1
|
Vidman S, Ma YHE, Fullenkamp N, Plant GW. Human induced pluripotent stem cell-derived therapies for regeneration after central nervous system injury. Neural Regen Res 2025; 20:3063-3075. [PMID: 39715081 DOI: 10.4103/nrr.nrr-d-24-00901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024] Open
Abstract
In recent years, the progression of stem cell therapies has shown great promise in advancing the nascent field of regenerative medicine. Considering the non-regenerative nature of the mature central nervous system, the concept that "blank" cells could be reprogrammed and functionally integrated into host neural networks remained intriguing. Previous work has also demonstrated the ability of such cells to stimulate intrinsic growth programs in post-mitotic cells, such as neurons. While embryonic stem cells demonstrated great potential in treating central nervous system pathologies, ethical and technical concerns remained. These barriers, along with the clear necessity for this type of treatment, ultimately prompted the advent of induced pluripotent stem cells. The advantage of pluripotent cells in central nervous system regeneration is multifaceted, permitting differentiation into neural stem cells, neural progenitor cells, glia, and various neuronal subpopulations. The precise spatiotemporal application of extrinsic growth factors in vitro, in addition to microenvironmental signaling in vivo, influences the efficiency of this directed differentiation. While the pluri- or multipotency of these cells is appealing, it also poses the risk of unregulated differentiation and teratoma formation. Cells of the neuroectodermal lineage, such as neuronal subpopulations and glia, have been explored with varying degrees of success. Although the risk of cancer or teratoma formation is greatly reduced, each subpopulation varies in effectiveness and is influenced by a myriad of factors, such as the timing of the transplant, pathology type, and the ratio of accompanying progenitor cells. Furthermore, successful transplantation requires innovative approaches to develop delivery vectors that can mitigate cell death and support integration. Lastly, host immune responses to allogeneic grafts must be thoroughly characterized and further developed to reduce the need for immunosuppression. Translation to a clinical setting will involve careful consideration when assessing both physiologic and functional outcomes. This review will highlight both successes and challenges faced when using human induced pluripotent stem cell-derived cell transplantation therapies to promote endogenous regeneration.
Collapse
Affiliation(s)
- Stephen Vidman
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | | | | | | |
Collapse
|
2
|
Maejima I, Sato K. New aspects of a small GTPase RAB35 in brain development and function. Neural Regen Res 2025; 20:1971-1980. [PMID: 39254551 DOI: 10.4103/nrr.nrr-d-23-01543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/30/2023] [Indexed: 09/11/2024] Open
Abstract
In eukaryotic cells, organelles in the secretory, lysosomal, and endocytic pathways actively exchange biological materials with each other through intracellular membrane trafficking, which is the process of transporting the cargo of proteins, lipids, and other molecules to appropriate compartments via transport vesicles or intermediates. These processes are strictly regulated by various small GTPases such as the RAS-like in rat brain (RAB) protein family, which is the largest subfamily of the RAS superfamily. Dysfunction of membrane trafficking affects tissue homeostasis and leads to a wide range of diseases, including neurological disorders and neurodegenerative diseases. Therefore, it is important to understand the physiological and pathological roles of RAB proteins in brain function. RAB35, a member of the RAB family, is an evolutionarily conserved protein in metazoans. A wide range of studies using cultured mammalian cells and model organisms have revealed that RAB35 mediates various processes such as cytokinesis, endocytic recycling, actin bundling, and cell migration. RAB35 is also involved in neurite outgrowth and turnover of synaptic vesicles. We generated brain-specific Rab35 knockout mice to study the physiological roles of RAB35 in brain development and function. These mice exhibited defects in anxiety-related behaviors and spatial memory. Strikingly, RAB35 is required for the precise positioning of pyramidal neurons during hippocampal development, and thereby for normal hippocampal lamination. In contrast, layer formation in the cerebral cortex occurred superficially, even in the absence of RAB35, suggesting a predominant role for RAB35 in hippocampal development rather than in cerebral cortex development. Recent studies have suggested an association between RAB35 and neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. In this review, we provide an overview of the current understanding of subcellular functions of RAB35. We also provide insights into the physiological role of RAB35 in mammalian brain development and function, and discuss the involvement of RAB35 dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ikuko Maejima
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | | |
Collapse
|
3
|
Jun S, Alderson TH, Malone SM, Harper J, Hunt RH, Thomas KM, Iacono WG, Wilson S, Sadaghiani S. Rapid dynamics of electrophysiological connectome states are heritable. Netw Neurosci 2024; 8:1065-1088. [PMID: 39735507 PMCID: PMC11674403 DOI: 10.1162/netn_a_00391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/17/2024] [Indexed: 12/31/2024] Open
Abstract
Time-varying changes in whole-brain connectivity patterns, or connectome state dynamics, are a prominent feature of brain activity with broad functional implications. While infraslow (<0.1 Hz) connectome dynamics have been extensively studied with fMRI, rapid dynamics highly relevant for cognition are poorly understood. Here, we asked whether rapid electrophysiological connectome dynamics constitute subject-specific brain traits and to what extent they are under genetic influence. Using source-localized EEG connectomes during resting state (N = 928, 473 females), we quantified the heritability of multivariate (multistate) features describing temporal or spatial characteristics of connectome dynamics. States switched rapidly every ∼60-500 ms. Temporal features were heritable, particularly Fractional Occupancy (in theta, alpha, beta, and gamma bands) and Transition Probability (in theta, alpha, and gamma bands), representing the duration spent in each state and the frequency of state switches, respectively. Genetic effects explained a substantial proportion of the phenotypic variance of these features: Fractional Occupancy in beta (44.3%) and gamma (39.8%) bands and Transition Probability in theta (38.4%), alpha (63.3%), beta (22.6%), and gamma (40%) bands. However, we found no evidence for the heritability of dynamic spatial features, specifically states' Modularity and connectivity pattern. We conclude that genetic effects shape individuals' connectome dynamics at rapid timescales, specifically states' overall occurrence and sequencing.
Collapse
Affiliation(s)
- Suhnyoung Jun
- Department of Psychology, University of Illinois Urbana-Champaign, Champaign, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Thomas H. Alderson
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Stephen M. Malone
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Jeremy Harper
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Ruskin H. Hunt
- Institute of Child Development, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Kathleen M. Thomas
- Institute of Child Development, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - William G. Iacono
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Sylia Wilson
- Institute of Child Development, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Sepideh Sadaghiani
- Department of Psychology, University of Illinois Urbana-Champaign, Champaign, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, USA
- Neuroscience Program, University of Illinois Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
4
|
Yang R, Ji F, Jiao J. Early central nervous system development and neuron regeneration. Curr Opin Genet Dev 2024; 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] [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
|
5
|
Libé-Philippot B, Polleux F, Vanderhaeghen P. If you please, draw me a neuron - linking evolutionary tinkering with human neuron evolution. Curr Opin Genet Dev 2024; 89:102260. [PMID: 39357501 PMCID: PMC11625661 DOI: 10.1016/j.gde.2024.102260] [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: 06/14/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024]
Abstract
Animal speciation often involves novel behavioral features that rely on nervous system evolution. Human-specific brain features have been proposed to underlie specialized cognitive functions and to be linked, at least in part, to the evolution of synapses, neurons, and circuits of the cerebral cortex. Here, we review recent results showing that, while the human cortex is composed of a repertoire of cells that appears to be largely similar to the one found in other mammals, human cortical neurons do display specialized features at many levels, from gene expression to intrinsic physiological properties. The molecular mechanisms underlying human species-specific neuronal features remain largely unknown but implicate hominid-specific gene duplicates that encode novel molecular modifiers of neuronal function. The identification of human-specific genetic modifiers of neuronal function brings novel insights on brain evolution and function and, could also provide new insights on human species-specific vulnerabilities to brain disorders.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium; Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille (IBDM), NeuroMarseille, Marseille, France.
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA. https://twitter.com/@fpolleux
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium.
| |
Collapse
|
6
|
Guo X, Lee T, Sun J, Sun J, Cai W, Yang Q, Sun T. Molecular Lineages and Spatial Distributions of Subplate Neurons in the Human Fetal Cerebral Cortex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407137. [PMID: 39495628 DOI: 10.1002/advs.202407137] [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: 06/26/2024] [Revised: 10/22/2024] [Indexed: 11/06/2024]
Abstract
The expansion of neural progenitors and production of distinct neurons are crucial for architectural assembly and formation of connectivity in human brains. Subplate neurons (SPNs) are among the firstborn neurons in the human fetal cerebral cortex, and play a critical role in establishing intra- and extracortical connections. However, little is known about SPN origin and developmental lineages. In this study, spatial landscapes and molecular trajectories of SPNs in the human fetal cortices from gestational weeks (GW) 10 to 25 are created by performing spatial transcriptomics and single-cell RNA sequencing. Genes known to be evolutionarily human-specific and genes associated with extracellular matrices (ECMs) are found to maintain stable proportions of subplate neurons among other neuronal types. Enriched ECM gene expression in SPNs varies in distinct cortical regions, with the highest level in the frontal lobe of human fetal brains. This study reveals molecular origin and lineage specification of subplate neurons in the human fetal cerebral cortices, and highlights underpinnings of SPNs to cortical neurogenesis and early structural folding.
