1
|
So KF. Commentary on: "Human neural stem cell-derived artificial organelles to improve oxidative phosphorylation". Neural Regen Res 2025; 20:3040. [PMID: 39610109 PMCID: PMC11826448 DOI: 10.4103/nrr.nrr-d-24-01079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 11/30/2024] Open
Affiliation(s)
- Kwok-Fai So
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China; State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
| |
Collapse
|
2
|
Gaston-Breton R, Disdier C, Hagberg H, Mabondzo A. Hypoxia-ischemia and sexual dimorphism: modeling mitochondrial dysfunction using brain organoids. Cell Biosci 2025; 15:67. [PMID: 40413513 PMCID: PMC12103005 DOI: 10.1186/s13578-025-01402-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/27/2025] [Indexed: 05/27/2025] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a leading cause of neurodevelopmental morbidities in full-term infants. There is strong evidence of sexual differences in hypoxic-ischemic (HI) injury where male neonates are at higher risk as they are subject to more pronounced neurological deficits and death than females. The cellular and molecular mechanisms underlying these sexual discrepancies in HI injury are poorly understood. Mitochondrial dysregulation has been increasingly explored in brain diseases and represents a major target during HI events. In this review, we discuss (1) different mitochondrial functions in the central nervous system (2), mitochondrial dysregulation in the context of HI injury (3), sex-dependent mitochondrial pathways in HIE and (4) modeling of mitochondrial dysfunction using human brain organoids. Gaining insight into these novel aspects of mitochondrial function will offer valuable understanding of brain development and neurological disorders such as HI injury, paving the way for the discovery and creation of new treatment approaches.
Collapse
Affiliation(s)
- Romane Gaston-Breton
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France
| | - Clémence Disdier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France
| | | | - Aloïse Mabondzo
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France.
| |
Collapse
|
3
|
Guan Y, Li L, Yang R, Lu Y, Tang J. Targeting mitochondria with natural polyphenols for treating Neurodegenerative Diseases: a comprehensive scoping review from oxidative stress perspective. J Transl Med 2025; 23:572. [PMID: 40410831 PMCID: PMC12100838 DOI: 10.1186/s12967-025-06605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
Neurodegenerative diseases are a class of conditions with widespread detrimental impacts, currently lacking effective therapeutic drugs. Recent studies have identified mitochondrial dysfunction and the resultant oxidative stress as crucial contributors to the pathogenesis of neurodegenerative diseases. Polyphenols, naturally occurring compounds with inherent antioxidant properties, have demonstrated the potential to target mitochondria and mitigate oxidative stress. This therapeutic potential has garnered significant attention in recent years. Investigating the mitochondrial targeting capacity of polyphenols, their role in functional regulation, and their ability to modulate oxidative stress, along with exploring novel technologies and strategies for modifying polyphenol compounds and their formulations, holds promise for providing new avenues for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yueyue Guan
- Department of Encephalopathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Lei Li
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Rui Yang
- Department of Encephalopathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Yun Lu
- Department of Emergency Medicine, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Jun Tang
- Department of Encephalopathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China.
| |
Collapse
|
4
|
Lane AR, Roberts BR, Fahrni CJ, Faundez V. A primer on copper biology in the brain. Neurobiol Dis 2025; 212:106974. [PMID: 40414313 DOI: 10.1016/j.nbd.2025.106974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 05/14/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025] Open
Abstract
This primer aims to expose scientists who study the brain to the field of copper biology. We briefly discuss key copper homeostasis mechanisms and proteins and place these functions in the context of the brain and neurodevelopment. A small number of key copper genes are explored as representative examples of the importance of this metal to the brain. We show that these genes are expressed throughout the brain and their defects are linked to a diverse array of neurological phenotypes, which we discuss further in the context of several neurological and neurodegenerative diseases associated with dysregulation of copper. This review aims to expose interested scientists to the fundamental roles for copper in the brain, the primary proteins responsible for maintaining copper homeostasis in the brain, and the classic neurological diseases associated with this metal.
Collapse
Affiliation(s)
- Alicia R Lane
- Department of Cell Biology, Emory University, 615 Michael St, Atlanta, GA 30322, USA.
| | - Blaine R Roberts
- Department of Biochemistry, Emory University, 1510 Clifton Rd, Atlanta, GA 30322, USA; Department of Neurology, Emory University, 12 Executive Park Dr NE, Atlanta, GA 30322, USA.
| | - Christoph J Fahrni
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Victor Faundez
- Department of Cell Biology, Emory University, 615 Michael St, Atlanta, GA 30322, USA.
| |
Collapse
|
5
|
Pipicelli F, Villalba A, Hippenmeyer S. How radial glia progenitor lineages generate cell-type diversity in the developing cerebral cortex. Curr Opin Neurobiol 2025; 93:103046. [PMID: 40383049 DOI: 10.1016/j.conb.2025.103046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/05/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
The cerebral cortex is arguably the most complex organ in humans. The cortical architecture is characterized by a remarkable diversity of neuronal and glial cell types that make up its neuronal circuits. Following a precise temporally ordered program, radial glia progenitor (RGP) cells generate all cortical excitatory projection neurons and glial cell-types. Cortical excitatory projection neurons are produced either directly or via intermediate progenitors, through indirect neurogenesis. How the extensive cortical cell-type diversity is generated during cortex development remains, however, a fundamental open question. How do RGPs quantitatively and qualitatively generate all the neocortical neurons? How does direct and indirect neurogenesis contribute to the establishment of neuronal and lineage heterogeneity? Whether RGPs represent a homogeneous and/or multipotent progenitor population, or if RGPs consist of heterogeneous groups is currently also not known. In this review, we will summarize the latest findings that contributed to a deeper insight into the above key questions.
Collapse
Affiliation(s)
- Fabrizia Pipicelli
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Ana Villalba
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
6
|
Robertson GL, Bodnya C, Gama V. Mitochondrial and peroxisomal fission in cortical neurogenesis. Int J Biochem Cell Biol 2025; 182-183:106774. [PMID: 40158688 DOI: 10.1016/j.biocel.2025.106774] [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: 01/03/2024] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human brain is unique in its cellular diversity, intricate cytoarchitecture, function, and complex metabolic and bioenergetic demands, for which mitochondria and peroxisomes are essential. Mitochondria are multifunctional organelles that coordinate various signaling pathways central to neurogenesis. The dynamic morphological changes of the mitochondrial network have been linked to the regulation of bioenergetic and metabolic states. Specific protein machinery is dedicated to mitochondrial fission and fusion, allowing organelle distribution during cell division, organelle repair, and adaptation to environmental stimuli (excellent reviews have been published on these topics [Kondadi and Reichert, 2024; Giacomello et al., 2020; Tilokani et al., 2018; Kraus et al., 2021; Navaratnarajah et al., 2021]). In parallel, peroxisomes contain over 50 different enzymes which regulate metabolic functions that are critical for neurogenesis (Berger et al., 2016; Hulshagen et al., 2008). Peroxisomes share many of the components of their fission machinery with the mitochondria and undergo fission to help meet metabolic demands in response to environmental stimuli (Schrader et al., 2016). This review focuses primarily on the machinery involved in mitochondrial and peroxisomal fission. Mitochondrial fission has been identified as a critical determinant of cell fate decisions (Iwata et al., 2023, 2020; Khacho et al., 2016; King et al., 2021; Prigione and Adjaye, 2010; Vantaggiato et al., 2019; Kraus et al., 2021). The connection between alterations in peroxisomal fission and metabolic changes associated with cellular differentiation remains less clear. Here, we provide an overview of the functional and regulatory aspects of the mitochondrial and peroxisomal fission machinery and provide insight into the current mechanistic understanding by which mitochondrial and peroxisomal fission influence neurogenesis.
Collapse
Affiliation(s)
| | - Caroline Bodnya
- Vanderbilt University, Cell and Developmental Biology, Nashville, TN, United States
| | - Vivian Gama
- Vanderbilt University, Cell and Developmental Biology, Nashville, TN, United States; Vanderbilt University, Vanderbilt Center for Stem Cell Biology, Nashville, TN, United States; Vanderbilt University, Vanderbilt Brain Institute, Nashville, TN, United States.
| |
Collapse
|
7
|
Yang M, Qin X, Liu X. The effect of mitochondrial-associated endoplasmic reticulum membranes (MAMs) modulation: New insights into therapeutic targets for depression. Neurosci Biobehav Rev 2025; 172:106087. [PMID: 40031998 DOI: 10.1016/j.neubiorev.2025.106087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/07/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
Depression is a prevalent mental disorder with high morbidity and mortality and its pathogenesis remains exactly unclarified. However, mitochondria and endoplasmic reticulum (ER) are two highly dynamic organelles that perform an indispensable role in the development of depression. Mitochondrial dysfunction and ER stress are recognized as vital pathological hallmarks in depression. The changes of intracellular activities such as mitochondrial dynamics, mitophagy, energy metabolism and ER stress are closely correlated with the progression of depression. Moreover, organelles interactions are conducive to homeostasis and cellular functions, and mitochondrial-associated endoplasmic reticulum membranes (MAMs) serve as signaling hubs of the two organelles and the coupling of the pathological progression. The main roles of MAMs are involved in metabolism, signal transduction, lipid transport, and maintenance of its structure and function. At present, accumulating studies elucidated that MAMs have gradually become a novel therapeutic target in treatment of depression. In the review, we focus on influence of mitochondria dysfunction and ER stress on depression. Furthermore, we discuss the underlying role of MAMs in depression and highlight natural products targeting MAMs as potential antidepressants to treat depression.
Collapse
Affiliation(s)
- Maohui Yang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China.
| | - Xiaojie Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, Shanxi 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
8
|
Baumann N, Wagener RJ, Javed A, Conti E, Abe P, Lopes A, Sansevrino R, Lavalley A, Magrinelli E, Szalai T, Fuciec D, Ferreira C, Fièvre S, Fouassier A, D'Amico D, Harschnitz O, Jabaudon D. Regional differences in progenitor metabolism shape brain growth during development. Cell 2025:S0092-8674(25)00405-2. [PMID: 40300602 DOI: 10.1016/j.cell.2025.04.003] [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: 08/31/2023] [Revised: 12/17/2024] [Accepted: 04/02/2025] [Indexed: 05/01/2025]
Abstract
Mammals have particularly large forebrains compared with other brain parts, yet the developmental mechanisms underlying this regional expansion remain poorly understood. Here, we provide a single-cell-resolution birthdate atlas of the mouse brain (www.neurobirth.org), which reveals that while hindbrain neurogenesis is transient and restricted to early development, forebrain neurogenesis is temporally sustained through reduced consumptive divisions of ventricular zone progenitors. This atlas additionally reveals region-specific patterns of direct and indirect neurogenesis. Using single-cell RNA sequencing, we identify evolutionarily conserved cell-cycle programs and metabolism-related molecular pathways that control regional temporal windows of proliferation. We identify the late neocortex-enriched mitochondrial protein FAM210B as a key regulator using in vivo gain- and loss-of-function experiments. FAM210B elongates mitochondria and increases lactate production, which promotes progenitor self-replicative divisions and, ultimately, the larger clonal size of their progeny. Together, these findings indicate that spatiotemporal heterogeneity in mitochondrial function regulates regional progenitor cycling behavior and associated clonal neuronal production during brain development.
