51
|
Van der Veeken L, Russo FM, Bleeser T, Basurto D, Emam D, Regin Y, Gsell W, Himmelreich U, De Catte L, Rex S, Deprest J. Brain development is altered in rabbit fetuses with congenital diaphragmatic hernia. Prenat Diagn 2023; 43:359-369. [PMID: 36627261 DOI: 10.1002/pd.6309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/28/2022] [Accepted: 01/07/2023] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Children with congenital diaphragmatic hernia (CDH) are at risk for neurodevelopmental delay. Some changes are already present prenatally. Herein, we further examined how the brain develops in fetal rabbits with surgically created DH. METHODS Two fetuses underwent surgical DH creation on day 23 (term = d31). DH pups and littermate controls were harvested at term. Ten DH pups and 11 controls underwent transcardial perfusion for brain fixation and measurement of brain volume, brain folding, neuron and synaptic density, pre-oligodendrocyte count, proliferation, and vascularization. Twelve other DH and 11 controls had echocardiographic assessment of cardiac output and aortic and cerebral blood flow, magnetic resonance imaging (9.4 T) for cerebral volumetry, and molecular assessment of vascularization markers. RESULTS DH pups had lower lung-to-body weight ratio (1.3 ± 0.3 vs. 2.4 ± 0.3%; p < 0.0001) and lower heart-to-body weight ratio (0.007 ± 0.001 vs. 0.009 ± 0.001; p = 0.0006) but comparable body weight and brain-to-body weight ratio. DH pups had a lower left ventricular ejection fraction, aortic and cerebral blood flow (39 ± 8 vs. 54 ± 15 mm/beat; p = 0.03) as compared to controls but similar left cardiac ventricular morphology. Fetal DH-brains were similar in volume but the cerebellum was less folded (perimeter/surface area: 25.5 ± 1.5 vs. 26.8 ± 1.2; p = 0.049). Furthermore, DH brains had a thinner cortex (143 ± 9 vs. 156 ± 13 μm; p = 0.02). Neuron densities in the white matter were higher in DH fetuses (124 ± 18 vs. 104 ± 14; p = 0.01) with comparable proliferation rates. Pre-oligodendrocyte count was lower, coinciding with the lower endothelial cell count. CONCLUSION Rabbits with DH had altered brain development compared to controls prenatally, indicating that brain development is already altered prenatally in CDH.
Collapse
Affiliation(s)
- Lennart Van der Veeken
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Leuven, Belgium.,Clinical Department Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium.,Clinical Department Obstetrics and Gynaecology, University Hospital Antwerp, Antwerpen, Belgium
| | - Francesca Maria Russo
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Leuven, Belgium.,Clinical Department Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Tom Bleeser
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium.,Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - David Basurto
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Doaa Emam
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Leuven, Belgium.,Department Obstetrics and Gynaecology, University Hospitals Tanta, Tanta, Egypt
| | - Yannick Regin
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Luc De Catte
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Leuven, Belgium.,Clinical Department Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Steffen Rex
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium.,Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Jan Deprest
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Leuven, Belgium.,Clinical Department Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium.,Institute for Women's Health, University College London, London, UK
| |
Collapse
|
52
|
Qi J, Mo F, An NA, Mi T, Wang J, Qi J, Li X, Zhang B, Xia L, Lu Y, Sun G, Wang X, Li C, Hu B. A Human-Specific De Novo Gene Promotes Cortical Expansion and Folding. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204140. [PMID: 36638273 PMCID: PMC9982566 DOI: 10.1002/advs.202204140] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Newly originated de novo genes have been linked to the formation and function of the human brain. However, how a specific gene originates from ancestral noncoding DNAs and becomes involved in the preexisting network for functional outcomes remains elusive. Here, a human-specific de novo gene, SP0535, is identified that is preferentially expressed in the ventricular zone of the human fetal brain and plays an important role in cortical development and function. In human embryonic stem cell-derived cortical organoids, knockout of SP0535 compromises their growth and neurogenesis. In SP0535 transgenic (TG) mice, expression of SP0535 induces fetal cortex expansion and sulci and gyri-like structure formation. The progenitors and neurons in the SP0535 TG mouse cortex tend to proliferate and differentiate in ways that are unique to humans. SP0535 TG adult mice also exhibit improved cognitive ability and working memory. Mechanistically, SP0535 interacts with the membrane protein Na+ /K+ ATPase subunit alpha-1 (ATP1A1) and releases Src from the ATP1A1-Src complex, allowing increased level of Src phosphorylation that promotes cell proliferation. Thus, SP0535 is the first proven human-specific de novo gene that promotes cortical expansion and folding, and can function through incorporating into an existing conserved molecular network.
Collapse
Affiliation(s)
- Jianhuan Qi
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Fan Mo
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Ni A. An
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Tingwei Mi
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Jiaxin Wang
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Jun‐Tian Qi
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Xiangshang Li
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Boya Zhang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Longkuo Xia
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Yingfei Lu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Gaoying Sun
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xinyue Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Chuan‐Yun Li
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
| |
Collapse
|
53
|
Yuan F, Li Y, Hu R, Gong M, Chai M, Ma X, Cha J, Guo P, Yang K, Li M, Xu M, Ma Q, Su Q, Zhang C, Sheng Z, Wu H, Wang Y, Yuan W, Bian S, Shao L, Zhang R, Li K, Shao Z, Zhang ZN, Li W. Modeling disrupted synapse formation in wolfram syndrome using hESCs-derived neural cells and cerebral organoids identifies Riluzole as a therapeutic molecule. Mol Psychiatry 2023; 28:1557-1570. [PMID: 36750736 DOI: 10.1038/s41380-023-01987-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
Dysregulated neurite outgrowth and synapse formation underlie many psychiatric disorders, which are also manifested by wolfram syndrome (WS). Whether and how the causative gene WFS1 deficiency affects synapse formation remain elusive. By mirroring human brain development with cerebral organoids, WFS1-deficient cerebral organoids not only recapitulate the neuronal loss in WS patients, but also exhibit significantly impaired synapse formation and function associated with reduced astrocytes. WFS1 deficiency in neurons autonomously delays neuronal differentiation with altered expressions of genes associated with psychiatric disorders, and impairs neurite outgrowth and synapse formation with elevated cytosolic calcium. Intriguingly, WFS1 deficiency in astrocytes decreases the expression of glutamate transporter EAAT2 by NF-κB activation and induces excessive glutamate. When co-cultured with wildtype neurons, WFS1-deficient astrocytes lead to impaired neurite outgrowth and increased cytosolic calcium in neurons. Importantly, disrupted synapse formation and function in WFS1-deficient cerebral organoids and impaired neurite outgrowth affected by WFS1-deficient astrocytes are efficiently reversed with Riluzole treatment, by restoring EAAT2 expression in astrocytes. Furthermore, Riluzole rescues the depressive-like behavior in the forced swimming test and the impaired recognition and spatial memory in the novel object test and water maze test in Wfs1 conditional knockout mice. Altogether, our study provides novel insights into how WFS1 deficiency affects synapse formation and function, and offers a strategy to treat this disease.