Collapse
Affiliation(s)
- Xueyu Guo
- Center for Precision Medicine, Huaqiao University, Xiamen, Fujian, 361021, China
| | - Trevor Lee
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065, USA
| | - Jason Sun
- Xiamen Institute of Technology Attached School, Xiamen, Fujian, 361005, China
| | - Julianne Sun
- Xiamen Institute of Technology Attached School, Xiamen, Fujian, 361005, China
| | - Wenjie Cai
- Department of Radiation Oncology, First Hospital of Quanzhou, Fujian Medical University, Quanzhou, Fujian, 362046, China
| | - Qingwei Yang
- Department of Neurology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361006, China
| | - Tao Sun
- Center for Precision Medicine, Huaqiao University, Xiamen, Fujian, 361021, China
- School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, 361021, China
| |
Collapse
|
7
|
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
|
8
|
Moss ND, Lollis D, Silver DL. How our brains are built: emerging approaches to understand human-specific features. Curr Opin Genet Dev 2024; 89:102278. [PMID: 39549607 DOI: 10.1016/j.gde.2024.102278] [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: 06/28/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/18/2024]
Abstract
Understanding what makes us uniquely human is a long-standing question permeating fields from genomics, neuroscience, and developmental biology to medicine. The discovery of human-specific genomic sequences has enabled a new understanding of the molecular features of human brain evolution. Advances in sequencing, computational, and in vitro screening approaches collectively reveal new roles of uniquely human sequences in regulating gene expression. Here, we review the landscape of human-specific loci and describe how emerging technologies are being used to understand their molecular functions and impact on brain development. We describe current challenges in the field and the potential of integrating new hypotheses and approaches to propel our understanding of the human brain.
Collapse
Affiliation(s)
- Nicole D Moss
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Davoneshia Lollis
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA. https://twitter.com/@_mlollis
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences and Duke Regeneration Center, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
9
|
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] [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
|
10
|
Stimpson CD, Smaers JB, Raghanti MA, Phillips KA, Jacobs B, Hopkins WD, Hof PR, Sherwood CC. Evolutionary scaling and cognitive correlates of primate frontal cortex microstructure. Brain Struct Funct 2024; 229:1823-1838. [PMID: 37889302 DOI: 10.1007/s00429-023-02719-7] [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: 07/02/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Investigating evolutionary changes in frontal cortex microstructure is crucial to understanding how modifications of neuron and axon distributions contribute to phylogenetic variation in cognition. In the present study, we characterized microstructural components of dorsolateral prefrontal cortex, orbitofrontal cortex, and primary motor cortex from 14 primate species using measurements of neuropil fraction and immunohistochemical markers for fast-spiking inhibitory interneurons, large pyramidal projection neuron subtypes, serotonergic innervation, and dopaminergic innervation. Results revealed that the rate of evolutionary change was similar across these microstructural variables, except for neuropil fraction, which evolves more slowly and displays the strongest correlation with brain size. We also found that neuropil fraction in orbitofrontal cortex layers V-VI was associated with cross-species variation in performance on experimental tasks that measure self-control. These findings provide insight into the evolutionary reorganization of the primate frontal cortex in relation to brain size scaling and its association with cognitive processes.
Collapse
Affiliation(s)
- Cheryl D Stimpson
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
- DoD/USU Brain Tissue Repository and Neuropathology Program, Uniformed Services University (USU), Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Jeroen B Smaers
- Department of Anthropology, Stony Brook University, Stony Brook, NY, USA
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Kimberley A Phillips
- Department of Psychology, Trinity University, San Antonio, TX, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Bob Jacobs
- Department of Psychology, Laboratory of Quantitative Neuromorphology, Colorado College, Colorado Springs, CO, USA
| | - William D Hopkins
- Department of Comparative Medicine, Michale E Keeling Center for Comparative Medicine and Research, M D Anderson Cancer Center, Bastrop, TX, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Center for Discovery and Innovation, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA.
| |
Collapse
|
11
|
He S, Zhang X, Zhu H. Human-specific protein-coding and lncRNA genes cast sex-biased genes in the brain and their relationships with brain diseases. Biol Sex Differ 2024; 15:86. [PMID: 39472939 PMCID: PMC11520681 DOI: 10.1186/s13293-024-00659-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Gene expression shows sex bias in the brain as it does in other organs. Since female and male humans exhibit noticeable differences in emotions, logical thinking, movement, spatial orientation, and even the incidence of neurological disorders, sex biases in the brain are especially interesting, but how they are determined, whether they are conserved or lineage specific, and what the consequences of the biases are, remain poorly explored and understood. METHODS Based on RNA-seq datasets from 16 and 14 brain regions in humans and macaques across developmental periods and from patients with brain diseases, we used linear mixed models (LMMs) to differentiate variations in gene expression caused by factors of interest and confounding factors and identify four types of sex-biased genes. Effect size and confidence in each effect were measured upon the local false sign rate (LFSR). We utilized the biomaRt R package to acquire orthologous genes in humans and macaques from the BioMart Ensembl website. Transcriptional regulation of sex-biased genes by sex hormones and lncRNAs were analyzed using the CellOracle, GENIE3, and Longtarget programs. Sex-biased genes' functions were revealed by gene set enrichment analysis using multiple methods. RESULTS Lineage-specific sex-biased genes greatly determine the distinct sex biases in human and macaque brains. In humans, those encoding proteins contribute directly to immune-related functions, and those encoding lncRNAs intensively regulate the expression of other sex-biased genes, especially genes with immune-related functions. The identified sex-specific differentially expressed genes (ssDEGs) upon gene expression in disease and normal samples also indicate that protein-coding ssDEGs are conserved in humans and macaques but that lncRNA ssDEGs are not conserved. The results answer the above questions, reveal an intrinsic relationship between sex biases in the brain and sex-biased susceptibility to brain diseases, and will help researchers investigate human- and sex-specific ncRNA targets for brain diseases. CONCLUSIONS Human-specific genes greatly cast sex-biased genes in the brain and their relationships with brain diseases, with protein-coding genes contributing to immune response related functions and lncRNA genes critically regulating sex-biased genes. The high proportions of lineage-specific lncRNAs in mammalian genomes indicate that sex biases may have evolved rapidly in not only the brain but also other organs.
Collapse
Affiliation(s)
- Sha He
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xuecong Zhang
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Shenzhen Clinical Research Center for Tuberculosis, National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Hao Zhu
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
12
|
Yang Y, Chen BR, Ye XC, Ni LY, Zhang XY, Liu YZ, Lyu TJ, Tian Y, Fu YJ, Wang Y. The chromodomain protein CDYL confers forebrain identity to human cortical organoids by inhibiting neuronatin. Cell Rep 2024; 43:114814. [PMID: 39378153 DOI: 10.1016/j.celrep.2024.114814] [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: 12/01/2023] [Revised: 08/02/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024] Open
Abstract
Fate determination of neural stem cells (NSCs) is crucial for cortex development and is closely linked to neurodevelopmental disorders when gene expression networks are disrupted. The transcriptional corepressor chromodomain Y-like (CDYL) is widely expressed across diverse cell populations within the human embryonic cortex. However, its precise role in cortical development remains unclear. Here, we show that CDYL is critical for human cortical neurogenesis and that its deficiency leads to a substantial increase in gamma-aminobutyric acid (GABA)-ergic neurons in cortical organoids. Subsequently, neuronatin (NNAT) is identified as a significant target of CDYL, and its abnormal expression obviously influences the fate commitment of cortical NSCs. Cross-species comparisons of CDYL targets unravel a distinct developmental trajectory between human cortical organoids and the mouse cortex at an analogous stage. Collectively, our data provide insight into the evolutionary roles of CDYL in human cortex development, emphasizing its critical function in maintaining the fate of human cortical NSCs.
Collapse
Affiliation(s)
- Yaming Yang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Bai-Rong Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Xi-Chun Ye
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Liang-Yu Ni
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Xi-Yin Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Yun-Ze Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Tian-Jie Lyu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Yue Tian
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Yun-Jie Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
13
|
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
|
14
|
Biagioni M, Baronchelli F, Fossati M. Multiscale spatio-temporal dynamics of UBE3A gene in brain physiology and neurodevelopmental disorders. Neurobiol Dis 2024; 201:106669. [PMID: 39293689 DOI: 10.1016/j.nbd.2024.106669] [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: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024] Open
Abstract
The UBE3A gene, located in the chromosomal region 15q11-13, is subject to neuron-specific genomic imprinting and it plays a critical role in brain development. Genetic defects of UBE3A cause severe neurodevelopmental disorders, namely the Angelman syndrome (AS) and the 15q11.2-q13.3 duplication syndrome (Dup15q). In the last two decades, the development of in vitro and in vivo models of AS and Dup15q were fundamental to improve the understanding of UBE3A function in the brain. However, the pathogenic mechanisms of these diseases remain elusive and effective treatments are lacking. Recent evidence suggests that UBE3A functions are both spatially and temporally specific, varying across subcellular compartments, brain regions, and neuronal circuits. In the present review, we summarize current knowledge on the role of UBE3A in neuronal pathophysiology under this spatio-temporal perspective. Additionally, we propose key research questions that will be instrumental to better understand the pathogenic mechanisms underpinning AS and Dup15q disorders and provide the rationale to develop novel therapies.
Collapse
Affiliation(s)
- Martina Biagioni
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano 20089, MI, Italy
| | - Federica Baronchelli
- CNR - Institute of Neuroscience, Section of Milano, via Manzoni 56, Rozzano 20089, MI, Italy; Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini, 20072 Pieve Emanuele, MI, Italy
| | - Matteo Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano 20089, MI, Italy; CNR - Institute of Neuroscience, Section of Milano, via Manzoni 56, Rozzano 20089, MI, Italy.
| |
Collapse
|
15
|
Caglayan E, Konopka G. Evolutionary neurogenomics at single-cell resolution. Curr Opin Genet Dev 2024; 88:102239. [PMID: 39094380 DOI: 10.1016/j.gde.2024.102239] [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: 05/14/2024] [Revised: 06/20/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
The human brain is composed of increasingly recognized heterogeneous cell types. Applying single-cell genomics to brain tissue can elucidate relative cell type proportions as well as differential gene expression and regulation among humans and other species. Here, we review recent studies that utilized high-throughput genomics approaches to compare brains among species at single-cell resolution. These studies identified genomic elements that are similar among species as well as evolutionary novelties on the human lineage. We focus on those human-relevant innovations and discuss the biological implications of these modifications. Finally, we discuss areas of comparative single-cell genomics that remain unexplored either due to needed technological advances or due to biological availability at the brain region or species level.