Collapse
Affiliation(s)
- Natalia Baumann
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Robin J Wagener
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Awais Javed
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Eleonora Conti
- Human Technopole, Viale Rita Levi-Montalcini, 1, 20157 Milan, Italy
| | - Philipp Abe
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland; Institute for Clinical Genetics, University Hospital Carl Gustav Carus at TUD Dresden University of Technology and Faculty of Medicine of TUD Dresden University of Technology, Dresden, Germany
| | - Andrea Lopes
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | | | - Adrien Lavalley
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Elia Magrinelli
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Timea Szalai
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Daniel Fuciec
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Clothilde Ferreira
- Department of Genetics and Evolution, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Sabine Fièvre
- Department of Genetics and Evolution, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | | | - Davide D'Amico
- Amazentis SA, EPFL Innovation Park, Lausanne, Switzerland
| | | | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland; Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland; Université Paris Cité, Imagine Institute, Paris, France.
| |
Collapse
|
9
|
Johnson KJ, Johnson K, Grant A, Taglialatela G, Micci MA. Photobiomodulation therapy increases neural stem cell pool in aged 3xTg-AD mice. PLoS One 2025; 20:e0321668. [PMID: 40261888 PMCID: PMC12013953 DOI: 10.1371/journal.pone.0321668] [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: 12/20/2024] [Accepted: 03/10/2025] [Indexed: 04/24/2025] Open
Abstract
Presently approved Alzheimer's Disease (AD) therapeutics are designed for targeted removal of the AD-related toxic protein aggregate amyloid-β (Aβ) and have only shown moderate efficacy at slowing disease progression. Reversal of cognitive decline requires both removal of toxic aggregates and repair of the cellular systems damaged by decades of exposure to these aggregates. Adult hippocampal neurogenesis (AHN) is one such system that is known to be affected early and severely in the development of AD. Moreover, preserved AHN is associated with cognitive resilience to AD neuropathology. Therefore, targeted therapies to improve or enhance neurogenesis should be considered in addition to the removal of toxic protein aggregates. Photobiomodulation (PBM) using 670 nm LED light has been shown to induce synaptic resilience to and removal of AD-related toxic protein aggregates. In this study, we aimed to assess the effect of PBM on a mouse model of advanced AD neuropathology. Transgenic 3xTg-AD mice (15- to 17-month old) were randomized to receive PBM or SHAM therapy for one month, followed by neuropathological assessments. Our results show that one month of PBM therapy reduces hyperphosphorylated tau burden and partially rescues AHN in aged 3xTg-AD mice as compared to SHAM-treated transgenic mice. These data support the notion that PBM has the potential to be an effective non-invasive therapy to help preserve AHN and reduce cognitive dysfunction in moderate to advanced AD.
Collapse
Affiliation(s)
- Kevin J. Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Neurobiology, Neuroscience Graduate Program, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Auston Grant
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Giulio Taglialatela
- The Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
10
|
Papetti AV, Jin M, Ma Z, Stillitano AC, Jiang P. Chimeric brain models: Unlocking insights into human neural development, aging, diseases, and cell therapies. Neuron 2025:S0896-6273(25)00256-9. [PMID: 40300597 DOI: 10.1016/j.neuron.2025.03.036] [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: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Human-rodent chimeric brain models serve as a unique platform for investigating the pathophysiology of human cells within a living brain environment. These models are established by transplanting human tissue- or human pluripotent stem cell (hPSC)-derived macroglial, microglial, or neuronal lineage cells, as well as cerebral organoids, into the brains of host animals. This approach has opened new avenues for exploring human brain development, disease mechanisms, and regenerative processes. Here, we highlight recent advancements in using chimeric models to study human neural development, aging, and disease. Additionally, we explore the potential applications of these models for studying human glial cell-replacement therapies, studying in vivo human glial-to-neuron reprogramming, and harnessing single-cell omics and advanced functional assays to uncover detailed insights into human neurobiology. Finally, we discuss strategies to enhance the precision and translational relevance of these models, expanding their impact in stem cell and neuroscience research.
Collapse
Affiliation(s)
- Ava V Papetti
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Alessandro C Stillitano
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA.
| |
Collapse
|
11
|
Mostajo-Radji MA, Leon WRM, Breevoort A, Gonzalez-Ferrer J, Schweiger HE, Lehrer J, Zhou L, Schmitz MT, Perez Y, Mukhtar T, Robbins A, Chu J, Andrews MG, Sullivan FN, Tejera D, Choy EC, Paredes MF, Teodorescu M, Kriegstein AR, Alvarez-Buylla A, Pollen AA. Fate plasticity of interneuron specification. iScience 2025; 28:112295. [PMID: 40264797 PMCID: PMC12013500 DOI: 10.1016/j.isci.2025.112295] [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: 10/15/2024] [Revised: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Neuronal subtype generation in the mammalian central nervous system is governed by competing genetic programs. The medial ganglionic eminence (MGE) produces two major cortical interneuron (IN) populations, somatostatin (Sst) and parvalbumin (Pvalb), which develop on different timelines. The extent to which external signals influence these identities remains unclear. Pvalb-positive INs are crucial for cortical circuit regulation but challenging to model in vitro. We grafted mouse MGE progenitors into diverse 2D and 3D co-culture systems, including mouse and human cortical, MGE, and thalamic models. Strikingly, only 3D human corticogenesis models promoted efficient, non-autonomous Pvalb differentiation, characterized by upregulation of Pvalb maturation markers, downregulation of Sst-specific markers, and the formation of perineuronal nets. Additionally, lineage-traced postmitotic Sst-positive INs upregulated Pvalb when grafted onto human cortical models. These findings reveal unexpected fate plasticity in MGE-derived INs, suggesting that their identities can be dynamically shaped by the environment.
Collapse
Affiliation(s)
- Mohammed A. Mostajo-Radji
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Walter R. Mancia Leon
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arnar Breevoort
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jesus Gonzalez-Ferrer
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hunter E. Schweiger
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Julian Lehrer
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Li Zhou
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew T. Schmitz
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yonatan Perez
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tanzila Mukhtar
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ash Robbins
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Julia Chu
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Madeline G. Andrews
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Dario Tejera
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric C. Choy
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mercedes F. Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mircea Teodorescu
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Arnold R. Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alex A. Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Feng Z, Hou Y, Yu C, Li T, Fu H, Lv F, Li P. Mitophagy in perioperative neurocognitive disorder: mechanisms and therapeutic strategies. Eur J Med Res 2025; 30:270. [PMID: 40211418 PMCID: PMC11987364 DOI: 10.1186/s40001-025-02400-1] [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: 12/04/2024] [Accepted: 02/20/2025] [Indexed: 04/13/2025] Open
Abstract
Perioperative neurocognitive disorder (PND) is a common neurological complication after surgery/anesthesia in elderly patients that affect postoperative outcome and long-term quality of life, which increases the cost of family and social resources. The pathological mechanism of PND is complex and not fully understood, and the methods of prevention and treatment of PND are very limited, so it is particularly important to analyze the mechanism of PND. Research indicates that mitochondrial dysfunction is pivotal in the initiation and progression of PND, although the precise mechanisms remain elusive and could involve disrupted mitophagy. We reviewed recent studies on the link between mitophagy and PND, highlighting the role of key proteins in abnormal mitophagy and discussing therapeutic strategies aimed at mitophagy regulation. This provides insights into the mechanisms underlying PND and potential therapeutic targets.
Collapse
Affiliation(s)
- Zhen Feng
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, 301 Nancheng Avenue, Nan'an District, Chongqing, Chongqing, 400000, People's Republic of China
| | - Yan Hou
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, 301 Nancheng Avenue, Nan'an District, Chongqing, Chongqing, 400000, People's Republic of China
| | - Chang Yu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Ting Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Haoyang Fu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Feng Lv
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China.
| | - Ping Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
13
|
Chen ZH, Pan TB, Zhang YH, Wang B, Sun XL, Gao M, Sun Y, Xu M, Han S, Shi X, Correa-da-Silva F, Yang C, Guo J, Wu H, Li YZ, Liu XQ, Gao F, Xu Z, Xu S, Liu X, Zhu Y, Deng Z, Liu S, Zhou Y, Yi CX, Liu L, Wu QF. Transcriptional conservation and evolutionary divergence of cell types across mammalian hypothalamus development. Dev Cell 2025:S1534-5807(25)00156-X. [PMID: 40203835 DOI: 10.1016/j.devcel.2025.03.009] [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: 05/31/2024] [Revised: 02/07/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025]
Abstract
The hypothalamus, an "ancient" subcortical brain structure, maintains physiological homeostasis and controls native behaviors. The evolution of homeostatic regulation and behavioral control in mammals may rely on adaptable neuronal identity establishment but conserved neural patterning mechanisms during neurodevelopment. Here, we combined single-cell, single-nucleus, and spatial transcriptomic datasets to map the spatial patterning of diverse progenitor domains and reconstruct their neurogenic lineages in the developing human and mouse hypothalamus. While the regional organizers orchestrating neural patterning are conserved between primates and rodents, we identified a human-enriched neuronal subtype and found a substantial increase in neuromodulatory gene expression among human neurons. Furthermore, cross-species comparison demonstrated a potential redistribution of two neuroendocrine neuronal subtypes and a shift in inter-transmitter and transmitter-peptide coupling within hypothalamic dopamine neurons. Together, our study lays a critical foundation for understanding cellular development and evolution of the mammalian hypothalamus.