Collapse
Affiliation(s)
- Fei Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Yana Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Hu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Mengting Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Mengyao Chai
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Xuefei Ma
- QuietD Biotechnology, Ltd., Shanghai, 201210, China
| | - Jiaxue Cha
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Pan Guo
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaijiang Yang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Mushan Li
- Department of Statistics, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Minglu Xu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Qing Ma
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Qiang Su
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Chuan Zhang
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zhejin Sheng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Heng Wu
- Department of Psychosomatic Medicine, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yuan Wang
- Department of Neurology and Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Wen Yuan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Shan Bian
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China
| | - Li Shao
- Department of VIP Clinic, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ru Zhang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaicheng Li
- QuietD Biotechnology, Ltd., Shanghai, 201210, China
| | - Zhen Shao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhen-Ning Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China. .,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China.
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China. .,Tsingtao Advanced Research Institute, Tongji University, Qingdao, 266071, China. .,Reg-Verse Therapeutics (Shanghai) Co. Ltd., Shanghai, 200120, China.
| |
Collapse
|
54
|
Lafferty MJ, Aygün N, Patel NK, Krupa O, Liang D, Wolter JM, Geschwind DH, de la Torre-Ubieta L, Stein JL. MicroRNA-eQTLs in the developing human neocortex link miR-4707-3p expression to brain size. eLife 2023; 12:e79488. [PMID: 36629315 PMCID: PMC9859047 DOI: 10.7554/elife.79488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 01/10/2023] [Indexed: 01/12/2023] Open
Abstract
Expression quantitative trait loci (eQTL) data have proven important for linking non-coding loci to protein-coding genes. But eQTL studies rarely measure microRNAs (miRNAs), small non-coding RNAs known to play a role in human brain development and neurogenesis. Here, we performed small-RNA sequencing across 212 mid-gestation human neocortical tissue samples, measured 907 expressed miRNAs, discovering 111 of which were novel, and identified 85 local-miRNA-eQTLs. Colocalization of miRNA-eQTLs with GWAS summary statistics yielded one robust colocalization of miR-4707-3p expression with educational attainment and brain size phenotypes, where the miRNA expression increasing allele was associated with decreased brain size. Exogenous expression of miR-4707-3p in primary human neural progenitor cells decreased expression of predicted targets and increased cell proliferation, indicating miR-4707-3p modulates progenitor gene regulation and cell fate decisions. Integrating miRNA-eQTLs with existing GWAS yielded evidence of a miRNA that may influence human brain size and function via modulation of neocortical brain development.
Collapse
Affiliation(s)
- Michael J Lafferty
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Nil Aygün
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Niyanta K Patel
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Oleh Krupa
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Dan Liang
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Justin M Wolter
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Luis de la Torre-Ubieta
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel HillChapel HillUnited States
| |
Collapse
|
55
|
Xing Y, Zan C, Liu L. Recent advances in understanding neuronal diversity and neural circuit complexity across different brain regions using single-cell sequencing. Front Neural Circuits 2023; 17:1007755. [PMID: 37063385 PMCID: PMC10097998 DOI: 10.3389/fncir.2023.1007755] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 02/16/2023] [Indexed: 04/18/2023] Open
Abstract
Neural circuits are characterized as interconnecting neuron networks connected by synapses. Some kinds of gene expression and/or functional changes of neurons and synaptic connections may result in aberrant neural circuits, which has been recognized as one crucial pathological mechanism for the onset of many neurological diseases. Gradual advances in single-cell sequencing approaches with strong technological advantages, as exemplified by high throughput and increased resolution for live cells, have enabled it to assist us in understanding neuronal diversity across diverse brain regions and further transformed our knowledge of cellular building blocks of neural circuits through revealing numerous molecular signatures. Currently published transcriptomic studies have elucidated various neuronal subpopulations as well as their distribution across prefrontal cortex, hippocampus, hypothalamus, and dorsal root ganglion, etc. Better characterization of brain region-specific circuits may shed light on new pathological mechanisms involved and assist in selecting potential targets for the prevention and treatment of specific neurological disorders based on their established roles. Given diverse neuronal populations across different brain regions, we aim to give a brief sketch of current progress in understanding neuronal diversity and neural circuit complexity according to their locations. With the special focus on the application of single-cell sequencing, we thereby summarize relevant region-specific findings. Considering the importance of spatial context and connectivity in neural circuits, we also discuss a few published results obtained by spatial transcriptomics. Taken together, these single-cell sequencing data may lay a mechanistic basis for functional identification of brain circuit components, which links their molecular signatures to anatomical regions, connectivity, morphology, and physiology. Furthermore, the comprehensive characterization of neuron subtypes, their distributions, and connectivity patterns via single-cell sequencing is critical for understanding neural circuit properties and how they generate region-dependent interactions in different context.