Collapse
Affiliation(s)
- Emre Caglayan
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Genevieve Konopka
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
16
|
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
|
17
|
Karageorgiou C, Gokcumen O, Dennis MY. Deciphering the role of structural variation in human evolution: a functional perspective. Curr Opin Genet Dev 2024; 88:102240. [PMID: 39121701 PMCID: PMC11485010 DOI: 10.1016/j.gde.2024.102240] [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: 03/30/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
Advances in sequencing technologies have enabled the comparison of high-quality genomes of diverse primate species, revealing vast amounts of divergence due to structural variation. Given their large size, structural variants (SVs) can simultaneously alter the function and regulation of multiple genes. Studies estimate that collectively more than 3.5% of the genome is divergent in humans versus other great apes, impacting thousands of genes. Functional genomics and gene-editing tools in various model systems recently emerged as an exciting frontier - investigating the wide-ranging impacts of SVs on molecular, cellular, and systems-level phenotypes. This review examines existing research and identifies future directions to broaden our understanding of the functional roles of SVs on phenotypic innovations and diversity impacting uniquely human features, ranging from cognition to metabolic adaptations.
Collapse
Affiliation(s)
- Charikleia Karageorgiou
- Department of Biological Sciences, University at Buffalo, 109 Cooke Hall, Buffalo, NY 14260, USA. https://twitter.com/@evobioclio
| | - Omer Gokcumen
- Department of Biological Sciences, University at Buffalo, 109 Cooke Hall, Buffalo, NY 14260, USA
| | - Megan Y Dennis
- Department of Biochemistry & Molecular Medicine, Genome Center, and MIND Institute, University of California, Davis, CA 95616, USA.
| |
Collapse
|
18
|
Namba T, Huttner WB. What Makes Us Human: Insights from the Evolution and Development of the Human Neocortex. Annu Rev Cell Dev Biol 2024; 40:427-452. [PMID: 39356810 DOI: 10.1146/annurev-cellbio-112122-032521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
"What makes us human?" is a central question of many research fields, notably anthropology. In this review, we focus on the development of the human neocortex, the part of the brain with a key role in cognition, to gain neurobiological insight toward answering this question. We first discuss cortical stem and progenitor cells and human-specific genes that affect their behavior. We thus aim to understand the molecular foundation of the expansion of the neocortex that occurred in the course of human evolution, as this expansion is generally thought to provide a basis for our unique cognitive abilities. We then review the emerging evidence pointing to differences in the development of the neocortex between present-day humans and Neanderthals, our closest relatives. Finally, we discuss human-specific genes that have been implicated in neuronal circuitry and offer a perspective for future studies addressing the question of what makes us human.
Collapse
Affiliation(s)
- Takashi Namba
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany;
| |
Collapse
|
19
|
Vermaercke B, Iwata R, Wierda K, Boubakar L, Rodriguez P, Ditkowska M, Bonin V, Vanderhaeghen P. SYNGAP1 deficiency disrupts synaptic neoteny in xenotransplanted human cortical neurons in vivo. Neuron 2024; 112:3058-3068.e8. [PMID: 39111306 PMCID: PMC11446607 DOI: 10.1016/j.neuron.2024.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/31/2024] [Accepted: 07/10/2024] [Indexed: 09/28/2024]
Abstract
Human brain ontogeny is characterized by a considerably prolonged neotenic development of cortical neurons and circuits. Neoteny is thought to be essential for the acquisition of advanced cognitive functions, which are typically altered in intellectual disability (ID) and autism spectrum disorders (ASDs). Human neuronal neoteny could be disrupted in some forms of ID and/or ASDs, but this has never been tested. Here, we use xenotransplantation of human cortical neurons into the mouse brain to model SYNGAP1 haploinsufficiency, one of the most prevalent genetic causes of ID/ASDs. We find that SYNGAP1-deficient human neurons display strong acceleration of morphological and functional synaptic formation and maturation alongside disrupted synaptic plasticity. At the circuit level, SYNGAP1-haploinsufficient neurons display precocious acquisition of responsiveness to visual stimulation months ahead of time. Our findings indicate that SYNGAP1 is required cell autonomously for human neuronal neoteny, providing novel links between human-specific developmental mechanisms and ID/ASDs.
Collapse
Affiliation(s)
- Ben Vermaercke
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000 Leuven, Belgium
| | - Ryohei Iwata
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000 Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Leïla Boubakar
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000 Leuven, Belgium
| | - Paula Rodriguez
- Neuro-Electronics Research Flanders, Kapeldreef 75, 3001 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; imec, 3001 Leuven, Belgium
| | - Martyna Ditkowska
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000 Leuven, Belgium
| | - Vincent Bonin
- Neuro-Electronics Research Flanders, Kapeldreef 75, 3001 Leuven, Belgium; Department of Biology, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; imec, 3001 Leuven, Belgium.
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences & Leuven Brain Institute, 3000 Leuven, Belgium.
| |
Collapse
|
20
|
Lotter LD, Saberi A, Hansen JY, Misic B, Paquola C, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Paillère ML, Artiges E, Papadopoulos Orfanos D, Paus T, Poustka L, Hohmann S, Fröhner JH, Smolka MN, Vaidya N, Walter H, Whelan R, Schumann G, Nees F, Banaschewski T, Eickhoff SB, Dukart J. Regional patterns of human cortex development correlate with underlying neurobiology. Nat Commun 2024; 15:7987. [PMID: 39284858 PMCID: PMC11405413 DOI: 10.1038/s41467-024-52366-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Human brain morphology undergoes complex changes over the lifespan. Despite recent progress in tracking brain development via normative models, current knowledge of underlying biological mechanisms is highly limited. We demonstrate that human cortical thickness development and aging trajectories unfold along patterns of molecular and cellular brain organization, traceable from population-level to individual developmental trajectories. During childhood and adolescence, cortex-wide spatial distributions of dopaminergic receptors, inhibitory neurons, glial cell populations, and brain-metabolic features explain up to 50% of the variance associated with a lifespan model of regional cortical thickness trajectories. In contrast, modeled cortical thickness change patterns during adulthood are best explained by cholinergic and glutamatergic neurotransmitter receptor and transporter distributions. These relationships are supported by developmental gene expression trajectories and translate to individual longitudinal data from over 8000 adolescents, explaining up to 59% of developmental change at cohort- and 18% at single-subject level. Integrating neurobiological brain atlases with normative modeling and population neuroimaging provides a biologically meaningful path to understand brain development and aging in living humans.
Collapse
Affiliation(s)
- Leon D Lotter
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany.
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
- Max Planck School of Cognition; Stephanstrasse 1A, Leipzig, Germany.
| | - Amin Saberi
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Otto Hahn Research Group for Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Justine Y Hansen
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Bratislav Misic
- McConnell Brain Imaging Centre, Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Casey Paquola
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham; University Park, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB); Braunschweig and Berlin, Berlin, Germany
| | - Jean-Luc Martinot
- Ecole Normale Supérieure Paris-Saclay, Université Paris-Saclay, Université paris Cité, INSERM U1299 "Trajectoires Développementales & Psychiatrie"; Centre Borelli, Gif-sur-Yvette, France
| | - Marie-Laure Paillère
- Ecole Normale Supérieure Paris-Saclay, Université Paris-Saclay, Université paris Cité, INSERM U1299 "Trajectoires Développementales & Psychiatrie"; Centre Borelli, Gif-sur-Yvette, France
- AP-HP Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
| | - Eric Artiges
- Ecole Normale Supérieure Paris-Saclay, Université Paris-Saclay, Université paris Cité, INSERM U1299 "Trajectoires Développementales & Psychiatrie"; Centre Borelli, Gif-sur-Yvette, France
- Department of Psychiatry, EPS Barthélémy Durand, Etampes, France
| | | | - Tomáš Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juliane H Fröhner
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Neuroscience, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Neuroscience, Charité Universitätsmedizin Berlin, Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | - Frauke Nees
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Center for Mental Health (DZPG), partner site Mannheim-Heidelberg-Ulm, Heidelberg, Germany
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Juergen Dukart
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany.
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
21
|
Glausier JR, Bouchet-Marquis C, Maier M, Banks-Tibbs T, Wu K, Ning J, Melchitzky D, Lewis DA, Freyberg Z. Volume electron microscopy reveals 3D synaptic nanoarchitecture in postmortem human prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582174. [PMID: 38463986 PMCID: PMC10925168 DOI: 10.1101/2024.02.26.582174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Synaptic function is directly reflected in quantifiable ultrastructural features using electron microscopy (EM) approaches. This coupling of synaptic function and ultrastructure suggests that in vivo synaptic function can be inferred from EM analysis of ex vivo human brain tissue. To investigate this, we employed focused ion beam-scanning electron microscopy (FIB-SEM), a volume EM (VEM) approach, to generate ultrafine-resolution, three-dimensional (3D) micrographic datasets of postmortem human dorsolateral prefrontal cortex (DLPFC), a region with cytoarchitectonic characteristics distinct to human brain. Synaptic, sub-synaptic, and organelle measures were highly consistent with findings from experimental models that are free from antemortem or postmortem effects. Further, 3D neuropil reconstruction revealed a unique, ultrastructurally-complex, spiny dendritic shaft that exhibited features characteristic of heightened synaptic communication, integration, and plasticity. Altogether, our findings provide critical proof-of-concept data demonstrating that ex vivo VEM analysis is an effective approach to infer in vivo synaptic functioning in human brain.