Collapse
Affiliation(s)
- Zhen-Hua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | | | - Yu-Hong Zhang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Ben Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xue-Lian Sun
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | | | - Yang Sun
- BGI Research, Beijing 102601, China
| | - Mingrui Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | | | - Xiang Shi
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Felipe Correa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | | | - Junfu Guo
- BGI Research, Beijing 102601, China; BGI Research, Shenzhen 518083, China
| | - Haoda Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Zheng Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiu-Qin Liu
- Department of Obstetrics and Gynecology, Baoding Second Central Hospital, Baoding 072750, China
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhiheng Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Shengjin Xu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin Liu
- BGI Research, Beijing 102601, China
| | - Ying Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University Shanghai, Shanghai 200032, China
| | | | | | - Yi Zhou
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | | | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China; Beijing Key Laboratory for Genetics of Birth Defects, Beijing 100045, China.
| |
Collapse
|
14
|
Antico O, Thompson PW, Hertz NT, Muqit MMK, Parton LE. Targeting mitophagy in neurodegenerative diseases. Nat Rev Drug Discov 2025; 24:276-299. [PMID: 39809929 DOI: 10.1038/s41573-024-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Mitochondrial dysfunction is a hallmark of idiopathic neurodegenerative diseases, including Parkinson disease, amyotrophic lateral sclerosis, Alzheimer disease and Huntington disease. Familial forms of Parkinson disease and amyotrophic lateral sclerosis are often characterized by mutations in genes associated with mitophagy deficits. Therefore, enhancing the mitophagy pathway may represent a novel therapeutic approach to targeting an underlying pathogenic cause of neurodegenerative diseases, with the potential to deliver neuroprotection and disease modification, which is an important unmet need. Accumulating genetic, molecular and preclinical model-based evidence now supports targeting mitophagy in neurodegenerative diseases. Despite clinical development challenges, small-molecule-based approaches for selective mitophagy enhancement - namely, USP30 inhibitors and PINK1 activators - are entering phase I clinical trials for the first time.
Collapse
Affiliation(s)
- Odetta Antico
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul W Thompson
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK
| | | | - Miratul M K Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura E Parton
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
15
|
Imada T, Sasaki S, Yamaguchi H, Ueda A, Kawamori D, Katakami N, Shimomura I. Imeglimin, unlike metformin, does not perturb differentiation of human induced pluripotent stem cells towards pancreatic β-like cells and rather enhances gain in β cell identity gene sets. J Diabetes Investig 2025; 16:584-597. [PMID: 39829307 PMCID: PMC11970301 DOI: 10.1111/jdi.14410] [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: 07/23/2024] [Revised: 12/26/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
AIMS/INTRODUCTION Metformin treatment for hyperglycemia in pregnancy (HIP) beneficially improves maternal glucose metabolism and reduces perinatal complications. However, metformin could impede pancreatic β cell development via impaired mitochondrial function. A new anti-diabetes drug imeglimin, developed based on metformin, improves mitochondrial function. Here we examine the effect of imeglimin on β cell differentiation using human induced pluripotent stem cell (iPSC)-derived pancreatic islet-like spheroid (SC-islet) models. MATERIALS AND METHODS Human iPSCs are differentiated into SC-islets by three-dimensional culture with and without imeglimin or metformin. Differentiation efficiencies of SC-islets were analyzed by flow cytometry, immunostaining, quantitative PCR, and insulin secretion assay. RNA sequencing and oxygen consumption rate were obtained for further characterization of SC-islets. SC-islets were cultured with proinflammatory cytokines, in part mimicking the uterus environment in HIP. RESULTS Metformin perturbed SC-islet differentiation while imeglimin did not alter it. Furthermore, imeglimin enhanced the gene expressions of β cell lineage markers. Maintenance of mitochondrial function and optimization of TGF-β and Wnt signaling were considered potential mechanisms for augmented β cell maturation by imeglimin. In the presence of proinflammatory cytokines, imeglimin ameliorated β cell differentiation impaired by cytokines and metformin. CONCLUSIONS Imeglimin does not perturb differentiation of SC-islet cells and rather enhances gain in β cell identity gene sets in contrast to metformin. This may lead to the improvement of in vitro β cell differentiation protocols.
Collapse
Affiliation(s)
- Tasuku Imada
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Shugo Sasaki
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Hiroki Yamaguchi
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Ayaka Ueda
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Dan Kawamori
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
- Medical Education Center, Faculty of MedicineOsaka UniversityOsakaJapan
- Postgraduate Medical Training CenterOsaka University Hospital, Osaka UniversityOsakaJapan
| | - Naoto Katakami
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Iichiro Shimomura
- Department of Metabolic MedicineOsaka University Graduate School of MedicineOsakaJapan
| |
Collapse
|
16
|
Sasidharan Y, Suryavanshi V, Smit ME. A space for time. Exploring temporal regulation of plant development across spatial scales. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2025; 122:e70130. [PMID: 40163327 PMCID: PMC11956849 DOI: 10.1111/tpj.70130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Plants continuously undergo change during their life cycle, experiencing dramatic phase transitions altering plant form, and regulating the assignment and progression of cell fates. The relative timing of developmental events is tightly controlled and involves integration of environmental, spatial, and relative age-related signals and actors. While plant phase transitions have been studied extensively and many of their regulators have been described, less is known about temporal regulation on a smaller, cell-level scale. Here, using examples from both plant and animal systems, we outline time-dependent changes. Looking at systemic scale changes, we discuss the timing of germination, juvenile-to-adult transition, flowering, and senescence, together with regeneration timing. Switching to temporal regulation on a cellular level, we discuss several instances from the animal field in which temporal control has been examined extensively at this scale. Then, we switch back to plants and summarize examples where plant cell-level changes are temporally regulated. As time cannot easily be separated from signaling derived from the environment and tissue context, we next discuss factors that have been implicated in controlling the timing of developmental events, reviewing temperature, photoperiod, nutrient availability, as well as tissue context and mechanical cues on the cellular scale. Afterwards, we provide an overview of mechanisms that have been shown or implicated in the temporal control of development, considering metabolism, division control, mobile signals, epigenetic regulation, and the action of transcription factors. Lastly, we look at remaining questions for the future study of developmental timing in plants and how recent technical advancement can enable these efforts.
Collapse
Affiliation(s)
- Yadhusankar Sasidharan
- Department of Developmental Genetics, Centre for Plant Molecular Biology (ZMBP)Eberhard Karls UniversityTuebingenD‐72076Germany
| | - Vijayalakshmi Suryavanshi
- Department of Developmental Genetics, Centre for Plant Molecular Biology (ZMBP)Eberhard Karls UniversityTuebingenD‐72076Germany
| | - Margot E. Smit
- Department of Developmental Genetics, Centre for Plant Molecular Biology (ZMBP)Eberhard Karls UniversityTuebingenD‐72076Germany
| |
Collapse
|
17
|
Carter AC, Koreman GT, Petrocelli JE, Robb JE, Bushinsky EM, Trowbridge SK, Kingsley DM, Walsh CA, Song JHT, Greenberg ME. FOS binding sites are a hub for the evolution of activity-dependent gene regulatory programs in human neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646366. [PMID: 40236085 PMCID: PMC11996375 DOI: 10.1101/2025.03.31.646366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
After birth, sensory inputs to neurons trigger the induction of activity-dependent genes (ADGs) that mediate many aspects of neuronal maturation and plasticity. To identify human-specific ADGs, we characterized these genes in human-chimpanzee tetraploid neurons. We identified 235 ADGs that are differentially expressed between human and chimpanzee neurons and found that their nearby regulatory sites are species-biased in their binding of the transcription factor FOS. An assessment of these sites revealed that many are enriched for single nucleotide variants that promote or eliminate FOS binding in human neurons. Disrupting the function of individual species-biased FOS-bound enhancers diminishes expression of nearby genes and affects the firing dynamics of human neurons. Our findings indicate that FOS-bound enhancers are frequent sites of evolution and that they regulate human-specific ADGs that may contribute to the unusually protracted and complex process of postnatal human brain development.
Collapse
|
18
|
Lv J, Liang S, Qin P, Liu X, Ge X, Guo Y, Xia S, Jing W, Lu Y, Zhang T, Li H. WWC1 mutation drives dopamine dysregulation and synaptic imbalance in Tourette's syndrome. SCIENCE ADVANCES 2025; 11:eadr4588. [PMID: 40153501 PMCID: PMC11952098 DOI: 10.1126/sciadv.adr4588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/25/2025] [Indexed: 03/30/2025]
Abstract
Tourette's syndrome (TS) is a major neurodevelopmental disorder characterized by childhood-onset motor and vocal tics. A W88C mutation in WWC1 gene is a notable risk factor for TS, but the underlying molecular mechanisms remain unclear due to the lack of suitable animal models. Here, we generate a mutant mouse line with human W88C mutation (W88CMut mice), which exhibits behavioral deficits similar to those observed in patients with TS, including repetitive motor behaviors and sensorimotor gating abnormalities. The W88C mutation leads to the degradation of kidney and brain (KIBRA) protein via a proteasomal pathway, evokes dopamine release in the dorsal striatum, and disrupts synaptic function through the dysregulation of Hippo pathway. Neuron-specific overexpression of wild-type WWC1 rescues synaptic and behavioral phenotypes in W88CMut mice. Together, this study not only provides a valuable mouse model for studying TS but also offers fresh insights into the molecular and synaptic mechanisms underlying neurodevelopmental abnormalities in TS.
Collapse
Affiliation(s)
- Junkai Lv
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiqi Liang
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pengwei Qin
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinlu Liu
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangyu Ge
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yiqing Guo
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shili Xia
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Jing
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Youming Lu
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 4030030, China
| | - Tongmei Zhang
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Histology and embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Li
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
19
|
Mil J, Soto JA, Matulionis N, Krall A, Day F, Stiles L, Montales KP, Azizad DJ, Gonzalez CE, Nano PR, Martija AA, Perez-Ramirez CA, Nguyen CV, Kan RL, Andrews MG, Christofk HR, Bhaduri A. Metabolic Atlas of Early Human Cortex Identifies Regulators of Cell Fate Transitions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642470. [PMID: 40161647 PMCID: PMC11952424 DOI: 10.1101/2025.03.10.642470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Characterization of cell type emergence during human cortical development, which enables unique human cognition, has focused primarily on anatomical and transcriptional characterizations. Metabolic processes in the human brain that allow for rapid expansion, but contribute to vulnerability to neurodevelopmental disorders, remain largely unexplored. We performed a variety of metabolic assays in primary tissue and stem cell derived cortical organoids and observed dynamic changes in core metabolic functions, including an unexpected increase in glycolysis during late neurogenesis. By depleting glucose levels in cortical organoids, we increased outer radial glia, astrocytes, and inhibitory neurons. We found the pentose phosphate pathway (PPP) was impacted in these experiments and leveraged pharmacological and genetic manipulations to recapitulate these radial glia cell fate changes. These data identify a new role for the PPP in modulating radial glia cell fate specification and generate a resource for future exploration of additional metabolic pathways in human cortical development.