Collapse
Affiliation(s)
- Yu Xing
- Department of Neurology, Beidahuang Industry Group General Hospital, Harbin, China
| | - Chunfang Zan
- Institute for Stroke and Dementia Research (ISD), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Lu Liu
- Munich Medical Research School (MMRS), LMU Klinikum, Ludwig-Maximilian-University (LMU), Munich, Germany
- *Correspondence: Lu Liu, ,
| |
Collapse
|
56
|
Zhu Z, Huang T, Zhen Z, Wang B, Wu X, Li S. From sMRI to task-fMRI: A unified geometric deep learning framework for cross-modal brain anatomo-functional mapping. Med Image Anal 2023; 83:102681. [PMID: 36459804 DOI: 10.1016/j.media.2022.102681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 07/28/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
Achieving predictions of brain functional activation patterns/task-fMRI maps from its underlying anatomy is an important yet challenging problem. Once successful, it will not only open up new ways to understand how brain anatomy influences functional organization of the brain, but also provide new technical support for the clinical use of anatomical information to guide the localization of cortical functional areas. However, due to the non-Euclidean complex architecture of brain anatomy and the inherent low signal-to-noise ratio (SNR) properties of fMRI signals, the key challenge in building such a cross-modal brain anatomo-functional mapping is how to effectively learn the context-aware information of brain anatomy and overcome the interference of noise-containing task-fMRI labels on the learning process. In this work, we propose a Unified Geometric Deep Learning framework (BrainUGDL) to perform the cross-modal brain anatomo-functional mapping task. Considering that both global and local structures of brain anatomy have an impact on brain functions from their respective perspectives, we innovatively propose the novel Global Graph Encoding (GGE) unit and Local Graph Attention (LGA) unit embedded into two parallel branches, focusing on learning the high-level global and local context information, respectively. Specifically, GGE learns the global context information of each mesh vertex by building and encoding global interactions, and LGA learns the local context information of each mesh vertex by selectively aggregating patch structure enhanced features from its spatial neighbors. The information learnt from the two branches is then fused to form a comprehensive representation of brain anatomical features for final brain function predictions. To address the inevitable measurement noise in task-fMRI labels, we further elaborate a novel uncertainty-filtered learning mechanism, which enables BrainUGDL to realize revised learning from the noise-containing labels through the estimated uncertainty. Experiments across seven open task-fMRI datasets from human connectome project (HCP) demonstrate the superiority of BrainUGDL. To our best knowledge, our proposed BrainUGDL is the first to achieve the prediction of individual task-fMRI maps solely based on brain sMRI data.
Collapse
Affiliation(s)
- Zhiyuan Zhu
- School of Artificial Intelligence, Beijing Normal University, Beijing, China; Engineering Research Center of Intelligent Technology and Educational Application (Beijing Normal University), Ministry of Education, Beijing, China
| | - Taicheng Huang
- Faculty of Psychology, Beijing Normal University, Beijing, China
| | - Zonglei Zhen
- Faculty of Psychology, Beijing Normal University, Beijing, China
| | - Boyu Wang
- Department of Computer Science, Western University, ON, Canada
| | - Xia Wu
- School of Artificial Intelligence, Beijing Normal University, Beijing, China; Engineering Research Center of Intelligent Technology and Educational Application (Beijing Normal University), Ministry of Education, Beijing, China.
| | - Shuo Li
- Department of Computer and Data Science, Case Western Reserve University, Ohio, USA; Department of Biomedical Engineering, Case Western Reserve University, Ohio, USA
| |
Collapse
|
57
|
Zhao J, Feng C, Wang W, Su L, Jiao J. Human SERPINA3 induces neocortical folding and improves cognitive ability in mice. Cell Discov 2022; 8:124. [DOI: 10.1038/s41421-022-00469-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
AbstractNeocortex expansion and folding are related to human intelligence and cognition, but the molecular and cellular mechanisms underlying cortical folding remain poorly understood. Here, we report that the human gene SERPINA3 is linked to gyrification. Specifically, the overexpression of SERPINA3 induced neocortical folding, increased the abundance of neurons, and improved cognitive abilities. Further, SERPINA3 promoted proliferation of the outer radial glia (oRG, also referred to as the basal radial glia) and increased the number of upper-layer neurons. The downstream target Glo1 was determined to be involved in SERPINA3-induced gyrification. Moreover, SERPINA3 increased the proliferation of oRG by binding to the Glo1 promoter. Assessment of behavior performance showed enhanced cognitive abilities in SERPINA3 knock-in mice. Our findings will enrich the understanding of neocortical expansion and gyrification and provide insights into possible treatments for intellectual disability and lissencephaly syndrome.
Collapse
|
58
|
Duński E, Pękowska A. Keeping the balance: Trade-offs between human brain evolution, autism, and schizophrenia. Front Genet 2022; 13:1009390. [DOI: 10.3389/fgene.2022.1009390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022] Open
Abstract
The unique qualities of the human brain are a product of a complex evolutionary process. Evolution, famously described by François Jacob as a “tinkerer,” builds upon existing genetic elements by modifying and repurposing them for new functions. Genetic changes in DNA may lead to the emergence of new genes or cause altered gene expression patterns. Both gene and regulatory element mutations may lead to new functions. Yet, this process may lead to side-effects. An evolutionary trade-off occurs when an otherwise beneficial change, which is important for evolutionary success and is under strong positive selection, concurrently results in a detrimental change in another trait. Pleiotropy occurs when a gene affects multiple traits. Antagonistic pleiotropy is a phenomenon whereby a genetic variant leads to an increase in fitness at one life-stage or in a specific environment, but simultaneously decreases fitness in another respect. Therefore, it is conceivable that the molecular underpinnings of evolution of highly complex traits, including brain size or cognitive ability, under certain conditions could result in deleterious effects, which would increase the susceptibility to psychiatric or neurodevelopmental diseases. Here, we discuss possible trade-offs and antagonistic pleiotropies between evolutionary change in a gene sequence, dosage or activity and the susceptibility of individuals to autism spectrum disorders and schizophrenia. We present current knowledge about genes and alterations in gene regulatory landscapes, which have likely played a role in establishing human-specific traits and have been implicated in those diseases.
Collapse
|
59
|
Fischer J, Fernández Ortuño E, Marsoner F, Artioli A, Peters J, Namba T, Eugster Oegema C, Huttner WB, Ladewig J, Heide M. Human-specific ARHGAP11B ensures human-like basal progenitor levels in hominid cerebral organoids. EMBO Rep 2022; 23:e54728. [PMID: 36098218 PMCID: PMC9646322 DOI: 10.15252/embr.202254728] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 02/06/2023] Open
Abstract
The human-specific gene ARHGAP11B has been implicated in human neocortex expansion. However, the extent of ARHGAP11B's contribution to this expansion during hominid evolution is unknown. Here we address this issue by genetic manipulation of ARHGAP11B levels and function in chimpanzee and human cerebral organoids. ARHGAP11B expression in chimpanzee cerebral organoids doubles basal progenitor levels, the class of cortical progenitors with a key role in neocortex expansion. Conversely, interference with ARHGAP11B's function in human cerebral organoids decreases basal progenitors down to the chimpanzee level. Moreover, ARHGAP11A or ARHGAP11B rescue experiments in ARHGAP11A plus ARHGAP11B double-knockout human forebrain organoids indicate that lack of ARHGAP11B, but not of ARHGAP11A, decreases the abundance of basal radial glia-the basal progenitor type thought to be of particular relevance for neocortex expansion. Taken together, our findings demonstrate that ARHGAP11B is necessary and sufficient to ensure the elevated basal progenitor levels that characterize the fetal human neocortex, suggesting that this human-specific gene was a major contributor to neocortex expansion during human evolution.