Collapse
Affiliation(s)
- Jill R. Glausier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | | | - Matthew Maier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Tabitha Banks-Tibbs
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA
- College of Medicine, The Ohio State University, Columbus, OH
| | - Ken Wu
- Materials and Structural Analysis, Thermo Fisher Scientific, Hillsboro, OR
| | - Jiying Ning
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | | | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
22
|
von Mueffling A, Garcia-Forn M, De Rubeis S. DDX3X syndrome: From clinical phenotypes to biological insights. J Neurochem 2024; 168:2147-2154. [PMID: 38976626 PMCID: PMC11449660 DOI: 10.1111/jnc.16174] [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: 05/16/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024]
Abstract
DDX3X syndrome is a neurodevelopmental disorder accounting for up to 3% of cases of intellectual disability (ID) and affecting primarily females. Individuals diagnosed with DDX3X syndrome can also present with behavioral challenges, motor delays and movement disorders, epilepsy, and congenital malformations. DDX3X syndrome is caused by mutations in the X-linked gene DDX3X, which encodes a DEAD-box RNA helicase with critical roles in RNA metabolism, including mRNA translation. Emerging discoveries from animal models are unveiling a fundamental role of DDX3X in neuronal differentiation and development, especially in the neocortex. Here, we review the current knowledge of genetic and neurobiological mechanisms underlying DDX3X syndrome and their relationship with clinical phenotypes.
Collapse
Affiliation(s)
- Alexa von Mueffling
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Barnard College, Columbia University, New York, NY 10027, USA
| | - Marta Garcia-Forn
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
23
|
Pavlinek A, Adhya D, Tsompanidis A, Warrier V, Vernon AC, Lancaster M, Mill J, Srivastava DP, Baron-Cohen S. Using Organoids to Model Sex Differences in the Human Brain. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100343. [PMID: 39092139 PMCID: PMC11292257 DOI: 10.1016/j.bpsgos.2024.100343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 08/04/2024] Open
Abstract
Sex differences are widespread during neurodevelopment and play a role in neuropsychiatric conditions such as autism, which is more prevalent in males than females. In humans, males have been shown to have larger brain volumes than females with development of the hippocampus and amygdala showing prominent sex differences. Mechanistically, sex steroids and sex chromosomes drive these differences in brain development, which seem to peak during prenatal and pubertal stages. Animal models have played a crucial role in understanding sex differences, but the study of human sex differences requires an experimental model that can recapitulate complex genetic traits. To fill this gap, human induced pluripotent stem cell-derived brain organoids are now being used to study how complex genetic traits influence prenatal brain development. For example, brain organoids from individuals with autism and individuals with X chromosome-linked Rett syndrome and fragile X syndrome have revealed prenatal differences in cell proliferation, a measure of brain volume differences, and excitatory-inhibitory imbalances. Brain organoids have also revealed increased neurogenesis of excitatory neurons due to androgens. However, despite growing interest in using brain organoids, several key challenges remain that affect its validity as a model system. In this review, we discuss how sex steroids and the sex chromosomes each contribute to sex differences in brain development. Then, we examine the role of X chromosome inactivation as a factor that drives sex differences. Finally, we discuss the combined challenges of modeling X chromosome inactivation and limitations of brain organoids that need to be taken into consideration when studying sex differences.
Collapse
Affiliation(s)
- Adam Pavlinek
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Dwaipayan Adhya
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Alex Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Varun Warrier
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | | | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
24
|
Al Harrach M, Yochum M, Ruffini G, Bartolomei F, Wendling F, Benquet P. NeoCoMM: A neocortical neuroinspired computational model for the reconstruction and simulation of epileptiform events. Comput Biol Med 2024; 180:108934. [PMID: 39079417 DOI: 10.1016/j.compbiomed.2024.108934] [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: 01/18/2024] [Revised: 06/13/2024] [Accepted: 07/20/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Understanding the pathophysiological dynamics that underline Interictal Epileptiform Events (IEEs) such as epileptic spikes, spike-and-waves or High-Frequency Oscillations (HFOs) is of major importance in the context of neocortical refractory epilepsy, as it paves the way for the development of novel therapies. Typically, these events are detected in Local Field Potential (LFP) recordings obtained through depth electrodes during pre-surgical investigations. Although essential, the underlying pathophysiological mechanisms for the generation of these epileptic neuromarkers remain unclear. The aim of this paper is to propose a novel neurophysiologically relevant reconstruction of the neocortical microcircuitry in the context of epilepsy. This reconstruction intends to facilitate the analysis of a comprehensive set of parameters encompassing physiological, morphological, and biophysical aspects that directly impact the generation and recording of different IEEs. METHOD a novel microscale computational model of an epileptic neocortical column was introduced. This model incorporates the intricate multilayered structure of the cortex and allows for the simulation of realistic interictal epileptic signals. The proposed model was validated through comparisons with real IEEs recorded using intracranial stereo-electroencephalography (SEEG) signals from both humans and animals. Using the model, the user can recreate epileptiform patterns observed in different species (human, rodent, and mouse) and study the intracellular activity associated with these patterns. RESULTS Our model allowed us to unravel the relationship between glutamatergic and GABAergic synaptic transmission of the epileptic neural network and the type of generated IEE. Moreover, sensitivity analyses allowed for the exploration of the pathophysiological parameters responsible for the transitions between these events. Finally, the presented modeling framework also provides an Electrode Tissue Model (ETI) that adds realism to the simulated signals and offers the possibility of studying their sensitivity to the electrode characteristics. CONCLUSION The model (NeoCoMM) presented in this work can be of great use in different applications since it offers an in silico framework for sensitivity analysis and hypothesis testing. It can also be used as a starting point for more complex studies.
Collapse
Affiliation(s)
- M Al Harrach
- University of Rennes, INSERM, LTSI-U1099, 35000 Rennes, France.
| | - M Yochum
- Neuroelectrics, Av. Tibidabo 47b, 08035 Barcelona, Spain
| | - G Ruffini
- Neuroelectrics, Av. Tibidabo 47b, 08035 Barcelona, Spain
| | - F Bartolomei
- Hopitaux de Marseille, Service d'Epileptologie et de Rythmologie Cerebrale, Hopital La Timone, Marseille, France
| | - F Wendling
- University of Rennes, INSERM, LTSI-U1099, 35000 Rennes, France
| | - P Benquet
- University of Rennes, INSERM, LTSI-U1099, 35000 Rennes, France
| |
Collapse
|
25
|
Gavenas J, Rutishauser U, Schurger A, Maoz U. Slow ramping emerges from spontaneous fluctuations in spiking neural networks. Nat Commun 2024; 15:7285. [PMID: 39179554 PMCID: PMC11344096 DOI: 10.1038/s41467-024-51401-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
The capacity to initiate actions endogenously is critical for goal-directed behavior. Spontaneous voluntary actions are typically preceded by slow-ramping activity in medial frontal cortex that begins around two seconds before movement, which may reflect spontaneous fluctuations that influence action timing. However, the mechanisms by which these slow ramping signals emerge from single-neuron and network dynamics remain poorly understood. Here, we developed a spiking neural-network model that produces spontaneous slow ramping activity in single neurons and population activity with onsets ~2 s before threshold crossings. A key prediction of our model is that neurons that ramp together have correlated firing patterns before ramping onset. We confirmed this model-derived hypothesis in a dataset of human single neuron recordings from medial frontal cortex. Our results suggest that slow ramping signals reflect bounded spontaneous fluctuations that emerge from quasi-winner-take-all dynamics in clustered networks that are temporally stabilized by slow-acting synapses.
Collapse
Affiliation(s)
- Jake Gavenas
- Institute for Interdisciplinary Brain and Behavioral Sciences, Chapman University, Orange, CA, USA.
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA.
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Ueli Rutishauser
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Aaron Schurger
- Institute for Interdisciplinary Brain and Behavioral Sciences, Chapman University, Orange, CA, USA
- Crean College of Health and Behavioral Sciences, Chapman University, Orange, CA, USA
- INSERM U992, Cognitive Neuroimaging Unit, NeuroSpin Center, Gif sur Yvette, 91191, France
- Commissariat à l'Energie Atomique, Direction des Sciences du Vivant, I2BM, NeuroSpin Center, Gif sur Yvette, 91191, France
| | - Uri Maoz
- Institute for Interdisciplinary Brain and Behavioral Sciences, Chapman University, Orange, CA, USA.
- Schmid College of Science and Technology, Chapman University, Orange, CA, USA.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Crean College of Health and Behavioral Sciences, Chapman University, Orange, CA, USA.
- Fowler School of Engineering, Chapman University, Orange, CA, USA.