Collapse
Affiliation(s)
- Jessenya Mil
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jose A. Soto
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nedas Matulionis
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Abigail Krall
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Francesca Day
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Katrina P. Montales
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daria J. Azizad
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Carlos E. Gonzalez
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Patricia R. Nano
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Antoni A. Martija
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cesar A. Perez-Ramirez
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Claudia V. Nguyen
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ryan L. Kan
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Madeline G. Andrews
- School of Biological and Health Systems Engineering, Arizona State University, Phoenix, AZ, United States
| | - Heather R. Christofk
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
20
|
Zelco A, Joshi A. Single-Cell Analysis of Sex and Gender Differences in the Human Brain During Development and Disease. Cell Mol Neurobiol 2025; 45:20. [PMID: 40016536 PMCID: PMC11868228 DOI: 10.1007/s10571-025-01536-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
Sex and gender (SG) differences in the human brain are of interest to society and science as numerous processes are impacted by them, including brain development, behavior, and diseases. By collecting publicly available single-cell data from the in-utero to elderly age in healthy, Alzheimer's disease and multiple sclerosis samples, we identified and characterized SG-biased genes in ten brain cell types across 9 age and disease groups. Sex and gender differences in the transcriptome were present throughout the lifespan and across all cell types. Although there was limited overlap among SG-biased genes across different age and disease groups, we observed significant functional overlap. Female-biased genes are consistently enriched for brain-related processes, while male-biased genes are enriched for metabolic pathways. Additionally, mitochondrial genes showed a consistent female bias across cell types. We also found that androgen response elements (not estrogen) were significantly enriched in both male- and female-biased genes, and thymosin hormone targets being consistently enriched only in male-biased genes. We systematically characterised SG differences in brain development and brain-related disorders at a single-cell level, by analysing a total of publicly available 419,885 single nuclei from 161 human brain samples (72 females, 89 males). The significant enrichment of androgen (not estrogen) response elements in both male- and female-biased genes suggests that androgens are important regulators likely establishing these SG differences. Finally, we provide full characterization of SG-biased genes at different thresholds for the scientific community as a web resource.
Collapse
Affiliation(s)
- Aura Zelco
- Department of Clinical Science, Computational Biology Unit, University of Bergen, Bergen, Norway.
| | - Anagha Joshi
- Department of Clinical Science, Computational Biology Unit, University of Bergen, Bergen, Norway.
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India.
| |
Collapse
|
21
|
Venkatesan S, Werner JM, Li Y, Gillis J. Cell Type-Agnostic Transcriptomic Signatures Enable Uniform Comparisons of Neurodevelopment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639936. [PMID: 40060479 PMCID: PMC11888278 DOI: 10.1101/2025.02.24.639936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Single-cell transcriptomics has revolutionized our understanding of neurodevelopmental cell identities, yet, predicting a cell type's developmental state from its transcriptome remains a challenge. We perform a meta-analysis of developing human brain datasets comprising over 2.8 million cells, identifying both tissue-level and cell-autonomous predictors of developmental age. While tissue composition predicts age within individual studies, it fails to generalize, whereas specific cell type proportions reliably track developmental time across datasets. Training regularized regression models to infer cell-autonomous maturation, we find that a cell type-agnostic model achieves the highest accuracy (error = 2.6 weeks), robustly capturing developmental dynamics across diverse cell types and datasets. This model generalizes to human neural organoids, accurately predicting normal developmental trajectories (R = 0.91) and disease-induced shifts in vitro. Furthermore, it extends to the developing mouse brain, revealing an accelerated developmental tempo relative to humans. Our work provides a unified framework for comparing neurodevelopment across contexts, model systems, and species.
Collapse
Affiliation(s)
- Sridevi Venkatesan
- Department of Physiology, University of Toronto, Canada
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Jonathan M Werner
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Yun Li
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Canada
| | - Jesse Gillis
- Department of Physiology, University of Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Canada
| |
Collapse
|
22
|
Yang R, Ji F, Jiao J. Early central nervous system development and neuron regeneration. Curr Opin Genet Dev 2025; 90:102286. [PMID: 39637751 DOI: 10.1016/j.gde.2024.102286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/25/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
The nervous system is the most complex system in the human body, and the normal development of the central nervous system (CNS) is essential for maintaining the healthy life activities of the individual. CNS development requires the orchestration of multiple internal or external or direct or indirect factors to regulate neural stem cell fate specification. Here, we provide a broad overview of the regulatory system of nerve cell fate decisions and discuss the latest technological approaches to achieve neural regeneration. Understanding the CNS development and regeneration mechanisms has shifted the paradigm from traditional experiments to high-throughput sequencing.
Collapse
Affiliation(s)
- Runhua Yang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing 100101, China
| | - Fen Ji
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing 100101, China
| | - Jianwei Jiao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing 100101, China.
| |
Collapse
|
23
|
Caporale N, Leonardi O, Villa CE, Vitriolo A, Boeckx C, Testa G. Tile by tile: capturing the evolutionary mosaic of human conditions. Curr Opin Genet Dev 2025; 90:102297. [PMID: 39705881 DOI: 10.1016/j.gde.2024.102297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024]
Abstract
The collection of Homo sapiens anatomical hallmarks hypothesized to support the 'human condition' did not appear at one specific time and place, but gradually, creating a reticulate evolutionary trajectory. The recent reconstruction of migration patterns and gene flows across different hominin species and populations draws a mosaic that we contend can be illuminated by genomic comparisons and specific experiments. Here, we first review key discoveries that could allow this experimental endeavor by describing recent advances in a variety of fields, stressing the importance of charting the current human neurodiversity as an interpretive substrate for evolutionary changes. Then, we identify key cellular and molecular observables. Finally, given the vast amount of possible variants, we focus the discussion on technologies that could allow their interrogation in a way that is compatible with the staggering amount of contemporary genomic and phenotypic characterization.
Collapse
Affiliation(s)
- Nicolò Caporale
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@NicoloCaporale
| | - Oliviero Leonardi
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@OlivieroLeonar2
| | - Carlo Emanuele Villa
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@CarloEmanueleV1
| | - Alessandro Vitriolo
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy. https://twitter.com/@AVitriolScience
| | - Cedric Boeckx
- University of Barcelona, 08007 Barcelona, Spain; University of Barcelona Institute of Complex Systems, 08007 Barcelona, Spain; University of Barcelona Institute of Neurosciences, 08007 Barcelona, Spain; Catalan Institute for Research and Advanced Studies (ICREA), 08007 Barcelona, Spain.
| | - Giuseppe Testa
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| |
Collapse
|
24
|
Yang X, Zhou B. Unleashing metabolic power for axonal regeneration. Trends Endocrinol Metab 2025; 36:161-175. [PMID: 39069446 DOI: 10.1016/j.tem.2024.07.001] [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: 05/02/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024]
Abstract
Axon regeneration requires the mobilization of intracellular resources, including proteins, lipids, and nucleotides. After injury, neurons need to adapt their metabolism to meet the biosynthetic demands needed to achieve axonal regeneration. However, the exact contribution of cellular metabolism to this process remains elusive. Insights into the metabolic characteristics of proliferative cells may illuminate similar mechanisms operating in axon regeneration; therefore, unraveling previously unappreciated roles of metabolic adaptation is critical to achieving neuron regrowth, which is connected to the therapeutic strategies for neurological conditions necessitating nerve repairs, such as spinal cord injury and stroke. Here, we outline the metabolic role in axon regeneration and discuss factors enhancing nerve regrowth, highlighting potential novel metabolic treatments for restoring nerve function.
Collapse
Affiliation(s)
- Xiaoyan Yang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China; School of Engineering Medicine, Beihang University, Beijing 100191, China.
| |
Collapse
|
25
|
Matsuda M, Lázaro J, Ebisuya M. Metabolic activities are selective modulators for individual segmentation clock processes. Nat Commun 2025; 16:845. [PMID: 39833174 PMCID: PMC11746943 DOI: 10.1038/s41467-025-56120-5] [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: 06/04/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Numerous cellular and molecular processes during embryonic development prompt the fundamental question of how their tempos are coordinated and whether a common global modulator exists. While the segmentation clock tempo scales with the kinetics of gene expression and degradation processes of the core clock gene Hes7 across mammals, the coordination of these processes remains unclear. This study examines whether metabolic activities serve as a global modulator for the segmentation clock, finding them to be selective instead. Several metabolic inhibitions extend the clock period but affect key processes differently: glycolysis inhibition slows Hes7 protein degradation and production delay without altering intron delay, while electron transport chain inhibition extends intron delay without influencing the other processes. Combinations of distinct metabolic inhibitions exhibit synergistic effects. We propose that the scaled kinetics of segmentation clock processes across species may result from combined selective modulators shaped by evolutionary constraints, rather than a single global modulator.
Collapse
Affiliation(s)
- Mitsuhiro Matsuda
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain.
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
| | - Jorge Lázaro
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Miki Ebisuya
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain.
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
26
|
Winden KD, Gisser I, Sahin M. Using cortical organoids to understand the pathogenesis of malformations of cortical development. Front Neurosci 2025; 18:1522652. [PMID: 39881808 PMCID: PMC11774837 DOI: 10.3389/fnins.2024.1522652] [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: 11/04/2024] [Accepted: 12/31/2024] [Indexed: 01/31/2025] Open
Abstract
Malformations of cortical development encompass a broad range of disorders associated with abnormalities in corticogenesis. Widespread abnormalities in neuronal formation or migration can lead to small head size or microcephaly with disorganized placement of cell types. Specific, localized malformations are termed focal cortical dysplasias (FCD). Neurodevelopmental disorders are common in all types of malformations of cortical development with the most prominent being refractory epilepsy, behavioral disorders such as autism spectrum disorder (ASD), and learning disorders. Several genetic pathways have been associated with these disorders from control of cell cycle and cytoskeletal dynamics in global malformations to variants in growth factor signaling pathways, especially those interacting with the mechanistic target of rapamycin (mTOR), in FCDs. Despite advances in understanding these disorders, the underlying developmental pathways that lead to lesion formation and mechanisms through which defects in cortical development cause specific neurological symptoms often remains unclear. One limitation is the difficulty in modeling these disorders, as animal models frequently do not faithfully mirror the human phenotype. To circumvent this obstacle, many investigators have turned to three-dimensional human stem cell models of the brain, known as organoids, because they recapitulate early neurodevelopmental processes. High throughput analysis of these organoids presents a promising opportunity to model pathophysiological processes across the breadth of malformations of cortical development. In this review, we highlight advances in understanding the pathophysiology of brain malformations using organoid models.