Collapse
Affiliation(s)
- Jan Fischer
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Institute for Clinical GeneticsUniversity Hospital Carl Gustav CarusDresdenGermany
| | | | - Fabio Marsoner
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Annasara Artioli
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Neuroscience Center, HiLIFE ‐ Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | | | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Julia Ladewig
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- German Primate CenterLeibniz Institute for Primate ResearchGöttingenGermany
| |
Collapse
|
60
|
Bok’s equi-volume principle: Translation, historical context, and a modern perspective. BRAIN MULTIPHYSICS 2022. [DOI: 10.1016/j.brain.2022.100057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
61
|
Galakhova AA, Hunt S, Wilbers R, Heyer DB, de Kock CPJ, Mansvelder HD, Goriounova NA. Evolution of cortical neurons supporting human cognition. Trends Cogn Sci 2022; 26:909-922. [PMID: 36117080 PMCID: PMC9561064 DOI: 10.1016/j.tics.2022.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 01/12/2023]
Abstract
Human cognitive abilities are generally thought to arise from cortical expansion over the course of human brain evolution. In addition to increased neuron numbers, this cortical expansion might be driven by adaptations in the properties of single neurons and their local circuits. We review recent findings on the distinct structural, functional, and transcriptomic features of human cortical neurons and their organization in cortical microstructure. We focus on the supragranular cortical layers, which showed the most prominent expansion during human brain evolution, and the properties of their principal cells: pyramidal neurons. We argue that the evolutionary adaptations in neuronal features that accompany the expansion of the human cortex partially underlie interindividual variability in human cognitive abilities.
Collapse
Affiliation(s)
- A A Galakhova
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - S Hunt
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - R Wilbers
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - D B Heyer
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - C P J de Kock
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - H D Mansvelder
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - N A Goriounova
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands.
| |
Collapse
|
62
|
Zhang S, Chavoshnejad P, Li X, Guo L, Jiang X, Han J, Wang L, Li G, Wang X, Liu T, Razavi MJ, Zhang S, Zhang T. Gyral peaks: Novel gyral landmarks in developing macaque brains. Hum Brain Mapp 2022; 43:4540-4555. [PMID: 35713202 PMCID: PMC9491295 DOI: 10.1002/hbm.25971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 04/22/2022] [Accepted: 05/23/2022] [Indexed: 11/09/2022] Open
Abstract
Cerebral cortex development undergoes a variety of processes, which provide valuable information for the study of the developmental mechanism of cortical folding as well as its relationship to brain structural architectures and brain functions. Despite the variability in the anatomy-function relationship on the higher-order cortex, recent studies have succeeded in identifying typical cortical landmarks, such as sulcal pits, that bestow specific functional and cognitive patterns and remain invariant across subjects and ages with their invariance being related to a gene-mediated proto-map. Inspired by the success of these studies, we aim in this study at defining and identifying novel cortical landmarks, termed gyral peaks, which are the local highest foci on gyri. By analyzing data from 156 MRI scans of 32 macaque monkeys with the age spanned from 0 to 36 months, we identified 39 and 37 gyral peaks on the left and right hemispheres, respectively. Our investigation suggests that these gyral peaks are spatially consistent across individuals and relatively stable within the age range of this dataset. Moreover, compared with other gyri, gyral peaks have a thicker cortex, higher mean curvature, more pronounced hub-like features in structural connective networks, and are closer to the borders of structural connectivity-based cortical parcellations. The spatial distribution of gyral peaks was shown to correlate with that of other cortical landmarks, including sulcal pits. These results provide insights into the spatial arrangement and temporal development of gyral peaks as well as their relation to brain structure and function.
Collapse
Affiliation(s)
- Songyao Zhang
- School of AutomationNorthwestern Polytechnical UniversityXi'anChina
| | - Poorya Chavoshnejad
- Department of Mechanical EngineeringState University of New York at BinghamtonNew YorkUSA
| | - Xiao Li
- School of Information TechnologyNorthwest UniversityXi'anChina
| | - Lei Guo
- School of AutomationNorthwestern Polytechnical UniversityXi'anChina
| | - Xi Jiang
- School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Junwei Han
- School of AutomationNorthwestern Polytechnical UniversityXi'anChina
| | - Li Wang
- Department of Radiology and BRICUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Gang Li
- Department of Radiology and BRICUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Xianqiao Wang
- College of EngineeringThe University of GeorgiaAthensGeorgiaUSA
| | - Tianming Liu
- Cortical Architecture Imaging and Discovery Lab, Department of Computer Science and Bioimaging Research CenterThe University of GeorgiaAthensGeorgiaUSA
| | - Mir Jalil Razavi
- Department of Mechanical EngineeringState University of New York at BinghamtonNew YorkUSA
| | - Shu Zhang
- Center for Brain and Brain‐Inspired Computing Research, Department of Computer ScienceNorthwestern Polytechnical UniversityXi'anChina
| | - Tuo Zhang
- School of AutomationNorthwestern Polytechnical UniversityXi'anChina
| |
Collapse
|
63
|
Massimo M, Long KR. Orchestrating human neocortex development across the scales; from micro to macro. Semin Cell Dev Biol 2022; 130:24-36. [PMID: 34583893 DOI: 10.1016/j.semcdb.2021.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/27/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
How our brains have developed to perform the many complex functions that make us human has long remained a question of great interest. Over the last few decades, many scientists from a wide range of fields have tried to answer this question by aiming to uncover the mechanisms that regulate the development of the human neocortex. They have approached this on different scales, focusing microscopically on individual cells all the way up to macroscopically imaging entire brains within living patients. In this review we will summarise these key findings and how they fit together.