- Anderson School of Management, University of California, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Moriano J, Leonardi O, Vitriolo A, Testa G, Boeckx C. A multi-layered integrative analysis reveals a cholesterol metabolic program in outer radial glia with implications for human brain evolution. Development 2024; 151:dev202390. [PMID: 39114968 PMCID: PMC11385646 DOI: 10.1242/dev.202390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 07/18/2024] [Indexed: 08/28/2024]
Abstract
The definition of molecular and cellular mechanisms contributing to brain ontogenetic trajectories is essential to investigate the evolution of our species. Yet their functional dissection at an appropriate level of granularity remains challenging. Capitalizing on recent efforts that have extensively profiled neural stem cells from the developing human cortex, we develop an integrative computational framework to perform trajectory inference and gene regulatory network reconstruction, (pseudo)time-informed non-negative matrix factorization for learning the dynamics of gene expression programs, and paleogenomic analysis for a higher-resolution mapping of derived regulatory variants in our species in comparison with our closest relatives. We provide evidence for cell type-specific regulation of gene expression programs during indirect neurogenesis. In particular, our analysis uncovers a key role for a cholesterol program in outer radial glia, regulated by zinc-finger transcription factor KLF6. A cartography of the regulatory landscape impacted by Homo sapiens-derived variants reveals signals of selection clustering around regulatory regions associated with GLI3, a well-known regulator of radial glial cell cycle, and impacting KLF6 regulation. Our study contributes to the evidence of significant changes in metabolic pathways in recent human brain evolution.
Collapse
Affiliation(s)
- Juan Moriano
- Department of General Linguistics, University of Barcelona, 08007 Barcelona, Spain
- University of Barcelona Institute of Complex Systems, 08007 Barcelona, Spain
| | | | - Alessandro Vitriolo
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy
| | - Giuseppe Testa
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy
| | - Cedric Boeckx
- Department of General Linguistics, 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
| |
Collapse
|
27
|
Thor S. Indirect neurogenesis in space and time. Nat Rev Neurosci 2024; 25:519-534. [PMID: 38951687 DOI: 10.1038/s41583-024-00833-x] [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: 05/29/2024] [Indexed: 07/03/2024]
Abstract
During central nervous system (CNS) development, neural progenitor cells (NPCs) generate neurons and glia in two different ways. In direct neurogenesis, daughter cells differentiate directly into neurons or glia, whereas in indirect neurogenesis, neurons or glia are generated after one or more daughter cell divisions. Intriguingly, indirect neurogenesis is not stochastically deployed and plays instructive roles during CNS development: increased generation of cells from specific lineages; increased generation of early or late-born cell types within a lineage; and increased cell diversification. Increased indirect neurogenesis might contribute to the anterior CNS expansion evident throughout the Bilateria and help to modify brain-region size without requiring increased NPC numbers or extended neurogenesis. Increased indirect neurogenesis could be an evolutionary driver of the gyrencephalic (that is, folded) cortex that emerged during mammalian evolution and might even have increased during hominid evolution. Thus, selection of indirect versus direct neurogenesis provides a powerful developmental and evolutionary instrument that drives not only the evolution of CNS complexity but also brain expansion and modulation of brain-region size, and thereby the evolution of increasingly advanced cognitive abilities. This Review describes indirect neurogenesis in several model species and humans, and highlights some of the molecular genetic mechanisms that control this important process.
Collapse
Affiliation(s)
- Stefan Thor
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
28
|
Iwata R, Vanderhaeghen P. Metabolic mechanisms of species-specific developmental tempo. Dev Cell 2024; 59:1628-1639. [PMID: 38906137 PMCID: PMC11266843 DOI: 10.1016/j.devcel.2024.05.027] [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: 01/19/2024] [Revised: 04/27/2024] [Accepted: 05/23/2024] [Indexed: 06/23/2024]
Abstract
Development consists of a highly ordered suite of steps and transitions, like choreography. Although these sequences are often evolutionarily conserved, they can display species variations in duration and speed, thereby modifying final organ size or function. Despite their evolutionary significance, the mechanisms underlying species-specific scaling of developmental tempo have remained unclear. Here, we will review recent findings that implicate global cellular mechanisms, particularly intermediary and protein metabolism, as species-specific modifiers of developmental tempo. In various systems, from somitic cell oscillations to neuronal development, metabolic pathways display species differences. These have been linked to mitochondrial metabolism, which can influence the species-specific speed of developmental transitions. Thus, intermediary metabolic pathways regulate developmental tempo together with other global processes, including proteostasis and chromatin remodeling. By linking metabolism and the evolution of developmental trajectories, these findings provide opportunities to decipher how species-specific cellular timing can influence organism fitness.
Collapse
Affiliation(s)
- Ryohei Iwata
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium.
| |
Collapse
|
29
|
Morales-Vicente DA, Tahira AC, Woellner-Santos D, Amaral MS, Berzoti-Coelho MG, Verjovski-Almeida S. The Human Developing Cerebral Cortex Is Characterized by an Elevated De Novo Expression of Long Noncoding RNAs in Excitatory Neurons. Mol Biol Evol 2024; 41:msae123. [PMID: 38913688 PMCID: PMC11221658 DOI: 10.1093/molbev/msae123] [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: 11/23/2023] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024] Open
Abstract
The outstanding human cognitive capacities are computed in the cerebral cortex, a mammalian-specific brain region and the place of massive biological innovation. Long noncoding RNAs have emerged as gene regulatory elements with higher evolutionary turnover than mRNAs. The many long noncoding RNAs identified in neural tissues make them candidates for molecular sources of cerebral cortex evolution and disease. Here, we characterized the genomic and cellular shifts that occurred during the evolution of the long noncoding RNA repertoire expressed in the developing cerebral cortex and explored putative roles for these long noncoding RNAs in the evolution of the human brain. Using transcriptomics and comparative genomics, we comprehensively annotated the cortical transcriptomes of humans, rhesus macaques, mice, and chickens and classified human cortical long noncoding RNAs into evolutionary groups as a function of their predicted minimal ages. Long noncoding RNA evolutionary groups showed differences in expression levels, splicing efficiencies, transposable element contents, genomic distributions, and transcription factor binding to their promoters. Furthermore, older long noncoding RNAs showed preferential expression in germinative zones, outer radial glial cells, and cortical inhibitory (GABAergic) neurons. In comparison, younger long noncoding RNAs showed preferential expression in cortical excitatory (glutamatergic) neurons, were enriched in primate and human-specific gene co-expression modules, and were dysregulated in neurodevelopmental disorders. These results suggest different evolutionary routes for older and younger cortical long noncoding RNAs, highlighting old long noncoding RNAs as a possible source of molecular evolution of conserved developmental programs; conversely, we propose that the de novo expression of primate- and human-specific young long noncoding RNAs is a putative source of molecular evolution and dysfunction of cortical excitatory neurons, warranting further investigation.
Collapse
Affiliation(s)
- David A Morales-Vicente
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Ana C Tahira
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
| | - Daisy Woellner-Santos
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Murilo S Amaral
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
| | - Maria G Berzoti-Coelho
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Verjovski-Almeida
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
30
|
Diaz-Salazar C, Krzisch M, Yoo J, Nano PR, Bhaduri A, Jaenisch R, Polleux F. Human-specific paralogs of SRGAP2 induce neotenic features of microglia structural and functional maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601266. [PMID: 38979266 PMCID: PMC11230448 DOI: 10.1101/2024.06.28.601266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Microglia play key roles in shaping synaptic connectivity during neural circuits development. Whether microglia display human-specific features of structural and functional maturation is currently unknown. We show that the ancestral gene SRGAP2A and its human-specific (HS) paralogs SRGAP2B/C are not only expressed in cortical neurons but are the only HS gene duplications expressed in human microglia. Here, using combination of xenotransplantation of human induced pluripotent stem cell (hiPSC)-derived microglia and mouse genetic models, we demonstrate that (1) HS SRGAP2B/C are necessary and sufficient to induce neotenic features of microglia structural and functional maturation in a cell-autonomous manner, and (2) induction of SRGAP2-dependent neotenic features of microglia maturation non-cell autonomously impacts synaptic development in cortical pyramidal neurons. Our results reveal that, during human brain evolution, human-specific genes SRGAP2B/C coordinated the emergence of neotenic features of synaptic development by acting as genetic modifiers of both neurons and microglia.
Collapse
Affiliation(s)
- Carlos Diaz-Salazar
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University; New York, NY, 10027, USA
| | - Marine Krzisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Juyoun Yoo
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University; New York, NY, 10027, USA
| | - Patricia R. Nano
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University; New York, NY, 10027, USA
| |
Collapse
|
31
|
Lindhout FW, Krienen FM, Pollard KS, Lancaster MA. A molecular and cellular perspective on human brain evolution and tempo. Nature 2024; 630:596-608. [PMID: 38898293 DOI: 10.1038/s41586-024-07521-x] [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: 06/29/2021] [Accepted: 04/29/2024] [Indexed: 06/21/2024]
Abstract
The evolution of the modern human brain was accompanied by distinct molecular and cellular specializations, which underpin our diverse cognitive abilities but also increase our susceptibility to neurological diseases. These features, some specific to humans and others shared with related species, manifest during different stages of brain development. In this multi-stage process, neural stem cells proliferate to produce a large and diverse progenitor pool, giving rise to excitatory or inhibitory neurons that integrate into circuits during further maturation. This process unfolds over varying time scales across species and has progressively become slower in the human lineage, with differences in tempo correlating with differences in brain size, cell number and diversity, and connectivity. Here we introduce the terms 'bradychrony' and 'tachycrony' to describe slowed and accelerated developmental tempos, respectively. We review how recent technical advances across disciplines, including advanced engineering of in vitro models, functional comparative genetics and high-throughput single-cell profiling, are leading to a deeper understanding of how specializations of the human brain arise during bradychronic neurodevelopment. Emerging insights point to a central role for genetics, gene-regulatory networks, cellular innovations and developmental tempo, which together contribute to the establishment of human specializations during various stages of neurodevelopment and at different points in evolution.