Collapse
Affiliation(s)
| | | | - Mustafa Sahin
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
27
|
Lichtarge J, Cappuccio G, Pati S, Dei-Ampeh AK, Sing S, Ma L, Liu Z, Maletic-Savatic M. MetaboLINK is a novel algorithm for unveiling cell-specific metabolic pathways in longitudinal datasets. Front Neurosci 2025; 18:1520982. [PMID: 39872998 PMCID: PMC11769959 DOI: 10.3389/fnins.2024.1520982] [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: 11/01/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction In the rapidly advancing field of 'omics research, there is an increasing demand for sophisticated bioinformatic tools to enable efficient and consistent data analysis. As biological datasets, particularly metabolomics, become larger and more complex, innovative strategies are essential for deciphering the intricate molecular and cellular networks. Methods We introduce a pioneering analytical approach that combines Principal Component Analysis (PCA) with Graphical Lasso (GLASSO). This method is designed to reduce the dimensionality of large datasets while preserving significant variance. For the first time, we applied the PCA-GLASSO algorithm (i.e., MetaboLINK) to metabolomics data derived from Nuclear Magnetic Resonance (NMR) spectroscopy performed on neural cells at various developmental stages, from human embryonic stem cells to neurons. Results The MetaboLINK analysis of longitudinal metabolomics data has revealed distinct pathways related to amino acids, lipids, and energy metabolism, uniquely associated with specific cell progenies. These findings suggest that different metabolic pathways play a critical role at different stages of cellular development, each contributing to diverse cellular functions. Discussion Our study demonstrates the efficacy of the MetaboLINK approach in analyzing NMR-based longitudinal metabolomic datasets, highlighting key metabolic shifts during cellular transitions. We share the methodology and the code to advance general 'omics research, providing a powerful tool for dissecting large datasets in neurobiology and other fields.
Collapse
Affiliation(s)
- Jared Lichtarge
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
| | - Gerarda Cappuccio
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Soumya Pati
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Alfred Kwabena Dei-Ampeh
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Senghong Sing
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
- College of Natural Sciences and Mathematics, University of Houston, Houston, TX, United States
| | - LiHua Ma
- Shared Equipment Authority, Rice University, Houston, TX, United States
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
| | - Mirjana Maletic-Savatic
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
28
|
Cheng W, Yin C, Yu S, Chen X, Hong N, Jin W. scMMO-atlas: a single cell multimodal omics atlas and portal for exploring fine cell heterogeneity and cell dynamics. Nucleic Acids Res 2025; 53:D1186-D1194. [PMID: 39315707 PMCID: PMC11701702 DOI: 10.1093/nar/gkae821] [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: 07/18/2024] [Revised: 08/27/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Single-cell multimodal sequencing parallelly captures multiple modalities of the same cell, providing unparalleled insights into cell heterogeneity and cell dynamics. For example, joint profiling of chromatin accessibility and transcriptome from the same single cell (scATAC + RNA) identified new cell subsets within the well-defined clusters. However, lack of single-cell multimodal omics (scMMO) database has led to data fragmentation, seriously hindering access, utilization and mining of scMMO data. Here, we constructed a scMMO atlas by collecting and integrating various scMMO data, then constructed scMMO database and portal called scMMO-atlas (https://www.biosino.org/scMMO-atlas/). scMMO-atlas includes scATAC + RNA (ISSAAS-seq, SNARE-seq, paired-seq, sci-CAR, scCARE-seq, 10X Multiome and so on), scRNA + protein, scATAC + protein and scTri-modal omics data, with 3 168 824 cells from 27 cell tissues/organs. scMMO-atlas offered an interactive portal for visualization and featured analysis for each modality and the integrated data. Integrated analysis of scATAC + RNA data of mouse cerebral cortex in scMMO-atlas identified more cell subsets compared with unimodal omics data. Among these new cell subsets, there is an early astrocyte subset highly expressed Grm3, called Astro-Grm3. Furthermore, we identified Ex-L6-Tle4-Nrf1, a progenitor of Ex-L6-Tle4, indicating the statistical power provided by the big data in scMMO-atlas. In summary, scMMO-atlas offers cell atlas, database and portal to facilitate data utilization and biological insight.
Collapse
Affiliation(s)
- Wenwen Cheng
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Changhui Yin
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiya Yu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xi Chen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Ni Hong
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenfei Jin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
29
|
Kawatani K, Omana Suarez G, Perkerson RB, Parent EE, Nambara T, Knight JA, Parsons TM, Gupta K, Shue F, Alnobani A, Vibhute P, Cai H, Guerrero-Cázares H, Copland JA, Quiñones-Hinojosa A, Kanekiyo T. Human iPSC-Derived MSCs Induce Neurotrophic Effects and Improve Metabolic Activity in Acute Neuronal Injury Models. J Neurosci 2025; 45:e0606242024. [PMID: 39496487 DOI: 10.1523/jneurosci.0606-24.2024] [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: 03/31/2024] [Revised: 09/15/2024] [Accepted: 10/25/2024] [Indexed: 11/06/2024] Open
Abstract
Mesenchymal stromal cell (MSC) therapy has regenerative potentials to treat various pathological conditions including neurological diseases. MSCs isolated from various organs can differentiate into specific cell types to repair organ damages. However, their paracrine mechanisms are predicted to predominantly mediate their immunomodulatory, proangiogenic, and regenerative properties. While preclinical studies highlight the significant potential of MSC therapy in mitigating neurological damage from stroke and traumatic brain injury, the variability in clinical trial outcomes may stem from the inherent heterogeneity of somatic MSCs. Accumulating evidence has demonstrated that induced pluripotent stem cells (iPSCs) are an ideal alternative resource for the unlimited expansion and biomanufacturing of MSCs. Thus, we investigated how iPSC-derived MSCs (iMSCs) influence properties of iPSC-derived neurons. Our findings demonstrate that the secretome from iMSCs possesses neurotrophic effects, improving neuronal survival and promoting neuronal outgrowth and synaptic activity in vitro. Additionally, the iMSCs enhance metabolic activity via mitochondrial respiration in neurons, both in vitro and in mouse models. Glycolytic pathways also increased following the administration of iMSC secretome to iPSC-derived neurons. Consistently, in vivo experiments showed that intravenous administration of iMSCs compensated for the elevated energetic demand in male mice with irradiation-induced brain injury by restoring synaptic metabolic activity during acute brain damage. 18F-FDG PET imaging also detected an increase in brain glucose uptake following iMSC administration. Together, our results highlight the potential of iMSC-based therapy in treating neuronal damage in various neurological disorders, while paving the way for future research and potential clinical applications of iMSCs in regenerative medicine.
Collapse
Affiliation(s)
- Keiji Kawatani
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Genesis Omana Suarez
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, Florida 32224
| | - Ralph B Perkerson
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida 32224
| | - Ephraim E Parent
- Departments of Radiology, Mayo Clinic, Jacksonville, Florida 32224
| | - Toshihiko Nambara
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | | | - Tammee M Parsons
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida 32224
| | - Kshama Gupta
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Alla Alnobani
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Prasanna Vibhute
- Departments of Radiology, Mayo Clinic, Jacksonville, Florida 32224
| | - Hancheng Cai
- Departments of Radiology, Mayo Clinic, Jacksonville, Florida 32224
| | - Hugo Guerrero-Cázares
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
- Neurosurgery, Mayo Clinic, Jacksonville, Florida 32224
| | - John A Copland
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
| | - Alfredo Quiñones-Hinojosa
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
- Neurosurgery, Mayo Clinic, Jacksonville, Florida 32224
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, Florida 32224
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida 32224
| |
Collapse
|
30
|
Keerthiga R, Xie Y, Pei DS, Fu A. The multifaceted modulation of mitochondrial metabolism in tumorigenesis. Mitochondrion 2025; 80:101977. [PMID: 39505244 DOI: 10.1016/j.mito.2024.101977] [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: 11/24/2023] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Changes in mitochondrial metabolism produce a malignant transformation from normal cells to tumor cells. Mitochondrial metabolism, comprising bioenergetic metabolism, biosynthetic process, biomolecular decomposition, and metabolic signal conversion, obviously forms a unique sign in the process of tumorigenesis. Several oncometabolites produced by mitochondrial metabolism maintain tumor phenotype, which are recognized as tumor indicators. The mitochondrial metabolism synchronizes the metabolic and genetic outcome to the potent tumor microenvironmental signals, thereby further promoting tumor initiation. Moreover, the bioenergetic and biosynthetic metabolism within tumor mitochondria orchestrates dynamic contributions toward cancer progression and invasion. In this review, we describe the contribution of mitochondrial metabolism in tumorigenesis through shaping several hallmarks such as microenvironment modulation, plasticity, mitochondrial calcium, mitochondrial dynamics, and epithelial-mesenchymal transition. The review will provide a new insight into the abnormal mitochondrial metabolism in tumorigenesis, which will be conducive to tumor prevention and therapy through targeting tumor mitochondria.
Collapse
Affiliation(s)
- Rajendiran Keerthiga
- College of Pharmaceutical Science, Southwest University, Chongqing, 400716, China; Department of Computational Biology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Thandalam, Chennai 602105, Tamil Nadu, India
| | - Yafang Xie
- College of Pharmaceutical Science, Southwest University, Chongqing, 400716, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Ailing Fu
- College of Pharmaceutical Science, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
31
|
Rolletschek H, Borisjuk L, Gómez-Álvarez EM, Pucciariello C. Advances in seed hypoxia research. PLANT PHYSIOLOGY 2024; 197:kiae556. [PMID: 39471319 PMCID: PMC11852284 DOI: 10.1093/plphys/kiae556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 11/01/2024]
Abstract
Seeds represent essential stages of the plant life cycle: embryogenesis, the intermittent quiescence phase, and germination. Each stage has its own physiological requirements, genetic program, and environmental challenges. Consequently, the effects of developmental and environmental hypoxia can vary from detrimental to beneficial. Past and recent evidence shows how low-oxygen signaling and metabolic adaptations to hypoxia affect seed development and germination. Here, we review the recent literature on seed biology in relation to hypoxia research and present our perspective on key challenges and opportunities for future investigations.
Collapse
Affiliation(s)
- Hardy Rolletschek
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, 06466 Seeland, Germany
| | - Ljudmilla Borisjuk
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, 06466 Seeland, Germany
| | - Eva María Gómez-Álvarez
- PlantLab, Institute of Plant Sciences, Scuola Superiore Sant'Anna, 56010 Pisa, Italy
- nanoPlant Center @NEST, Institute of Plant Sciences, Scuola Superiore Sant'Anna, 56127 Pisa, Italy
| | - Chiara Pucciariello
- PlantLab, Institute of Plant Sciences, Scuola Superiore Sant'Anna, 56010 Pisa, Italy
- nanoPlant Center @NEST, Institute of Plant Sciences, Scuola Superiore Sant'Anna, 56127 Pisa, Italy
| |
Collapse
|
32
|
Ye J, Jiang H, Tiche S, He C, Liu J, Bian F, Jedoui M, Forgo B, Islam MT, Zhao M, Emengo P, He B, Li Y, Li A, Truong A, Ho J, Simmermaker C, Yang Y, Zhou MN, Hu Z, Svensson K, Cuthbertson D, Hazard F, Xing L, Shimada H, Chiu B. Restoring Mitochondrial Quantity and Quality to Reverse Warburg Effect and Drive Tumor Differentiation. RESEARCH SQUARE 2024:rs.3.rs-5494402. [PMID: 39711563 PMCID: PMC11661309 DOI: 10.21203/rs.3.rs-5494402/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Reduced mitochondrial quality and quantity in tumors is associated with dedifferentiation and increased malignancy. However, it remains unclear how to restore mitochondrial quantity and quality in tumors, and whether mitochondrial restoration can drive tumor differentiation. Our study shows that restoring mitochondrial function using retinoic acid (RA) to boost mitochondrial biogenesis and a mitochondrial uncoupler to enhance respiration synergistically drives neuroblastoma differentiation and inhibits proliferation. U-13C-glucose/glutamine isotope tracing revealed a metabolic shift from the pentose phosphate pathway to oxidative phosphorylation, accelerating the TCA cycle and switching substrate preference from glutamine to glucose. These effects were reversed by ETC inhibitors or in ρ0 cells lacking mtDNA, emphasizing the necessity of mitochondrial function for differentiation. Dietary RA and uncoupler treatment promoted tumor differentiation in an orthotopic neuroblastoma xenograft model, evidenced by neuropil production and Schwann cell recruitment. Single-cell RNA sequencing analysis of the orthotopic xenografts revealed that this strategy effectively eliminated the stem cell population, promoted differentiation, and increased mitochondrial gene signatures along the differentiation trajectory, which could potentially significantly improve patient outcomes. Collectively, our findings establish a mitochondria-centric therapeutic strategy for inducing tumor differentiation, suggesting that maintaining/driving differentiation in tumor requires not only ATP production but also continuous ATP consumption and sustained ETC activity.