Collapse
Affiliation(s)
- Marco Massimo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom
| | - Katherine R Long
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
64
|
Wagner NR, Sinha A, Siththanandan V, Kowalchuk AM, MacDonald JL, Tharin S. miR-409-3p represses Cited2 to refine neocortical layer V projection neuron identity. Front Neurosci 2022; 16:931333. [PMID: 36248641 PMCID: PMC9558290 DOI: 10.3389/fnins.2022.931333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/13/2022] [Indexed: 12/14/2022] Open
Abstract
The evolutionary emergence of the corticospinal tract and corpus callosum are thought to underpin the expansion of complex motor and cognitive abilities in mammals. Molecular mechanisms regulating development of the neurons whose axons comprise these tracts, the corticospinal and callosal projection neurons, remain incompletely understood. Our previous work identified a genomic cluster of microRNAs (miRNAs), Mirg/12qF1, that is unique to placental mammals and specifically expressed by corticospinal neurons, and excluded from callosal projection neurons, during development. We found that one of these, miR-409-3p, can convert layer V callosal into corticospinal projection neurons, acting in part through repression of the transcriptional regulator Lmo4. Here we show that miR-409-3p also directly represses the transcriptional co-regulator Cited2, which is highly expressed by callosal projection neurons from the earliest stages of neurogenesis. Cited2 is highly expressed by intermediate progenitor cells (IPCs) in the embryonic neocortex while Mirg, which encodes miR-409-3p, is excluded from these progenitors. miR-409-3p gain-of-function (GOF) in IPCs results in a phenocopy of established Cited2 loss-of-function (LOF). At later developmental stages, both miR-409-3p GOF and Cited2 LOF promote the expression of corticospinal at the expense of callosal projection neuron markers in layer V. Taken together, this work identifies previously undescribed roles for miR-409-3p in controlling IPC numbers and for Cited2 in controlling callosal fate. Thus, miR-409-3p, possibly in cooperation with other Mirg/12qF1 miRNAs, represses Cited2 as part of the multifaceted regulation of the refinement of neuronal cell fate within layer V, combining molecular regulation at multiple levels in both progenitors and post-mitotic neurons.
Collapse
Affiliation(s)
- Nikolaus R. Wagner
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| | - Ashis Sinha
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| | - Verl Siththanandan
- Department of Neurosurgery, Stanford University Medical Center, Center for Academic Medicine, Palo Alto, CA, United States
| | - Angelica M. Kowalchuk
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| | - Jessica L. MacDonald
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States,*Correspondence: Jessica L. MacDonald,
| | - Suzanne Tharin
- Department of Neurosurgery, Stanford University Medical Center, Center for Academic Medicine, Palo Alto, CA, United States,Division of Neurosurgery, Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, United States,Suzanne Tharin,
| |
Collapse
|
65
|
Fischer J, Fernández Ortuño E, Marsoner F, Artioli A, Peters J, Namba T, Eugster Oegema C, Huttner WB, Ladewig J, Heide M. Human-specific ARHGAP11B ensures human-like basal progenitor levels in hominid cerebral organoids. EMBO Rep 2022; 23:e54728. [PMID: 36381990 PMCID: PMC9646322 DOI: 10.1101/2020.10.01.322792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 06/16/2023] Open
Abstract
The human-specific gene ARHGAP11B has been implicated in human neocortex expansion. However, the extent of ARHGAP11B's contribution to this expansion during hominid evolution is unknown. Here we address this issue by genetic manipulation of ARHGAP11B levels and function in chimpanzee and human cerebral organoids. ARHGAP11B expression in chimpanzee cerebral organoids doubles basal progenitor levels, the class of cortical progenitors with a key role in neocortex expansion. Conversely, interference with ARHGAP11B's function in human cerebral organoids decreases basal progenitors down to the chimpanzee level. Moreover, ARHGAP11A or ARHGAP11B rescue experiments in ARHGAP11A plus ARHGAP11B double-knockout human forebrain organoids indicate that lack of ARHGAP11B, but not of ARHGAP11A, decreases the abundance of basal radial glia - the basal progenitor type thought to be of particular relevance for neocortex expansion. Taken together, our findings demonstrate that ARHGAP11B is necessary and sufficient to ensure the elevated basal progenitor levels that characterize the fetal human neocortex, suggesting that this human-specific gene was a major contributor to neocortex expansion during human evolution.
Collapse
Affiliation(s)
- Jan Fischer
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Institute for Clinical GeneticsUniversity Hospital Carl Gustav CarusDresdenGermany
| | | | - Fabio Marsoner
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Annasara Artioli
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Neuroscience Center, HiLIFE ‐ Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | | | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Julia Ladewig
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- German Primate CenterLeibniz Institute for Primate ResearchGöttingenGermany
| |
Collapse
|
66
|
Pinson A, Xing L, Namba T, Kalebic N, Peters J, Oegema CE, Traikov S, Reppe K, Riesenberg S, Maricic T, Derihaci R, Wimberger P, Pääbo S, Huttner WB. Human TKTL1 implies greater neurogenesis in frontal neocortex of modern humans than Neanderthals. Science 2022; 377:eabl6422. [PMID: 36074851 DOI: 10.1126/science.abl6422] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neanderthal brains were similar in size to those of modern humans. We sought to investigate potential differences in neurogenesis during neocortex development. Modern human transketolase-like 1 (TKTL1) differs from Neanderthal TKTL1 by a lysine-to-arginine amino acid substitution. Using overexpression in developing mouse and ferret neocortex, knockout in fetal human neocortical tissue, and genome-edited cerebral organoids, we found that the modern human variant, hTKTL1, but not the Neanderthal variant, increases the abundance of basal radial glia (bRG) but not that of intermediate progenitors (bIPs). bRG generate more neocortical neurons than bIPs. The hTKTL1 effect requires the pentose phosphate pathway and fatty acid synthesis. Inhibition of these metabolic pathways reduces bRG abundance in fetal human neocortical tissue. Our data suggest that neocortical neurogenesis in modern humans differs from that in Neanderthals.