Collapse
Affiliation(s)
- Feline W Lindhout
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK.
| | - Fenna M Krienen
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Katherine S Pollard
- Gladstone Institutes, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, Institute for Computational Health Sciences, and Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
32
|
Lee AT, Chang EF, Paredes MF, Nowakowski TJ. Large-scale neurophysiology and single-cell profiling in human neuroscience. Nature 2024; 630:587-595. [PMID: 38898291 DOI: 10.1038/s41586-024-07405-0] [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: 07/06/2023] [Accepted: 04/09/2024] [Indexed: 06/21/2024]
Abstract
Advances in large-scale single-unit human neurophysiology, single-cell RNA sequencing, spatial transcriptomics and long-term ex vivo tissue culture of surgically resected human brain tissue have provided an unprecedented opportunity to study human neuroscience. In this Perspective, we describe the development of these paradigms, including Neuropixels and recent brain-cell atlas efforts, and discuss how their convergence will further investigations into the cellular underpinnings of network-level activity in the human brain. Specifically, we introduce a workflow in which functionally mapped samples of human brain tissue resected during awake brain surgery can be cultured ex vivo for multi-modal cellular and functional profiling. We then explore how advances in human neuroscience will affect clinical practice, and conclude by discussing societal and ethical implications to consider. Potential findings from the field of human neuroscience will be vast, ranging from insights into human neurodiversity and evolution to providing cell-type-specific access to study and manipulate diseased circuits in pathology. This Perspective aims to provide a unifying framework for the field of human neuroscience as we welcome an exciting era for understanding the functional cytoarchitecture of the human brain.
Collapse
Affiliation(s)
- Anthony T Lee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mercedes F Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
33
|
Casimir P, Iwata R, Vanderhaeghen P. Linking mitochondria metabolism, developmental timing, and human brain evolution. Curr Opin Genet Dev 2024; 86:102182. [PMID: 38555796 PMCID: PMC11190843 DOI: 10.1016/j.gde.2024.102182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/02/2024]
Abstract
Changes in developmental timing are an important factor of evolution in organ shape and function. This is particularly striking for human brain development, which, compared with other mammals, is considerably prolonged at the level of the cerebral cortex, resulting in brain neoteny. Here, we review recent findings that indicate that mitochondria and metabolism contribute to species differences in the tempo of cortical neuron development. Mitochondria display species-specific developmental timeline and metabolic activity patterns that are highly correlated with the speed of neuron maturation. Enhancing mitochondrial activity in human cortical neurons results in their accelerated maturation, while its reduction leads to decreased maturation rates in mouse neurons. Together with other global and gene-specific mechanisms, mitochondria thus act as a cellular hourglass of neuronal developmental tempo and may thereby contribute to species-specific features of human brain ontogeny.
Collapse
Affiliation(s)
- Pierre Casimir
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium; Department of Neurology, Centre Hospitalier Universitaire Brugmann, ULB, 1020 Brussels, Belgium
| | - Ryohei Iwata
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium. https://twitter.com/@Ryo2Iwata
| | - Pierre Vanderhaeghen
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
34
|
Plaitakis A, Sidiropoulou K, Kotzamani D, Litso I, Zaganas I, Spanaki C. Evolution of Glutamate Metabolism via GLUD2 Enhances Lactate-Dependent Synaptic Plasticity and Complex Cognition. Int J Mol Sci 2024; 25:5297. [PMID: 38791334 PMCID: PMC11120665 DOI: 10.3390/ijms25105297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Human evolution is characterized by rapid brain enlargement and the emergence of unique cognitive abilities. Besides its distinctive cytoarchitectural organization and extensive inter-neuronal connectivity, the human brain is also defined by high rates of synaptic, mainly glutamatergic, transmission, and energy utilization. While these adaptations' origins remain elusive, evolutionary changes occurred in synaptic glutamate metabolism in the common ancestor of humans and apes via the emergence of GLUD2, a gene encoding the human glutamate dehydrogenase 2 (hGDH2) isoenzyme. Driven by positive selection, hGDH2 became adapted to function upon intense excitatory firing, a process central to the long-term strengthening of synaptic connections. It also gained expression in brain astrocytes and cortical pyramidal neurons, including the CA1-CA3 hippocampal cells, neurons crucial to cognition. In mice transgenic for GLUD2, theta-burst-evoked long-term potentiation (LTP) is markedly enhanced in hippocampal CA3-CA1 synapses, with patch-clamp recordings from CA1 pyramidal neurons revealing increased sNMDA receptor currents. D-lactate blocked LTP enhancement, implying that glutamate metabolism via hGDH2 potentiates L-lactate-dependent glia-neuron interaction, a process essential to memory consolidation. The transgenic (Tg) mice exhibited increased dendritic spine density/synaptogenesis in the hippocampus and improved complex cognitive functions. Hence, enhancement of neuron-glia communication, via GLUD2 evolution, likely contributed to human cognitive advancement by potentiating synaptic plasticity and inter-neuronal connectivity.
Collapse
Affiliation(s)
- Andreas Plaitakis
- Department of Neurology, School of Health Sciences, Faculty of Medicine, University of Crete, Voutes, 71003 Heraklion, Crete, Greece; (D.K.); (I.L.); (I.Z.)
| | - Kyriaki Sidiropoulou
- Department of Biology, University of Crete, Voutes, 71003 Heraklion, Crete, Greece;
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), 70013 Heraklion, Crete, Greece
| | - Dimitra Kotzamani
- Department of Neurology, School of Health Sciences, Faculty of Medicine, University of Crete, Voutes, 71003 Heraklion, Crete, Greece; (D.K.); (I.L.); (I.Z.)
| | - Ionela Litso
- Department of Neurology, School of Health Sciences, Faculty of Medicine, University of Crete, Voutes, 71003 Heraklion, Crete, Greece; (D.K.); (I.L.); (I.Z.)
| | - Ioannis Zaganas
- Department of Neurology, School of Health Sciences, Faculty of Medicine, University of Crete, Voutes, 71003 Heraklion, Crete, Greece; (D.K.); (I.L.); (I.Z.)
- Neurology Department, PaGNI University General Hospital of Heraklion, 71500 Heraklion, Crete, Greece
| | - Cleanthe Spanaki
- Department of Neurology, School of Health Sciences, Faculty of Medicine, University of Crete, Voutes, 71003 Heraklion, Crete, Greece; (D.K.); (I.L.); (I.Z.)
- Neurology Department, PaGNI University General Hospital of Heraklion, 71500 Heraklion, Crete, Greece
| |
Collapse
|
35
|
Chandel AS, Keseroglu K, Özbudak EM. Oscillatory control of embryonic development. Development 2024; 151:dev202191. [PMID: 38727565 PMCID: PMC11128281 DOI: 10.1242/dev.202191] [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: 05/28/2024]
Abstract
Proper embryonic development depends on the timely progression of a genetic program. One of the key mechanisms for achieving precise control of developmental timing is to use gene expression oscillations. In this Review, we examine how gene expression oscillations encode temporal information during vertebrate embryonic development by discussing the gene expression oscillations occurring during somitogenesis, neurogenesis, myogenesis and pancreas development. These oscillations play important but varied physiological functions in different contexts. Oscillations control the period of somite formation during somitogenesis, whereas they regulate the proliferation-to-differentiation switch of stem cells and progenitor cells during neurogenesis, myogenesis and pancreas development. We describe the similarities and differences of the expression pattern in space (i.e. whether oscillations are synchronous or asynchronous across neighboring cells) and in time (i.e. different time scales) of mammalian Hes/zebrafish Her genes and their targets in different tissues. We further summarize experimental evidence for the functional role of their oscillations. Finally, we discuss the outstanding questions for future research.
Collapse
Affiliation(s)
- Angad Singh Chandel
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Systems Biology and Physiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kemal Keseroglu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ertuğrul M. Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
36
|
Fernández V, Borrell V. Keep calm and make neurons: The effects of glucocorticoids on human cortical neurogenesis. Neuron 2024; 112:1373-1375. [PMID: 38697018 DOI: 10.1016/j.neuron.2024.04.004] [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: 04/01/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024]
Abstract
Maternal well-being is important for the development of the fetus, with a key influence on its nervous system. In this issue of Neuron, Krontira et al.1 implicate glucocorticoids, the stress hormones, in the regulation of neural stem cell identity and proliferation, with long-lasting consequences on brain architecture and educational attainment.
Collapse
Affiliation(s)
- Virginia Fernández
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain.
| |
Collapse
|
37
|
Heredia Cedillo A, Lambert D, Morsella E. Identifying Consciousness in Other Creatures: Three Initial Steps. Behav Sci (Basel) 2024; 14:337. [PMID: 38667133 PMCID: PMC11047643 DOI: 10.3390/bs14040337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/15/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
Identifying consciousness in other creatures, be they animals or exotic creatures that have yet to be discovered, remains a great scientific challenge. We delineate the first three steps that we think are necessary for identifying consciousness in other creatures. Step 1 is to define the particular kind of consciousness in which one is interested. Step 2 is to identify, in humans, the key differences between the brain processes that are associated with consciousness and the brain processes that are not associated with consciousness. For Step 2, to identify these differences, we focus on passive frame theory. Step 3 concerns how the insights derived from consciousness research on humans (e.g., concerning these differences) can be generalized to other creatures. We discuss the significance of examining how consciousness was fashioned by the process of evolution, a process that could be happenstance and replete with incessant tinkering, yielding adaptations that can be suboptimal and counterintuitive, far different in nature from our efficiently designed robotic systems. We conclude that the more that is understood about the differences between conscious processing and unconscious processing in humans, the easier it will be to identify consciousness in other creatures.