Collapse
|
33
|
Gao L, Gao B, Ge W, Li S, Wang F. Stimulated Emission Depletion Imaging Reveals Mitochondrial Phenotypic Heterogeneity under Apoptosis Stimuli across Living Glioma Models. NANO LETTERS 2024; 24:15904-15911. [PMID: 39587402 DOI: 10.1021/acs.nanolett.4c04986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The mitochondrial phenotypes contribute to the understanding of disease mechanisms and treatments, which are typically characterized through the omics methods. However, the high dynamics and phenotypic heterogeneity of mitochondria require high-resolution characterization within individual living cells. Therefore, we introduce a fluorescence analysis method, based on two-color and fluorescence lifetime stimulated emission depletion (STED) super-resolution imaging, to explore mitochondrial phenotypic heterogeneity in human (U87) and mouse (GL261) glioma models. Furthermore, we used rotenone and etoposide to simulate the effects of antitumor drugs, inducing apoptosis through mitochondrial dysfunction, respectively. The two-color labeling introduces intracellular parameters to qualitatively visualize changes in mitochondrial morphology, while fluorescence lifetime reflects the status of mitochondria and their microenvironment from the perspective of probe characteristics. This method reveals mitochondria phenotypic heterogeneity induced by the apoptotic stimuli in human and mouse glioma models from a morphological perspective.
Collapse
Affiliation(s)
- Lu Gao
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Beibei Gao
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Ge
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuxian Li
- College of Chemistry and Chemical Engineering, Xinjiang Normal University, Urumqi 830054, China
| | - Fu Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
34
|
Masin L, Bergmans S, Van Dyck A, Farrow K, De Groef L, Moons L. Local glycolysis supports injury-induced axonal regeneration. J Cell Biol 2024; 223:e202402133. [PMID: 39352499 PMCID: PMC11451009 DOI: 10.1083/jcb.202402133] [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: 02/22/2024] [Revised: 04/09/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024] Open
Abstract
Successful axonal regeneration following injury requires the effective allocation of energy. How axons withstand the initial disruption in mitochondrial energy production caused by the injury and subsequently initiate regrowth is poorly understood. Transcriptomic data showed increased expression of glycolytic genes after optic nerve crush in retinal ganglion cells with the co-deletion of Pten and Socs3. Using retinal cultures in a multicompartment microfluidic device, we observed increased regrowth and enhanced mitochondrial trafficking in the axons of Pten and Socs3 co-deleted neurons. While wild-type axons relied on mitochondrial metabolism, after injury, in the absence of Pten and Socs3, energy production was supported by local glycolysis. Specific inhibition of lactate production hindered injury survival and the initiation of regrowth while slowing down glycolysis upstream impaired regrowth initiation, axonal elongation, and energy production. Together, these observations reveal that glycolytic ATP, combined with sustained mitochondrial transport, is essential for injury-induced axonal regrowth, providing new insights into the metabolic underpinnings of axonal regeneration.
Collapse
Affiliation(s)
- Luca Masin
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Steven Bergmans
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Annelies Van Dyck
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Karl Farrow
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
- Neuro-Electronics Research Flanders, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- imec, Leuven, Belgium
| | - Lies De Groef
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Lieve Moons
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
35
|
Christopoulou E, Charrier C. Molecular mechanisms of the specialization of human synapses in the neocortex. Curr Opin Genet Dev 2024; 89:102258. [PMID: 39255688 DOI: 10.1016/j.gde.2024.102258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/05/2024] [Accepted: 08/18/2024] [Indexed: 09/12/2024]
Abstract
Synapses of the neocortex specialized during human evolution to develop over extended timescales, process vast amounts of information and increase connectivity, which is thought to underlie our advanced social and cognitive abilities. These features reflect species-specific regulations of neuron and synapse cell biology. However, despite growing understanding of the human genome and the brain transcriptome at the single-cell level, linking human-specific genetic changes to the specialization of human synapses has remained experimentally challenging. In this review, we describe recent progress in characterizing divergent morphofunctional and developmental properties of human synapses, and we discuss new insights into the underlying molecular mechanisms. We also highlight intersections between evolutionary innovations and disorder-related dysfunctions at the synapse.
Collapse
Affiliation(s)
- Eirini Christopoulou
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Cécile Charrier
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| |
Collapse
|
36
|
Wang S, Shi X, Xiong T, Chen Q, Yang Y, Chen W, Zhang K, Nan Y, Huang Q, Ai K. Inhibiting Mitochondrial Damage for Efficient Treatment of Cerebral Ischemia-Reperfusion Injury Through Sequential Targeting Nanomedicine of Neuronal Mitochondria in Affected Brain Tissue. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409529. [PMID: 39501980 DOI: 10.1002/adma.202409529] [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: 07/03/2024] [Revised: 10/25/2024] [Indexed: 12/13/2024]
Abstract
Oxidative stress, predominantly from neuronal mitochondrial damage and the resultant cytokine storm, is central to cerebral ischemia-reperfusion injury (CIRI). However, delivering drugs to neuronal mitochondria remains challenging due to the blood-brain barrier (BBB), which impedes drug entry into affected brain tissues. This study introduces an innovative tannic acid (TA) and melanin-modified heteropolyacid nanomedicine (MHT), which highly specifically eliminates the neuronal mitochondrial reactive oxygen radicals burst to efficiently reduce neuronal mitochondrial damage through a strategically designed sequential targeting strategy from affected brain tissue to neuronal mitochondria. TA endows MHT with sequential targeting ability by binding to matrix proteins exposed to the damaged BBB and mitochondrial outer membrane proteins of neurons, while melanin significantly enhances the antioxidant capacity of MHT. Consequently, MHT effectively inhibits neuronal apoptosis by protecting mitochondria and reversing the inflammatory immune environment through the deactivation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. MHT demonstrated a strong therapeutic effect on CIRI, with an ultralow dose (2 mg kg-1) proving effective in reversing the condition. This work not only introduces a new avenue to significantly reduce CIRI through sequential targeting therapy but also offers a new paradigm for treating other ischemia-reperfusion injury diseases.
Collapse
Affiliation(s)
- Shuya Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
| | - Tingli Xiong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
| | - Yongqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
| | - Wensheng Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Kexin Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, 750002, P. R. China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, P. R. China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| |
Collapse
|
37
|
Li W, Xue X, Li X, Wu X, Zhou P, Xia Y, Zhang J, Zhang M, Zhu F. Ancestral retrovirus envelope protein ERVWE1 upregulates circ_0001810, a potential biomarker for schizophrenia, and induces neuronal mitochondrial dysfunction via activating AK2. Cell Biosci 2024; 14:138. [PMID: 39543767 PMCID: PMC11566632 DOI: 10.1186/s13578-024-01318-1] [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: 04/11/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Increasingly studies highlight the crucial role of the ancestral retrovirus envelope protein ERVWE1 in the pathogenic mechanisms of schizophrenia, a severe psychiatric disorder affecting approximately 1% of the global population. Recent studies also underscore the significance of circular RNAs (circRNAs), crucial for neurogenesis and synaptogenesis, in maintaining neuronal functions. However, the precise relationship between ERVWE1 and circRNAs in the etiology of schizophrenia remains elusive. RESULTS This study observed elevated levels of hsa_circ_0001810 (circ_0001810) in the blood samples of schizophrenia patients, displaying a significant positive correlation with ERVWE1 expression. Interestingly, in vivo studies demonstrated that ERVWE1 upregulated circ_0001810 in neuronal cells. Circ_0001810, acting as a competing endogenous RNA (ceRNA), bound to miR-1197 and facilitated the release of adenylate kinase 2 (AK2). The bioinformatics analysis of the schizophrenia datasets revealed increased levels of AK2 and enrichment of mitochondrial dynamics. Notably, miR-1197 was reduced in schizophrenia patients, while AK2 levels were increased. Additionally, AK2 showed positive correlations with ERVWE1 and circ_0001810. Further studies demonstrated that AK2 led to mitochondrial dysfunction, characterized by loss of intracellular ATP, mitochondrial depolarization, and disruption of mitochondrial dynamics. Our comprehensive investigation suggested that ERVWE1 influenced ATP levels, promoted mitochondrial depolarization, and disrupted mitochondrial dynamics through the circ_0001810/AK2 pathway. CONCLUSIONS Circ_0001810 and AK2 were increased in schizophrenia and positively correlated with ERVWE1. Importantly, ERVWE1 triggered mitochondrial dysfunction through circ_0001810/miR-1197/AK2 pathway. Recent focus on the impact of mitochondrial dynamics on schizophrenia development had led to our discovery of a novel mechanism by which ERVWE1 contributed to the etiology of schizophrenia, particularly through mitochondrial dynamics. Moreover, these findings collectively proposed that circ_0001810 might serve as a potential blood-based biomarker for schizophrenia. Consistent with our previous theories, ERVWE1 is increasingly recognized as a promising therapeutic target for schizophrenia.