Collapse
Affiliation(s)
- Anneline Pinson
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | | | - Sofia Traikov
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Katrin Reppe
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stephan Riesenberg
- Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Tomislav Maricic
- Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Razvan Derihaci
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, 01307 Dresden, Germany
| | - Pauline Wimberger
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, 01307 Dresden, Germany
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
67
|
Willbrand EH, Voorhies WI, Yao JK, Weiner KS, Bunge SA. Presence or absence of a prefrontal sulcus is linked to reasoning performance during child development. Brain Struct Funct 2022; 227:2543-2551. [PMID: 35932310 PMCID: PMC9418286 DOI: 10.1007/s00429-022-02539-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/05/2022] [Indexed: 12/27/2022]
Abstract
The relationship between structural variability in late-developing association cortices like the lateral prefrontal cortex (LPFC) and the development of higher-order cognitive skills is not well understood. Recent findings show that the morphology of LPFC sulci predicts reasoning performance; this work led to the observation of substantial individual variability in the morphology of one of these sulci, the para-intermediate frontal sulcus (pimfs). Here, we sought to characterize this variability and assess its behavioral significance. To this end, we identified the pimfs in a developmental cohort of 72 participants, ages 6-18. Subsequent analyses revealed that the presence or absence of the ventral component of the pimfs was associated with reasoning, even when controlling for age. This finding shows that the cortex lining the banks of sulci can support the development of complex cognitive abilities and highlights the importance of considering individual differences in local morphology when exploring the neurodevelopmental basis of cognition.
Collapse
Affiliation(s)
- Ethan H Willbrand
- Department of Psychology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Willa I Voorhies
- Department of Psychology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Jewelia K Yao
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08540, USA
| | - Kevin S Weiner
- Department of Psychology, University of California Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
| | - Silvia A Bunge
- Department of Psychology, University of California Berkeley, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
68
|
Brault J, Bardin S, Lampic M, Carpentieri JA, Coquand L, Penisson M, Lachuer H, Victoria GS, Baloul S, El Marjou F, Boncompain G, Miserey‐Lenkei S, Belvindrah R, Fraisier V, Francis F, Perez F, Goud B, Baffet AD. RAB6
and dynein drive
post‐Golgi
apical transport to prevent neuronal progenitor delamination. EMBO Rep 2022; 23:e54605. [PMID: 35979738 PMCID: PMC9535803 DOI: 10.15252/embr.202254605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 12/03/2022] Open
Abstract
Radial glial (RG) cells are the neural stem cells of the developing neocortex. Apical RG (aRG) cells can delaminate to generate basal RG (bRG) cells, a cell type associated with human brain expansion. Here, we report that aRG delamination is regulated by the post‐Golgi secretory pathway. Using in situ subcellular live imaging, we show that post‐Golgi transport of RAB6+ vesicles occurs toward the minus ends of microtubules and depends on dynein. We demonstrate that the apical determinant Crumbs3 (CRB3) is also transported by dynein. Double knockout of RAB6A/A' and RAB6B impairs apical localization of CRB3 and induces a retraction of aRG cell apical process, leading to delamination and ectopic division. These defects are phenocopied by knockout of the dynein activator LIS1. Overall, our results identify a RAB6‐dynein‐LIS1 complex for Golgi to apical surface transport in aRG cells, and highlights the role of this pathway in the maintenance of neuroepithelial integrity.
Collapse
Affiliation(s)
| | - Sabine Bardin
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Marusa Lampic
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | | | - Laure Coquand
- Institut Curie PSL Research University, CNRS UMR144 Paris France
- Sorbonne University Paris France
| | - Maxime Penisson
- Sorbonne University Paris France
- INSERM UMR‐S 1270 Paris France
- Institut du Fer à Moulin Paris France
| | - Hugo Lachuer
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | | | - Sarah Baloul
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Fatima El Marjou
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | | | | | - Richard Belvindrah
- Sorbonne University Paris France
- INSERM UMR‐S 1270 Paris France
- Institut du Fer à Moulin Paris France
| | - Vincent Fraisier
- UMR 144‐Cell and Tissue Imaging Facility (PICT‐IBiSA) CNRS‐Institut Curie Paris France
| | - Fiona Francis
- Sorbonne University Paris France
- INSERM UMR‐S 1270 Paris France
- Institut du Fer à Moulin Paris France
| | - Franck Perez
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Bruno Goud
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Alexandre D Baffet
- Institut Curie PSL Research University, CNRS UMR144 Paris France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Paris France
| |
Collapse
|
69
|
Huang Y, Wu Z, Wang F, Hu D, Li T, Guo L, Wang L, Lin W, Li G. Mapping developmental regionalization and patterns of cortical surface area from 29 post-menstrual weeks to 2 years of age. Proc Natl Acad Sci U S A 2022; 119:e2121748119. [PMID: 35939665 PMCID: PMC9388141 DOI: 10.1073/pnas.2121748119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Surface area of the human cerebral cortex expands extremely dynamically and regionally heterogeneously from the third trimester of pregnancy to 2 y of age, reflecting the spatial heterogeneity of the underlying microstructural and functional development of the cerebral cortex. However, little is known about the developmental patterns and regionalization of cortical surface area during this critical stage, due to the lack of high-quality imaging data and accurate computational tools for pediatric brain MRI data. To fill this critical knowledge gap, by leveraging 1,037 high-quality MRI scans with the age between 29 post-menstrual weeks and 24 mo from 735 pediatric subjects in two complementary datasets, i.e., the Baby Connectome Project (BCP) and the developing Human Connectome Project (dHCP), and state-of-the-art dedicated image-processing tools, we unprecedentedly parcellate the cerebral cortex into a set of distinct subdivisions purely according to the developmental patterns of the cortical surface. Our discovered developmentally distinct subdivisions correspond well to structurally and functionally meaningful regions and reveal spatially contiguous, hierarchical, and bilaterally symmetric patterns of early cortical surface expansion. We also show that high-order association subdivisions, where cortical folds emerge later during prenatal stages, undergo more dramatic cortical surface expansion during infancy, compared with the central regions, especially the sensorimotor and insula cortices, thus forming a distinct central-pole division in early cortical surface expansion. These results provide an important reference for exploring and understanding dynamic early brain development in health and disease.
