric proliferative divisions at the expense of differentiative divisions. Conversely, loss of IL-22 in knockout mice led to a decrease in neurosphere yield, suggesting a reduction in the NSC population, which was confirmed by the decrease in cells retaining BrdU labeling in IL-22 knockout mice. Our study supports that IL-22 is involved in the development and/or maintenance of V-VSZ NSC and opens new avenues to further investigate the role of IL-22 in NSC biology in health and disease.
Collapse
Affiliation(s)
- Valérie Coronas
- 4CS, Laboratory Channels & Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Patricia Arnault
- 4CS, Laboratory Channels & Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Jean-François Jégou
- LITEC, Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, University of Poitiers, Poitiers, France
| | - Laetitia Cousin
- 4CS, Laboratory Channels & Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Hanitriniaina Rabeony
- LITEC, Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, University of Poitiers, Poitiers, France
| | - Sandrine Clarhaut
- LITEC, Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, University of Poitiers, Poitiers, France
| | - Thomas Harnois
- 4CS, Laboratory Channels & Connexins in Cancers and Cell Stemness, CNRS UMR 6041, University of Poitiers, Poitiers, France
| | - Jean-Claude Lecron
- LITEC, Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, University of Poitiers, Poitiers, France
- Service Immunologie et Inflammation, UBM, CHU de Poitiers, Poitiers, France
| | - Franck Morel
- LITEC, Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, University of Poitiers, Poitiers, France
| |
Collapse
|
45
|
Regulation of Cell Plasticity by Bromodomain and Extraterminal Domain (BET) Proteins: A New Perspective in Glioblastoma Therapy. Int J Mol Sci 2023; 24:ijms24065665. [PMID: 36982740 PMCID: PMC10055343 DOI: 10.3390/ijms24065665] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
BET proteins are a family of multifunctional epigenetic readers, mainly involved in transcriptional regulation through chromatin modelling. Transcriptome handling ability of BET proteins suggests a key role in the modulation of cell plasticity, both in fate decision and in lineage commitment during embryonic development and in pathogenic conditions, including cancerogenesis. Glioblastoma is the most aggressive form of glioma, characterized by a very poor prognosis despite the application of a multimodal therapy. Recently, new insights are emerging about the glioblastoma cellular origin, leading to the hypothesis that several putative mechanisms occur during gliomagenesis. Interestingly, epigenome dysregulation associated with loss of cellular identity and functions are emerging as crucial features of glioblastoma pathogenesis. Therefore, the emerging roles of BET protein in glioblastoma onco-biology and the compelling demand for more effective therapeutic strategies suggest that BET family members could be promising targets for translational breakthroughs in glioblastoma treatment. Primarily, “Reprogramming Therapy”, which is aimed at reverting the malignant phenotype, is now considered a promising strategy for GBM therapy.
Collapse
|
46
|
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
|
47
|
Liu Y, Bilen M, McNicoll MM, Harris RA, Fong BC, Iqbal MA, Paul S, Mayne J, Walker K, Wang J, Figeys D, Slack RS. Early postnatal defects in neurogenesis in the 3xTg mouse model of Alzheimer's disease. Cell Death Dis 2023; 14:138. [PMID: 36801910 PMCID: PMC9938901 DOI: 10.1038/s41419-023-05650-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 01/26/2023] [Accepted: 02/03/2023] [Indexed: 02/19/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder leading to dementia. The hippocampus, which is one of the sites where neural stem cells reside and new neurons are born, exhibits the most significant neuronal loss in AD. A decline in adult neurogenesis has been described in several animal models of AD. However, the age at which this defect first appears remains unknown. To determine at which stage, from birth to adulthood, the neurogenic deficits are found in AD, we used the triple transgenic mouse model of AD (3xTg). We show that defects in neurogenesis are present as early as postnatal stages, well before the onset of any neuropathology or behavioral deficits. We also show that 3xTg mice have significantly fewer neural stem/progenitor cells, with reduced proliferation and decreased numbers of newborn neurons at postnatal stages, consistent with reduced volumes of hippocampal structures. To determine whether there are early changes in the molecular signatures of neural stem/progenitor cells, we perform bulk RNA-seq on cells sorted directly from the hippocampus. We show significant changes in the gene expression profiles at one month of age, including genes of the Notch and Wnt pathways. These findings reveal impairments in neurogenesis very early in the 3xTg AD model, which provides new opportunities for early diagnosis and therapeutic interventions to prevent neurodegeneration in AD.