Collapse
Affiliation(s)
- Alejandro Heredia Cedillo
- Department of Psychology, San Francisco State University, San Francisco, CA 94132, USA; (D.L.); (E.M.)
| | - Dennis Lambert
- Department of Psychology, San Francisco State University, San Francisco, CA 94132, USA; (D.L.); (E.M.)
| | - Ezequiel Morsella
- Department of Psychology, San Francisco State University, San Francisco, CA 94132, USA; (D.L.); (E.M.)
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
38
|
Liu J, Mosti F, Zhao HT, Sotelo-Fonseca JE, Escobar-Tomlienovich CF, Lollis D, Musso CM, Mao Y, Massri AJ, Doll HM, Sousa AM, Wray GA, Schmidt E, Silver DL. A human-specific enhancer fine-tunes radial glia potency and corticogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588953. [PMID: 38645099 PMCID: PMC11030412 DOI: 10.1101/2024.04.10.588953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Humans evolved an extraordinarily expanded and complex cerebral cortex, associated with developmental and gene regulatory modifications 1-3 . Human accelerated regions (HARs) are highly conserved genomic sequences with human-specific nucleotide substitutions. Although there are thousands of annotated HARs, their functional contribution to human-specific cortical development is largely unknown 4,5 . HARE5 is a HAR transcriptional enhancer of the WNT signaling receptor Frizzled8 (FZD8) active during brain development 6 . Here, using genome-edited mouse and primate models, we demonstrate that human (Hs) HARE5 fine-tunes cortical development and connectivity by controlling the proliferative and neurogenic capacity of neural progenitor cells (NPCs). Hs-HARE5 knock-in mice have significantly enlarged neocortices containing more neurons. By measuring neural dynamics in vivo we show these anatomical features correlate with increased functional independence between cortical regions. To understand the underlying developmental mechanisms, we assess progenitor fate using live imaging, lineage analysis, and single-cell RNA sequencing. This reveals Hs-HARE5 modifies radial glial progenitor behavior, with increased self-renewal at early developmental stages followed by expanded neurogenic potential. We use genome-edited human and chimpanzee (Pt) NPCs and cortical organoids to assess the relative enhancer activity and function of Hs-HARE5 and Pt-HARE5. Using these orthogonal strategies we show four human-specific variants in HARE5 drive increased enhancer activity which promotes progenitor proliferation. These findings illustrate how small changes in regulatory DNA can directly impact critical signaling pathways and brain development. Our study uncovers new functions for HARs as key regulatory elements crucial for the expansion and complexity of the human cerebral cortex.
Collapse
|
39
|
Heesen SH, Köhr G. GABAergic interneuron diversity and organization are crucial for the generation of human-specific functional neural networks in cerebral organoids. Front Cell Neurosci 2024; 18:1389335. [PMID: 38665372 PMCID: PMC11044699 DOI: 10.3389/fncel.2024.1389335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
This mini review investigates the importance of GABAergic interneurons for the network function of human-induced pluripotent stem cells (hiPSC)-derived brain organoids. The presented evidence suggests that the abundance, diversity and three-dimensional cortical organization of GABAergic interneurons are the primary elements responsible for the creation of synchronous neuronal firing patterns. Without intricate inhibition, coupled oscillatory patterns cannot reach a sufficient complexity to transfer spatiotemporal information constituting physiological network function. Furthermore, human-specific brain network function seems to be mediated by a more complex and interconnected inhibitory structure that remains developmentally flexible for a longer period when compared to rodents. This suggests that several characteristics of human brain networks cannot be captured by rodent models, emphasizing the need for model systems like organoids that adequately mimic physiological human brain function in vitro.
Collapse
Affiliation(s)
- Sebastian H. Heesen
- Molecular and Behavioural Neurobiology, Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Georg Köhr
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany
- Physiology of Neural Networks, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| |
Collapse
|
40
|
Gavenas J, Rutishauser U, Schurger A, Maoz U. Slow ramping emerges from spontaneous fluctuations in spiking neural networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.27.542589. [PMID: 37398452 PMCID: PMC10312459 DOI: 10.1101/2023.05.27.542589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
1. We reveal a mechanism for slow-ramping signals before spontaneous voluntary movements. 2. Slow synapses stabilize spontaneous fluctuations in spiking neural network. 3. We validate model predictions in human frontal cortical single-neuron recordings. 4. The model recreates the readiness potential in an EEG proxy signal. 5. Neurons that ramp together had correlated activity before ramping onset. The capacity to initiate actions endogenously is critical for goal-directed behavior. Spontaneous voluntary actions are typically preceded by slow-ramping activity in medial frontal cortex that begins around two seconds before movement, which may reflect spontaneous fluctuations that influence action timing. However, the mechanisms by which these slow ramping signals emerge from single-neuron and network dynamics remain poorly understood. Here, we developed a spiking neural-network model that produces spontaneous slow ramping activity in single neurons and population activity with onsets ∼2 seconds before threshold crossings. A key prediction of our model is that neurons that ramp together have correlated firing patterns before ramping onset. We confirmed this model-derived hypothesis in a dataset of human single neuron recordings from medial frontal cortex. Our results suggest that slow ramping signals reflect bounded spontaneous fluctuations that emerge from quasi-winner-take-all dynamics in clustered networks that are temporally stabilized by slow-acting synapses.
Collapse
|
41
|
Ziak J, Dorskind JM, Trigg B, Sudarsanam S, Jin XO, Hand RA, Kolodkin AL. Microtubule-binding protein MAP1B regulates interstitial axon branching of cortical neurons via the tubulin tyrosination cycle. EMBO J 2024; 43:1214-1243. [PMID: 38388748 PMCID: PMC10987652 DOI: 10.1038/s44318-024-00050-3] [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: 08/11/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Regulation of directed axon guidance and branching during development is essential for the generation of neuronal networks. However, the molecular mechanisms that underlie interstitial (or collateral) axon branching in the mammalian brain remain unresolved. Here, we investigate interstitial axon branching in vivo using an approach for precise labeling of layer 2/3 callosal projection neurons (CPNs). This method allows for quantitative analysis of axonal morphology at high acuity and also manipulation of gene expression in well-defined temporal windows. We find that the GSK3β serine/threonine kinase promotes interstitial axon branching in layer 2/3 CPNs by releasing MAP1B-mediated inhibition of axon branching. Further, we find that the tubulin tyrosination cycle is a key downstream component of GSK3β/MAP1B signaling. These data suggest a cell-autonomous molecular regulation of cortical neuron axon morphology, in which GSK3β can release a MAP1B-mediated brake on interstitial axon branching upstream of the posttranslational tubulin code.
Collapse
Affiliation(s)
- Jakub Ziak
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Joelle M Dorskind
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
- Novartis Institutes for BioMedical Research, Boston, MA, USA
| | - Brian Trigg
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Sriram Sudarsanam
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Xinyu O Jin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
| | - Randal A Hand
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA
- Prilenia Therapeutics, Boston, MA, USA
| | - Alex L Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins Kavli Neuroscience Discovery Institute, The Johns Hopkins School of Medicine, 725 North Wolfe St., Baltimore, MD, 21205, USA.
| |
Collapse
|
42
|
Pigeon J, Hassan BA. Timing neurogenesis: a clock or an algorithm? Curr Opin Genet Dev 2024; 85:102156. [PMID: 38354530 DOI: 10.1016/j.gde.2024.102156] [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: 11/01/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/16/2024]
Abstract
Emerging evidence supports the existence of dedicated molecular mechanisms under evolutionary selection to control time during neurogenesis. Here, we briefly review these mechanisms and discuss a potentially useful conceptual framework inspired by computer science to think about how these biological mechanisms operate during brain development and evolution.
Collapse
Affiliation(s)
- Julien Pigeon
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| | - Bassem A Hassan
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
43
|
Dehay C, Huttner WB. Development and evolution of the primate neocortex from a progenitor cell perspective. Development 2024; 151:dev199797. [PMID: 38369736 DOI: 10.1242/dev.199797] [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: 02/20/2024]
Abstract
The generation of neurons in the developing neocortex is a major determinant of neocortex size. Crucially, the increase in cortical neuron numbers in the primate lineage, notably in the upper-layer neurons, contributes to increased cognitive abilities. Here, we review major evolutionary changes affecting the apical progenitors in the ventricular zone and focus on the key germinal zone constituting the foundation of neocortical neurogenesis in primates, the outer subventricular zone (OSVZ). We summarize characteristic features of the OSVZ and its key stem cell type, the basal (or outer) radial glia. Next, we concentrate on primate-specific and human-specific genes, expressed in OSVZ-progenitors, the ability of which to amplify these progenitors by targeting the regulation of the cell cycle ultimately underlies the evolutionary increase in upper-layer neurons. Finally, we address likely differences in neocortical development between present-day humans and Neanderthals that are based on human-specific amino acid substitutions in proteins operating in cortical progenitors.