Collapse
Affiliation(s)
- Ying Huang
- School of Automation, Northwestern Polytechnical University, Xi’an 710071, China
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Zhengwang Wu
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Fan Wang
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Dan Hu
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Tengfei Li
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Lei Guo
- School of Automation, Northwestern Polytechnical University, Xi’an 710071, China
| | - Li Wang
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Weili Lin
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Gang Li
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
70
|
Fasano G, Compagnucci C, Dallapiccola B, Tartaglia M, Lauri A. Teleost Fish and Organoids: Alternative Windows Into the Development of Healthy and Diseased Brains. Front Mol Neurosci 2022; 15:855786. [PMID: 36034498 PMCID: PMC9403253 DOI: 10.3389/fnmol.2022.855786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
The variety in the display of animals’ cognition, emotions, and behaviors, typical of humans, has its roots within the anterior-most part of the brain: the forebrain, giving rise to the neocortex in mammals. Our understanding of cellular and molecular events instructing the development of this domain and its multiple adaptations within the vertebrate lineage has progressed in the last decade. Expanding and detailing the available knowledge on regionalization, progenitors’ behavior and functional sophistication of the forebrain derivatives is also key to generating informative models to improve our characterization of heterogeneous and mechanistically unexplored cortical malformations. Classical and emerging mammalian models are irreplaceable to accurately elucidate mechanisms of stem cells expansion and impairments of cortex development. Nevertheless, alternative systems, allowing a considerable reduction of the burden associated with animal experimentation, are gaining popularity to dissect basic strategies of neural stem cells biology and morphogenesis in health and disease and to speed up preclinical drug testing. Teleost vertebrates such as zebrafish, showing conserved core programs of forebrain development, together with patients-derived in vitro 2D and 3D models, recapitulating more accurately human neurogenesis, are now accepted within translational workflows spanning from genetic analysis to functional investigation. Here, we review the current knowledge of common and divergent mechanisms shaping the forebrain in vertebrates, and causing cortical malformations in humans. We next address the utility, benefits and limitations of whole-brain/organism-based fish models or neuronal ensembles in vitro for translational research to unravel key genes and pathological mechanisms involved in neurodevelopmental diseases.
Collapse
|
71
|
Studer M, Rossini L, Spreafico R, Pelliccia V, Tassi L, de Curtis M, Garbelli R. Why are type II focal cortical dysplasias frequently located at the bottom of sulcus? A neurodevelopmental hypothesis. Epilepsia 2022; 63:2716-2721. [PMID: 35932101 DOI: 10.1111/epi.17386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022]
Affiliation(s)
| | - Laura Rossini
- Epilepsy Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milano, Italy
| | - Roberto Spreafico
- Epilepsy Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milano, Italy
| | - Veronica Pelliccia
- "Claudio Munari" Epilepsy Surgery Centre, Niguarda Hospital, Milano, Italy
| | - Laura Tassi
- "Claudio Munari" Epilepsy Surgery Centre, Niguarda Hospital, Milano, Italy
| | - Marco de Curtis
- Epilepsy Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milano, Italy
| | - Rita Garbelli
- Epilepsy Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milano, Italy
| |
Collapse
|
72
|
Brain organoids: the quest to decipher human-specific features of brain development. Curr Opin Genet Dev 2022; 75:101955. [PMID: 35816938 DOI: 10.1016/j.gde.2022.101955] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/31/2022] [Accepted: 06/10/2022] [Indexed: 11/21/2022]
Abstract
The development of the human brain occurs largely in utero over long periods of time and is thus experimentally inaccessible; therefore, tractable experimental models are needed. Human brain organoid have emerged as powerful model systems to investigate human-specific features of brain development. Focusing on the cerebral cortex, here, we discuss how brain, and more specifically cortical, organoid models have newly enabled discovery of aspects of progenitor biology and cortical-cell diversification that are unique to humans. We foresee that as advancements in organoid generation increase the complexity of these models, more complete replicas of the brain will empower future studies investigating higher-order aspects of brain biology, toward an understanding of the unique processing capabilities of the human brain.
Collapse
|
73
|
Farcy S, Albert A, Gressens P, Baffet AD, El Ghouzzi V. Cortical Organoids to Model Microcephaly. Cells 2022; 11:2135. [PMID: 35883578 PMCID: PMC9320662 DOI: 10.3390/cells11142135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
How the brain develops and achieves its final size is a fascinating issue that questions cortical evolution across species and man's place in the animal kingdom. Although animal models have so far been highly valuable in understanding the key steps of cortical development, many human specificities call for appropriate models. In particular, microcephaly, a neurodevelopmental disorder that is characterized by a smaller head circumference has been challenging to model in mice, which often do not fully recapitulate the human phenotype. The relatively recent development of brain organoid technology from induced pluripotent stem cells (iPSCs) now makes it possible to model human microcephaly, both due to genetic and environmental origins, and to generate developing cortical tissue from the patients themselves. These 3D tissues rely on iPSCs differentiation into cortical progenitors that self-organize into neuroepithelial rosettes mimicking the earliest stages of human neurogenesis in vitro. Over the last ten years, numerous protocols have been developed to control the identity of the induced brain areas, the reproducibility of the experiments and the longevity of the cultures, allowing analysis of the later stages. In this review, we describe the different approaches that instruct human iPSCs to form cortical organoids, summarize the different microcephalic conditions that have so far been modeled by organoids, and discuss the relevance of this model to decipher the cellular and molecular mechanisms of primary and secondary microcephalies.
Collapse
Affiliation(s)
- Sarah Farcy
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Alexandra Albert
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Pierre Gressens
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Alexandre D. Baffet
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Vincent El Ghouzzi
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| |
Collapse
|
74
|
Vuksanović V. Brain morphometric similarity and flexibility. Cereb Cortex Commun 2022; 3:tgac024. [PMID: 35854840 PMCID: PMC9283106 DOI: 10.1093/texcom/tgac024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 05/31/2022] [Accepted: 06/05/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
The cerebral cortex is represented through multiple multilayer morphometric similarity networks to study their modular structures. The approach introduces a novel way for studying brain networks' metrics across individuals, and can quantify network properties usually not revealed using conventional network analyses.
Methods
A total of 8 combinations or types of morphometric similarity networks were constructed – 4 combinations of the inter-regional cortical features on 2 brain atlases. The networks' modular structures were investigated by identifying those modular interactions that stay consistent across the combinations of inter-regional morphometric features and individuals.
Results
The results provide evidence of the community structures as the property of (i) cortical lobar divisions, and also as (ii) the product of different combinations of morphometric features used for the construction of the multilayer representations of the cortex. For the first time, this study has mapped out flexible and inflexible morphometric similarity hubs, and evidence has been provided about variations of the modular network topology across the multilayers with age and IQ.
Conclusions
The results contribute to understanding of intra-regional characteristics in cortical interactions, which potentially can be used to map heterogeneous neurodegeneration patterns in diseased brains.