Collapse
Affiliation(s)
- Yubing Liu
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| | - Maria Bilen
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| | - Marie-Michelle McNicoll
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| | - Richard A. Harris
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| | - Bensun C. Fong
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| | - Mohamed Ariff Iqbal
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| | - Smitha Paul
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| | - Janice Mayne
- grid.28046.380000 0001 2182 2255Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, K1H 8M5 Ottawa, Canada
| | - Krystal Walker
- grid.28046.380000 0001 2182 2255Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, K1H 8M5 Ottawa, Canada
| | - Jing Wang
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada ,grid.412687.e0000 0000 9606 5108Regenerative Medicine Program, Ottawa Hospital Research Institute, K1H 8L6 Ottawa, Canada
| | - Daniel Figeys
- grid.28046.380000 0001 2182 2255Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, K1H 8M5 Ottawa, Canada
| | - Ruth S. Slack
- grid.28046.380000 0001 2182 2255Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research institute, K1H 8M5 Ottawa, Canada
| |
Collapse
|
48
|
He L, Bhat K, Ioannidis A, Pajonk F. Effects of Dopamine Receptor Antagonists and Radiation on Mouse Neural Stem/Progenitor Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524632. [PMID: 36712018 PMCID: PMC9882258 DOI: 10.1101/2023.01.18.524632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background Dopamine receptor antagonists are psychotropic drugs that have been originally developed against psychiatric disorders. We recently identified dopamine receptor antagonists as potential anti-cancer agents and some have entered clinical trials against glioblastoma. Radiotherapy is known to cause cognitive impairment in patients receiving cranial irradiation through the elimination of neural stem/progenitor cells and subsequent loss of neurogenesis. Methods Using transgenic mice that report the presence of neural stem/progenitor cells through Nestin promoter-driven expression of enhanced green fluorescent protein, the effects of dopamine receptor antagonists alone or in combination with radiation on murine neural stem/progenitor cells were assessed in sphere-formation assays, flow cytometry and immunofluorescence in vitro and in vivo . Results We report that several dopamine receptor antagonists show sex-dependent effects on neural stem/progenitor cells both in vitro and in vivo . Hydroxyzine, trifluoperazine, amisulpride, nemonapride or quetiapine alone or in combination with radiation significantly increased the number of neural stem/progenitor cells in female neurospheres but not in male mice. Dopamine receptor antagonists either protected neural stem/progenitor cells from radiation or expanded the stem cell pool, thus indicating that this combination therapy against glioblastoma will not increase radiation-induced cognitive decline through increasing elimination of neural stem/progenitor cells and subsequent loss of neurogenesis. Conclusions We conclude that a therapeutic window for dopamine receptor antagonists in combination with radiation potentially exist, making it a novel combination therapy against glioblastoma. Normal tissue toxicity of this combination potentially differs depending on age and sex and should be taken into consideration when designing clinical trials. Key Points - Neural stem/progenitor cells show sex-dependent sensitivity to dopamine receptor antagonists- Dopamine receptor antagonists active against GBM increase Neural stem/progenitor cells counts. Importance of the Study Combination therapy of dopamine receptor antagonists with radiation have entered clinical trials against glioblastoma but the normal tissue toxicity of this combination has not been fully explored yet. Here we present evidence that some dopamine receptor antagonists show sex-dependent effects on neural stem/progenitor cells either by protecting neural stem/progenitor cells from radiation or inducing an expansion of the stem cell pool, suggesting that this combination therapy against glioblastoma will not increase radiation-induced cognitive decline through increasing elimination of neural stem/progenitor cells and subsequent loss of neurogenesis. Normal tissue toxicity of this combination potentially differs depending on age and sex and should be further explored in clinical trials.
Collapse
|
49
|
Identification of the Time Period during Which BMP Signaling Regulates Proliferation of Neural Progenitor Cells in Zebrafish. Int J Mol Sci 2023; 24:ijms24021733. [PMID: 36675251 PMCID: PMC9863262 DOI: 10.3390/ijms24021733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates neural induction, neuronal specification, and neuronal differentiation. However, the role of BMP signaling in neural progenitors remains unclear. This is because interruption of BMP signaling before or during neural induction causes severe effects on subsequent neural developmental processes. To examine the role of BMP signaling in the development of neural progenitors in zebrafish, we bypassed the effect of BMP signaling on neural induction and suppressed BMP signaling at different time points during gastrulation using a temporally controlled transgenic line carrying a dominant-negative form of Bmp receptor type 1aa and a chemical inhibitor of BMP signaling, DMH1. Inhibiting BMP signaling from 8 hpf could bypass BMP regulation on neural induction, induce the number of proliferating neural progenitors, and reduce the number of neuronal precursors. Inhibiting BMP signaling upregulates the expression of the Notch downstream gene hairy/E(spl)-related 2 (her2). Inhibiting Notch signaling or knocking down the Her2 function reduced neural progenitor proliferation, whereas inactivating BMP signaling in Notch-Her2 deficient background restored the number of proliferating neural progenitors. These results reveal the time window for the proliferation of neural progenitors during zebrafish development and a fine balance between BMP and Notch signaling in regulating the proliferation of neural progenitor cells.
Collapse
|
50
|
Zhang X, Lou Y, Zheng D, Lu J, Qi D. Quantitative proteomic analysis reveals the effects of mu opioid agonists on HT22 cells. Front Pharmacol 2023; 13:1022449. [PMID: 36699066 PMCID: PMC9868271 DOI: 10.3389/fphar.2022.1022449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction: At present, the mu opioid receptor is the most important neuroaesthetics receptor in anesthesiology research, and the damage that it does to the nervous system is unknown. Methods: We investigated the effects of loperamide, an agonist of the mu opioid receptor, on protein expression in HT22 cells using stable isotope labeling of amino acids in cell culture (SILAC), immobilized metal affinity chromatography (IMAC) enrichment, and high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of 7,823 proteins were identified. Results and Discussion: Bioinformatic analysis revealed that mu opioid receptor agonism can induce distinct changes in the proteome of HT22 cells. These findings improve our understanding of narcotic drugs.
Collapse
Affiliation(s)
- Xutong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yani Lou
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dongxu Zheng
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jialin Lu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dansi Qi
- Department of Pathology, Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China,*Correspondence: Dansi Qi,
| |
Collapse
|