Collapse
Affiliation(s)
- Colette Dehay
- Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, F-69500 Bron, France
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| |
Collapse
|
44
|
Wang B, Starr AL, Fraser HB. Cell-type-specific cis-regulatory divergence in gene expression and chromatin accessibility revealed by human-chimpanzee hybrid cells. eLife 2024; 12:RP89594. [PMID: 38358392 PMCID: PMC10942608 DOI: 10.7554/elife.89594] [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: 02/16/2024] Open
Abstract
Although gene expression divergence has long been postulated to be the primary driver of human evolution, identifying the genes and genetic variants underlying uniquely human traits has proven to be quite challenging. Theory suggests that cell-type-specific cis-regulatory variants may fuel evolutionary adaptation due to the specificity of their effects. These variants can precisely tune the expression of a single gene in a single cell-type, avoiding the potentially deleterious consequences of trans-acting changes and non-cell type-specific changes that can impact many genes and cell types, respectively. It has recently become possible to quantify human-specific cis-acting regulatory divergence by measuring allele-specific expression in human-chimpanzee hybrid cells-the product of fusing induced pluripotent stem (iPS) cells of each species in vitro. However, these cis-regulatory changes have only been explored in a limited number of cell types. Here, we quantify human-chimpanzee cis-regulatory divergence in gene expression and chromatin accessibility across six cell types, enabling the identification of highly cell-type-specific cis-regulatory changes. We find that cell-type-specific genes and regulatory elements evolve faster than those shared across cell types, suggesting an important role for genes with cell-type-specific expression in human evolution. Furthermore, we identify several instances of lineage-specific natural selection that may have played key roles in specific cell types, such as coordinated changes in the cis-regulation of dozens of genes involved in neuronal firing in motor neurons. Finally, using novel metrics and a machine learning model, we identify genetic variants that likely alter chromatin accessibility and transcription factor binding, leading to neuron-specific changes in the expression of the neurodevelopmentally important genes FABP7 and GAD1. Overall, our results demonstrate that integrative analysis of cis-regulatory divergence in chromatin accessibility and gene expression across cell types is a promising approach to identify the specific genes and genetic variants that make us human.
Collapse
Affiliation(s)
- Ban Wang
- Department of Biology, Stanford UniversityStanfordUnited States
| | | | - Hunter B Fraser
- Department of Biology, Stanford UniversityStanfordUnited States
| |
Collapse
|
45
|
Abstract
Brain development in humans is achieved through precise spatiotemporal genetic control, the mechanisms of which remain largely elusive. Recently, integration of technological advances in human stem cell-based modelling with genome editing has emerged as a powerful platform to establish causative links between genotypes and phenotypes directly in the human system. Here, we review our current knowledge of complex genetic regulation of each key step of human brain development through the lens of evolutionary specialization and neurodevelopmental disorders and highlight the use of human stem cell-derived 2D cultures and 3D brain organoids to investigate human-enriched features and disease mechanisms. We also discuss opportunities and challenges of integrating new technologies to reveal the genetic architecture of human brain development and disorders.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Usui N. Possible roles of deep cortical neurons and oligodendrocytes in the neural basis of human sociality. Anat Sci Int 2024; 99:34-47. [PMID: 38010534 PMCID: PMC10771383 DOI: 10.1007/s12565-023-00747-1] [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: 05/22/2023] [Accepted: 10/17/2023] [Indexed: 11/29/2023]
Abstract
Sociality is an instinctive property of organisms that live in relation to others and is a complex characteristic of higher order brain functions. However, the evolution of the human brain to acquire higher order brain functions, such as sociality, and the neural basis for executing these functions and their control mechanisms are largely unknown. Several studies have attempted to evaluate how human sociality was acquired during the course of evolution and the mechanisms controlling sociality from a neurodevelopment viewpoint. This review discusses these findings in the context of human brain evolution and the pathophysiology of autism spectrum disorder (ASD). Comparative genomic studies of postmortem primate brains have demonstrated human-specific regulatory mechanisms underlying higher order brain functions, providing evidence for the contribution of oligodendrocytes to human brain function. Functional analyses of the causative genes of ASD in animal models have demonstrated that the neural basis of social behavior is associated with layer 6 (L6) of the neocortex and oligodendrocytes. These findings demonstrate that both neurons and oligodendrocytes contribute to the neural basis and molecular mechanisms underlying human brain evolution and social functioning. This review provides novel insights into sociability and the corresponding neural bases of brain disorders and evolution.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan.
- Omics Center, Center of Medical Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan.
- United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan.
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, 565-0871, Japan.
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, 541-8567, Japan.
| |
Collapse
|
47
|
Nomura T. In Vitro Patch-Clamp. Methods Mol Biol 2024; 2794:221-244. [PMID: 38630233 DOI: 10.1007/978-1-0716-3810-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
The patch-clamp technique is one of the most useful tools to analyze the function of electrically active cells such as neurons. This technique allows for the analysis of proteins (ion channels and receptors), cells (neurons), and synapses that are the building blocks of neuronal networks. Cortical development involves coordinated changes in functional measures at each of these levels of analysis that reflect both cellular and circuit maturation. This chapter explains the technical and theoretical basis of patch-clamp methodology and introduces several examples of how this technique can be applied in the context of cortical development.
Collapse
Affiliation(s)
- Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
48
|
Libé-Philippot B, Lejeune A, Wierda K, Louros N, Erkol E, Vlaeminck I, Beckers S, Gaspariunaite V, Bilheu A, Konstantoulea K, Nyitrai H, De Vleeschouwer M, Vennekens KM, Vidal N, Bird TW, Soto DC, Jaspers T, Dewilde M, Dennis MY, Rousseau F, Comoletti D, Schymkowitz J, Theys T, de Wit J, Vanderhaeghen P. LRRC37B is a human modifier of voltage-gated sodium channels and axon excitability in cortical neurons. Cell 2023; 186:5766-5783.e25. [PMID: 38134874 PMCID: PMC10754148 DOI: 10.1016/j.cell.2023.11.028] [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: 12/02/2022] [Revised: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
The enhanced cognitive abilities characterizing the human species result from specialized features of neurons and circuits. Here, we report that the hominid-specific gene LRRC37B encodes a receptor expressed in human cortical pyramidal neurons (CPNs) and selectively localized to the axon initial segment (AIS), the subcellular compartment triggering action potentials. Ectopic expression of LRRC37B in mouse CPNs in vivo leads to reduced intrinsic excitability, a distinctive feature of some classes of human CPNs. Molecularly, LRRC37B binds to the secreted ligand FGF13A and to the voltage-gated sodium channel (Nav) β-subunit SCN1B. LRRC37B concentrates inhibitory effects of FGF13A on Nav channel function, thereby reducing excitability, specifically at the AIS level. Electrophysiological recordings in adult human cortical slices reveal lower neuronal excitability in human CPNs expressing LRRC37B. LRRC37B thus acts as a species-specific modifier of human neuron excitability, linking human genome and cell evolution, with important implications for human brain function and diseases.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Amélie Lejeune
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Nikolaos Louros
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Emir Erkol
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Ine Vlaeminck
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Sofie Beckers
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Vaiva Gaspariunaite
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Angéline Bilheu
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium
| | - Katerina Konstantoulea
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Hajnalka Nyitrai
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Matthias De Vleeschouwer
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Kristel M Vennekens
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Niels Vidal
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Thomas W Bird
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Daniela C Soto
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Tom Jaspers
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Maarten Dewilde
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Megan Y Dennis
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Frederic Rousseau
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Davide Comoletti
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Joost Schymkowitz
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Tom Theys
- KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Research Group Experimental Neurosurgery and Neuroanatomy, KUL, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium.
| |
Collapse
|
49
|
Mirdass C, Catala M, Bocel M, Nedelec S, Ribes V. Stem cell-derived models of spinal neurulation. Emerg Top Life Sci 2023; 7:423-437. [PMID: 38087891 DOI: 10.1042/etls20230087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Neurulation is a critical step in early embryonic development, giving rise to the neural tube, the primordium of the central nervous system in amniotes. Understanding this complex, multi-scale, multi-tissue morphogenetic process is essential to provide insights into normal development and the etiology of neural tube defects. Innovations in tissue engineering have fostered the generation of pluripotent stem cell-based in vitro models, including organoids, that are emerging as unique tools for delving into neurulation mechanisms, especially in the context of human development. Each model captures specific aspects of neural tube morphogenesis, from epithelialization to neural tissue elongation, folding and cavitation. In particular, the recent models of human and mouse trunk morphogenesis, such as gastruloids, that form a spinal neural plate-like or neural tube-like structure are opening new avenues to study normal and pathological neurulation. Here, we review the morphogenetic events generating the neural tube in the mammalian embryo and questions that remain unanswered. We discuss the advantages and limitations of existing in vitro models of neurulation and possible future technical developments.
Collapse
Affiliation(s)
- Camil Mirdass
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Institut du Fer à Moulin, 75005 Paris, France
- Inserm, UMR-S 1270, 75005 Paris, France
- Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Martin Catala
- Institut de Biologie Paris Seine (IBPS) - Developmental Biology Laboratory, UMR7622 CNRS, INSERM ERL 1156, Sorbonne Université, 9 Quai Saint-Bernard, 75005 Paris, France
| | - Mikaëlle Bocel
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Stéphane Nedelec
- Institut du Fer à Moulin, 75005 Paris, France
- Inserm, UMR-S 1270, 75005 Paris, France
- Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Vanessa Ribes
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| |
Collapse
|
50
|
Akula SK, Exposito-Alonso D, Walsh CA. Shaping the brain: The emergence of cortical structure and folding. Dev Cell 2023; 58:2836-2849. [PMID: 38113850 PMCID: PMC10793202 DOI: 10.1016/j.devcel.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/08/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The cerebral cortex-the brain's covering and largest region-has increased in size and complexity in humans and supports higher cognitive functions such as language and abstract thinking. There is a growing understanding of the human cerebral cortex, including the diversity and number of cell types that it contains, as well as of the developmental mechanisms that shape cortical structure and organization. In this review, we discuss recent progress in our understanding of molecular and cellular processes, as well as mechanical forces, that regulate the folding of the cerebral cortex. Advances in human genetics, coupled with experimental modeling in gyrencephalic species, have provided insights into the central role of cortical progenitors in the gyrification and evolutionary expansion of the cerebral cortex. These studies are essential for understanding the emergence of structural and functional organization during cortical development and the pathogenesis of neurodevelopmental disorders associated with cortical malformations.
Collapse
Affiliation(s)
- Shyam K Akula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - David Exposito-Alonso
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA.
| |
Collapse
|