Collapse
Affiliation(s)
- Wenshi Li
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Xing Xue
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Xuhang Li
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Xiulin Wu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Ping Zhou
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Yaru Xia
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Jiahang Zhang
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Mengqi Zhang
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China
| | - Fan Zhu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Science, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
38
|
Libé-Philippot B, Iwata R, Recupero AJ, Wierda K, Bernal Garcia S, Hammond L, van Benthem A, Limame R, Ditkowska M, Beckers S, Gaspariunaite V, Peze-Heidsieck E, Remans D, Charrier C, Theys T, Polleux F, Vanderhaeghen P. Synaptic neoteny of human cortical neurons requires species-specific balancing of SRGAP2-SYNGAP1 cross-inhibition. Neuron 2024; 112:3602-3617.e9. [PMID: 39406239 PMCID: PMC11546603 DOI: 10.1016/j.neuron.2024.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/09/2024] [Accepted: 08/29/2024] [Indexed: 10/26/2024]
Abstract
Human-specific (HS) genes have been implicated in brain evolution, but their impact on human neuron development and diseases remains unclear. Here, we study SRGAP2B/C, two HS gene duplications of the ancestral synaptic gene SRGAP2A, in human cortical pyramidal neurons (CPNs) xenotransplanted in the mouse cortex. Downregulation of SRGAP2B/C in human CPNs led to strongly accelerated synaptic development, indicating their requirement for the neoteny that distinguishes human synaptogenesis. SRGAP2B/C genes promoted neoteny by reducing the synaptic levels of SRGAP2A,thereby increasing the postsynaptic accumulation of the SYNGAP1 protein, encoded by a major intellectual disability/autism spectrum disorder (ID/ASD) gene. Combinatorial loss-of-function experiments in vivo revealed that the tempo of synaptogenesis is set by the reciprocal antagonism between SRGAP2A and SYNGAP1, which in human CPNs is tipped toward neoteny by SRGAP2B/C. Thus, HS genes can modify the phenotypic expression of genetic mutations leading to ID/ASD through the regulation of human synaptic neoteny.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium
| | - Ryohei Iwata
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium
| | - Aleksandra J Recupero
- Department of Neuroscience, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Keimpe Wierda
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Sergio Bernal Garcia
- Department of Neuroscience, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Luke Hammond
- Department of Neuroscience, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Neurology, The Ohio State University, Wexner Medical School, Columbus, OH, USA
| | - Anja van Benthem
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium
| | - Ridha Limame
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium
| | - Martyna Ditkowska
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Sofie Beckers
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Vaiva Gaspariunaite
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Eugénie Peze-Heidsieck
- Department of Neuroscience, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Daan Remans
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Cécile Charrier
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, Inserm, École Normale Supérieure, PSL Research University, Paris 75005, France
| | - Tom Theys
- Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium; Research Group Experimental Neurosurgery and Neuroanatomy, KUL, 3000 Leuven, Belgium
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Pierre Vanderhaeghen
- VIB-KULeuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium.
| |
Collapse
|
39
|
Qiao L, Yang G, Wang P, Xu C. The potential role of mitochondria in the microbiota-gut-brain axis: Implications for brain health. Pharmacol Res 2024; 209:107434. [PMID: 39332752 DOI: 10.1016/j.phrs.2024.107434] [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: 07/04/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Mitochondria are crucial organelles that regulate cellular energy metabolism, calcium homeostasis, and oxidative stress responses, playing pivotal roles in brain development and neurodegeneration. Concurrently, the gut microbiota has emerged as a key modulator of brain physiology and pathology through the microbiota-gut-brain axis. Recent evidence suggests an intricate crosstalk between the gut microbiota and mitochondrial function, mediated by microbial metabolites that can influence mitochondrial activities in the brain. This review aims to provide a comprehensive overview of the emerging role of mitochondria as critical mediators in the microbiota-gut-brain axis, shaping brain health and neurological disease pathogenesis. We discuss how gut microbial metabolites such as short-chain fatty acids, secondary bile acids, tryptophan metabolites, and trimethylamine N-oxide can traverse the blood-brain barrier and modulate mitochondrial processes including energy production, calcium regulation, mitophagy, and oxidative stress in neurons and glial cells. Additionally, we proposed targeting the mitochondria through diet, prebiotics, probiotics, or microbial metabolites as a promising potential therapeutic approach to maintain brain health by optimizing mitochondrial fitness. Overall, further investigations into how the gut microbiota and its metabolites regulate mitochondrial bioenergetics, dynamics, and stress responses will provide valuable insights into the microbiota-gut-brain axis in both health and disease states.
Collapse
Affiliation(s)
- Lei Qiao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ge Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Peng Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Department of Psychiatry, The Affiliated Xi'an Central Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710000, China
| | - Chunlan Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
40
|
Son G, Na Y, Kim Y, Son JH, Clemenson GD, Schafer ST, Yoo JY, Parylak SL, Paquola A, Do H, Kim D, Ahn I, Ju M, Kang CS, Ju Y, Jung E, McDonald AH, Park Y, Kim G, Paik SB, Hur J, Kim J, Han YM, Lee SH, Gage FH, Kim JS, Han J. miR-124 coordinates metabolic regulators acting at early stages of human neurogenesis. Commun Biol 2024; 7:1393. [PMID: 39455851 PMCID: PMC11511827 DOI: 10.1038/s42003-024-07089-2] [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: 07/28/2023] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic dysregulation of neurons is associated with diverse human brain disorders. Metabolic reprogramming occurs during neuronal differentiation, but it is not fully understood which molecules regulate metabolic changes at the early stages of neurogenesis. In this study, we report that miR-124 is a driver of metabolic change at the initiating stage of human neurogenesis. Proteome analysis has shown the oxidative phosphorylation pathway to be the most significantly altered among the differentially expressed proteins (DEPs) in the immature neurons after the knockdown of miR-124. In agreement with these proteomics results, miR-124-depleted neurons display mitochondrial dysfunctions, such as decreased mitochondrial membrane potential and cellular respiration. Moreover, morphological analyses of mitochondria in early differentiated neurons after miR-124 knockdown result in smaller and less mature shapes. Lastly, we show the potential of identified DEPs as novel metabolic regulators in early neuronal development by validating the effects of GSTK1 on cellular respiration. GSTK1, which is upregulated most significantly in miR-124 knockdown neurons, reduces the oxygen consumption rate of neural cells. Collectively, our data highlight the roles of miR-124 in coordinating metabolic maturation at the early stages of neurogenesis and provide insights into potential metabolic regulators associated with human brain disorders characterized by metabolic dysfunctions.
Collapse
Affiliation(s)
- Geurim Son
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Yongwoo Na
- Center for RNA Research, Institute for Basic Science, Seoul, Korea
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Yongsung Kim
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ji-Hoon Son
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Gregory D Clemenson
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Simon T Schafer
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jong-Yeon Yoo
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Sarah L Parylak
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Apua Paquola
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hyunsu Do
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Dayeon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Insook Ahn
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Mingyu Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Chanhee S Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Younghee Ju
- Department of Biological Sciences, KAIST, Daejeon, Korea
- Sovargen.CO., LTD., Daejeon, Korea
| | - Eunji Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Aidan H McDonald
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Youngjin Park
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
| | - Gilhyun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea
| | - Se-Bum Paik
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
- Department of Brain and Cognitive Sciences, KAIST, Daejeon, Korea
| | - Junho Hur
- College of Medicine, Hanyang University, Seoul, Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Yong-Mahn Han
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea
- Department of Brain and Cognitive Sciences, KAIST, Daejeon, Korea
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jong-Seo Kim
- Center for RNA Research, Institute for Basic Science, Seoul, Korea
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Jinju Han
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
- BioMedical Research Center, KAIST, Daejeon, Korea.
- KAIST Stem Cell Center, KAIST, Daejeon, Korea.
| |
Collapse
|
41
|
Keeney JG, Astling D, Andries V, Vandepoele K, Anderson N, Davis JM, Lopert P, Vandenbussche J, Gevaert K, Staes A, Paukovich N, Vögeli B, Jones KL, van Roy F, Patel M, Sikela JM. Olduvai domain expression downregulates mitochondrial pathways: implications for human brain evolution and neoteny. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619278. [PMID: 39484454 PMCID: PMC11526873 DOI: 10.1101/2024.10.21.619278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Encoded by the NBPF gene family, Olduvai (formerly DUF1220) protein domains have undergone the largest human lineage-specific copy number expansion of any coding region in the genome. Olduvai copy number shows a linear relationship with several brain size-related measures and cortical neuron number among primates and with normal and disease-associated (micro- and macrocephaly) variation in brain size in human populations. While Olduvai domains have been shown to promote proliferation of neural stem cells, the mechanism underlying such effects has remained unclear. Here, we investigate the function of Olduvai by transcriptome and proteome analyses of cells overexpressing NBPF1, a gene encoding 7 Olduvai domains. Our results from both RNAseq and mass spectrometry approaches suggest a potential downregulation of mitochondria. In our proteomics study, a Gene Ontology (GO) enrichment analysis for the downregulated proteins revealed a striking overrepresentation of the biological process related to the mitochondrial electron transport chain (p value: 1.81e-11) and identified deregulation of the NADH dehydrogenase activity (p value: 2.43e-11) as the primary molecular function. We verify the reduction of apparent mitochondria via live-cell imaging experiments. Given these and previous Olduvai findings, we suggest that the Olduvai-mediated, dosage-dependent reduction in available energy via mitochondrial downregulation may have resulted in a developmental slowdown such that the neurogenic window among primates, and most extremely in humans, was expanded over a greater time interval, allowing for production of greater numbers of neurons and a larger brain. We further suggest that such a slowdown may extend to other developmental processes that also exhibit neotenic features.
Collapse
Affiliation(s)
- Jonathon G. Keeney
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David Astling
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vanessa Andries
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Karl Vandepoele
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Nathan Anderson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jonathan M. Davis
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Pamela Lopert
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jonathan Vandenbussche
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
| | - Kris Gevaert
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
| | - An Staes
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- VIB Proteomics Core, 9052 Ghent, Belgium
| | - Natasia Paukovich
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Beat Vögeli
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth l. Jones
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Frans van Roy
- Inflammation Research Center, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Manisha Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James M. Sikela
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Human Medical Genetics and Neuroscience Programs, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
42
|
Lancaster MA. Unraveling mechanisms of human brain evolution. Cell 2024; 187:5838-5857. [PMID: 39423803 PMCID: PMC7617105 DOI: 10.1016/j.cell.2024.08.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/19/2024] [Accepted: 08/28/2024] [Indexed: 10/21/2024]
Abstract
Evolutionary changes in human brain structure and function have enabled our specialized cognitive abilities. How these changes have come about genetically and functionally has remained an open question. However, new methods are providing a wealth of information about the genetic, epigenetic, and transcriptomic differences that set the human brain apart. Combined with in vitro models that allow access to developing brain tissue and the cells of our closest living relatives, the puzzle pieces are now coming together to yield a much more complete picture of what is actually unique about the human brain. The challenge now will be linking these observations and making the jump from correlation to causation. However, elegant genetic manipulations are now possible and, when combined with model systems such as organoids, will uncover a mechanistic understanding of how evolutionary changes at the genetic level have led to key differences in development and function that enable human cognition.
Collapse
Affiliation(s)
- Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge, UK; Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
43
|
Zhao M, Wang J, Zhu S, Wang M, Chen C, Wang L, Liu J. Mitochondrion-based organellar therapies for central nervous system diseases. Cell Commun Signal 2024; 22:487. [PMID: 39390521 PMCID: PMC11468137 DOI: 10.1186/s12964-024-01843-z] [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: 06/26/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
As most traditional drugs used to treat central nervous system (CNS) diseases have a single therapeutic target, many of them cannot treat complex diseases or diseases whose mechanism is unknown and cannot effectively reverse the root changes underlying CNS diseases. This raises the question of whether multiple functional components are involved in the complex pathological processes of CNS diseases. Organelles are the core functional units of cells, and the replacement of damaged organelles with healthy organelles allows the multitargeted and integrated modulation of cellular functions. The development of therapies that target independent functional units in the cell, specifically, organelle-based therapies, is rapidly progressing. This article comprehensively discusses the pathogenesis of mitochondrial homeostasis disorders, which involve mitochondria, one of the most important organelles in CNS diseases, and the machanisms of mitochondrion-based therapies, as well as current preclinical and clinical studies on the efficacy of therapies targeting mitochondrial to treat CNS diseases, to provide evidence for use of organelle-based treatment strategies in the future.
Collapse
Affiliation(s)
- Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Shuaiyu Zhu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Meina Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Chong Chen
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China.
| |
Collapse
|
44
|
Identification of a chemical cocktail that drives the maturation of human neurons. Nat Biotechnol 2024; 42:1502-1503. [PMID: 38168997 DOI: 10.1038/s41587-023-02044-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
|
45
|
Whye D, Norabuena EM, Srinivasan GR, Wood D, Polanco TJ, Makhortova NR, Sahin M, Buttermore ED. A Hybrid 2D-to-3D in vitro Differentiation Platform Improves Outcomes of Cerebral Cortical Organoid Generation in hiPSCs. Curr Protoc 2024; 4:e70022. [PMID: 39400999 DOI: 10.1002/cpz1.70022] [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: 10/15/2024]
Abstract
Three-dimensional (3D) cerebral cortical organoids are popular in vitro cellular model systems widely used to study human brain development and disease, compared to traditional stem cell-derived methods that use two-dimensional (2D) monolayer cultures. Despite the advancements made in protocol development for cerebral cortical organoid derivation over the past decade, limitations due to biological, mechanistic, and technical variables remain in generating these complex 3D cellular systems. Building from our previously established differentiation system, we have made modifications to our existing 3D cerebral cortical organoid protocol that resolve several of these technical and biological challenges when working with diverse groups of human induced pluripotent stem cell (hiPSC) lines. This improved protocol blends a 2D monolayer culture format for the specification of neural stem cells and expansion of neuroepithelial progenitor cells with a 3D system for improved self-aggregation and subsequent organoid development. Furthermore, this "hybrid" approach is amenable to both an accelerated cerebral cortical organoid protocol as well as an alternative long-term differentiation protocol. In addition to establishing a hybrid technical format, this protocol also offers phenotypic and morphological characterization of stage-specific cellular profiles using antibodies and fluorescent-based dyes for live cell imaging. © 2024 Wiley Periodicals LLC. Basic Protocol 1: hiPSC-based 2D monolayer specification into neural stem cells (NSCs) Basic Protocol 2: Serial passaging and 2D monolayer expansion of neuroepithelial progenitor cells (NPCs) Support Protocol 1: Direct cryopreservation and rapid thawing of NSCs and NPCs Basic Protocol 3: Bulk aggregation of 3D neurospheres and accelerated cerebral cortical organoid differentiation Alternate Protocol 1: Bulk aggregation of 3D neurospheres and long-term cerebral cortical organoid differentiation Support Protocol 2: High-throughput 3D neurosphere formation and 2D neurosphere migration assay Support Protocol 3: LIVE/DEAD stain cell imaging assay of 3D neurospheres Support Protocol 4: NeuroFluor NeuO live cell dye for 3D cerebral cortical organoids.
Collapse
Affiliation(s)
- Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Erika M Norabuena
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Gayathri Rajaram Srinivasan
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Delaney Wood
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Taryn J Polanco
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Nina R Makhortova
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Mustafa Sahin
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth D Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
46
|
Zhao HT, Schmidt ER. Human-specific genetic modifiers of cortical architecture and function. Curr Opin Genet Dev 2024; 88:102241. [PMID: 39111228 PMCID: PMC11547859 DOI: 10.1016/j.gde.2024.102241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Evolution of the cerebral cortex is thought to have been critical for the emergence of our cognitive abilities. Major features of cortical evolution include increased neuron number and connectivity and altered morpho-electric properties of cortical neurons. Significant progress has been made in identifying human-specific genetic modifiers (HSGMs), some of which are involved in shaping these features of cortical architecture. But how did these evolutionary changes support the emergence of our cognitive abilities? Here, we highlight recent studies aimed at examining the impact of HSGMs on cortical circuit function and behavior. We also discuss the need for greater insight into the link between evolution of cortical architecture and the functional and computational properties of neuronal circuits, as we seek to provide a neurobiological foundation for human cognition.
Collapse
Affiliation(s)
- Hanzhi T Zhao
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA
| | - Ewoud Re Schmidt
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA.
| |
Collapse
|
47
|
Hergenreder E, Minotti AP, Zorina Y, Oberst P, Zhao Z, Munguba H, Calder EL, Baggiolini A, Walsh RM, Liston C, Levitz J, Garippa R, Chen S, Ciceri G, Studer L. Combined small-molecule treatment accelerates maturation of human pluripotent stem cell-derived neurons. Nat Biotechnol 2024; 42:1515-1525. [PMID: 38168993 PMCID: PMC11348887 DOI: 10.1038/s41587-023-02031-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/13/2023] [Indexed: 01/05/2024]
Abstract
The maturation of human pluripotent stem cell (hPSC)-derived neurons mimics the protracted timing of human brain development, extending over months to years for reaching adult-like function. Prolonged in vitro maturation presents a major challenge to stem cell-based applications in modeling and treating neurological disease. Therefore, we designed a high-content imaging assay based on morphological and functional readouts in hPSC-derived cortical neurons which identified multiple compounds that drive neuronal maturation including inhibitors of lysine-specific demethylase 1 and disruptor of telomerase-like 1 and activators of calcium-dependent transcription. A cocktail of four factors, GSK2879552, EPZ-5676, N-methyl-D-aspartate and Bay K 8644, collectively termed GENtoniK, triggered maturation across all parameters tested, including synaptic density, electrophysiology and transcriptomics. Maturation effects were further validated in cortical organoids, spinal motoneurons and non-neural lineages including melanocytes and pancreatic β-cells. The effects on maturation observed across a broad range of hPSC-derived cell types indicate that some of the mechanisms controlling the timing of human maturation might be shared across lineages.
Collapse
Affiliation(s)
- Emiliano Hergenreder
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Andrew P Minotti
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yana Zorina
- Gene Editing and Screening Core Facility, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Polina Oberst
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Zeping Zhao
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, USA
| | - Elizabeth L Calder
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Arianna Baggiolini
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ryan M Walsh
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Ralph Garippa
- Gene Editing and Screening Core Facility, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Gabriele Ciceri
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA.
- Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA.
| |
Collapse
|
48
|
Wang J, Zhao M, Wang M, Fu D, Kang L, Xu Y, Shen L, Jin S, Wang L, Liu J. Human neural stem cell-derived artificial organelles to improve oxidative phosphorylation. Nat Commun 2024; 15:7855. [PMID: 39245680 PMCID: PMC11381526 DOI: 10.1038/s41467-024-52171-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
Oxidative phosphorylation (OXPHOS) in the mitochondrial inner membrane is a therapeutic target in many diseases. Neural stem cells (NSCs) show progress in improving mitochondrial dysfunction in the central nervous system (CNS). However, translating neural stem cell-based therapies to the clinic is challenged by uncontrollable biological variability or heterogeneity, hindering uniform clinical safety and efficacy evaluations. We propose a systematic top-down design based on membrane self-assembly to develop neural stem cell-derived oxidative phosphorylating artificial organelles (SAOs) for targeting the central nervous system as an alternative to NSCs. We construct human conditionally immortal clone neural stem cells (iNSCs) as parent cells and use a streamlined closed operation system to prepare neural stem cell-derived highly homogenous oxidative phosphorylating artificial organelles. These artificial organelles act as biomimetic organelles to mimic respiration chain function and perform oxidative phosphorylation, thus improving ATP synthesis deficiency and rectifying excessive mitochondrial reactive oxygen species production. Conclusively, we provide a framework for a generalizable manufacturing procedure that opens promising prospects for disease treatment.
Collapse
Affiliation(s)
- Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Meina Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Dong Fu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Lin Kang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Yu Xu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Liming Shen
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Shilin Jin
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, PR China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian City, Liaoning Province, PR China.
| |
Collapse
|
49
|
Ji Y, McLean JL, Xu R. Emerging Human Pluripotent Stem Cell-Based Human-Animal Brain Chimeras for Advancing Disease Modeling and Cell Therapy for Neurological Disorders. Neurosci Bull 2024; 40:1315-1332. [PMID: 38466557 PMCID: PMC11365908 DOI: 10.1007/s12264-024-01189-z] [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: 08/08/2023] [Accepted: 11/23/2023] [Indexed: 03/13/2024] Open
Abstract
Human pluripotent stem cell (hPSC) models provide unprecedented opportunities to study human neurological disorders by recapitulating human-specific disease mechanisms. In particular, hPSC-based human-animal brain chimeras enable the study of human cell pathophysiology in vivo. In chimeric brains, human neural and immune cells can maintain human-specific features, undergo maturation, and functionally integrate into host brains, allowing scientists to study how human cells impact neural circuits and animal behaviors. The emerging human-animal brain chimeras hold promise for modeling human brain cells and their interactions in health and disease, elucidating the disease mechanism from molecular and cellular to circuit and behavioral levels, and testing the efficacy of cell therapy interventions. Here, we discuss recent advances in the generation and applications of using human-animal chimeric brain models for the study of neurological disorders, including disease modeling and cell therapy.
Collapse
Affiliation(s)
- Yanru Ji
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Jenna Lillie McLean
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Ranjie Xu
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
50
|
Zhao Y, Liu K, Wang Y, Ma Y, Guo W, Shi C. Human-mouse chimeric brain models constructed from iPSC-derived brain cells: Applications and challenges. Exp Neurol 2024; 379:114848. [PMID: 38857749 DOI: 10.1016/j.expneurol.2024.114848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.
Collapse
Affiliation(s)
- Ya Zhao
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ke Liu
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Yinghua Wang
- Medical College of Yan'an University, Yan'an 716000, PR China
| | - Yifan Ma
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Wenwen Guo
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Changhong Shi
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|