Collapse
Affiliation(s)
- Vesna Vuksanović
- Health Data Science, Swansea University Medical School, Swansea University, Data Science Building , Swansea SA2 8PP, Wales, United Kingdom
| |
Collapse
|
75
|
Lossi L. Anatomical features for an adequate choice of the experimental animal model in biomedicine: III. Ferret, goat, sheep, and horse. Ann Anat 2022; 244:151978. [PMID: 35787443 DOI: 10.1016/j.aanat.2022.151978] [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: 05/16/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022]
Abstract
The anatomical characteristics of each of the many species today employed in biomedical research are very important when selecting the correct animal model(s), especially for conducting translational research. In previous papers, these features have been considered for fish (D'Angelo et al. Ann. Anat, 2016, 205:75), the most common laboratory rodents, rabbits, and pigs (Lossi et al. 2016). I here follow this line of discussion by dealing with the importance of proper knowledge of ferrets, goats, sheep, and horses' main anatomical features in translational research.
Collapse
Affiliation(s)
- Laura Lossi
- University of Turin, Department of Veterinary Sciences, Turin, Italy; INN, Istituto Nazionale di Neuroscienze, Turin, Italy.
| |
Collapse
|
76
|
Application of the adverse outcome pathway concept for investigating developmental neurotoxicity potential of Chinese herbal medicines by using human neural progenitor cells in vitro. Cell Biol Toxicol 2022; 39:319-343. [PMID: 35701726 PMCID: PMC10042984 DOI: 10.1007/s10565-022-09730-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 05/10/2022] [Indexed: 12/16/2022]
Abstract
Adverse outcome pathways (AOPs) are organized sequences of key events (KEs) that are triggered by a xenobiotic-induced molecular initiating event (MIE) and summit in an adverse outcome (AO) relevant to human or ecological health. The AOP framework causally connects toxicological mechanistic information with apical endpoints for application in regulatory sciences. AOPs are very useful to link endophenotypic, cellular endpoints in vitro to adverse health effects in vivo. In the field of in vitro developmental neurotoxicity (DNT), such cellular endpoints can be assessed using the human "Neurosphere Assay," which depicts different endophenotypes for a broad variety of neurodevelopmental KEs. Combining this model with large-scale transcriptomics, we evaluated DNT hazards of two selected Chinese herbal medicines (CHMs) Lei Gong Teng (LGT) and Tian Ma (TM), and provided further insight into their modes-of-action (MoA). LGT disrupted hNPC migration eliciting an exceptional migration endophenotype. Time-lapse microscopy and intervention studies indicated that LGT disturbs laminin-dependent cell adhesion. TM impaired oligodendrocyte differentiation in human but not rat NPCs and activated a gene expression network related to oxidative stress. The LGT results supported a previously published AOP on radial glia cell adhesion due to interference with integrin-laminin binding, while the results of TM exposure were incorporated into a novel putative, stressor-based AOP. This study demonstrates that the combination of phenotypic and transcriptomic analyses is a powerful tool to elucidate compounds' MoA and incorporate the results into novel or existing AOPs for a better perception of the DNT hazard in a regulatory context.
Collapse
|
77
|
Espinós A, Fernández‐Ortuño E, Negri E, Borrell V. Evolution of genetic mechanisms regulating cortical neurogenesis. Dev Neurobiol 2022; 82:428-453. [PMID: 35670518 PMCID: PMC9543202 DOI: 10.1002/dneu.22891] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/26/2022] [Accepted: 05/24/2022] [Indexed: 11/20/2022]
Abstract
The size of the cerebral cortex increases dramatically across amniotes, from reptiles to great apes. This is primarily due to different numbers of neurons and glial cells produced during embryonic development. The evolutionary expansion of cortical neurogenesis was linked to changes in neural stem and progenitor cells, which acquired increased capacity of self‐amplification and neuron production. Evolution works via changes in the genome, and recent studies have identified a small number of new genes that emerged in the recent human and primate lineages, promoting cortical progenitor proliferation and increased neurogenesis. However, most of the mammalian genome corresponds to noncoding DNA that contains gene‐regulatory elements, and recent evidence precisely points at changes in expression levels of conserved genes as key in the evolution of cortical neurogenesis. Here, we provide an overview of basic cellular mechanisms involved in cortical neurogenesis across amniotes, and discuss recent progress on genetic mechanisms that may have changed during evolution, including gene expression regulation, leading to the expansion of the cerebral cortex.
Collapse
Affiliation(s)
- Alexandre Espinós
- Instituto de Neurociencias CSIC ‐ UMH, 03550 Sant Joan d'Alacant Spain
| | | | - Enrico Negri
- Instituto de Neurociencias CSIC ‐ UMH, 03550 Sant Joan d'Alacant Spain
| | - Víctor Borrell
- Instituto de Neurociencias CSIC ‐ UMH, 03550 Sant Joan d'Alacant Spain
| |
Collapse
|
78
|
Jin Y, Gao X, Lu M, Chen G, Yang X, Ren N, Song Y, Hou C, Li J, Liu Q, Gao J. Loss of BAF (mSWI/SNF) chromatin-remodeling ATPase Brg1 causes multiple malformations of cortical development in mice. Hum Mol Genet 2022; 31:3504-3520. [PMID: 35666215 DOI: 10.1093/hmg/ddac127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/12/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Mutations in genes encoding subunits of the BAF (BRG1/BRM-associated factor) complex cause various neurodevelopmental diseases. However, the underlying pathophysiology remains largely unknown. Here, we analyzed the function of Brg1, a core ATPase of BAF complexes, in the developing cerebral cortex. Loss of Brg1 causes several morphological defects resembling human malformations of cortical development (MCDs), including microcephaly, cortical dysplasia, cobblestone lissencephaly, and periventricular heterotopia. We demonstrated that neural progenitor cell (NPC) renewal, neuronal differentiation, neuronal migration, apoptotic cell death, pial basement membrane, and apical junctional complexes, which are associated with MCD formation, were impaired after Brg1 deletion. Furthermore, transcriptome profiling indicated that a large number of genes were deregulated. The deregulated genes were closely related to MCD formation, and most of these genes were bound by Brg1. Cumulatively, our study indicates an essential role of Brg1 in cortical development and provides a new possible pathogenesis underlying Brg1-based BAF complex-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yecheng Jin
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaotong Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Miaoqing Lu
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315040, China
| | - Ge Chen
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Xiaofan Yang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Naixia Ren
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Yuning Song
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Congzhe Hou
- Department of Reproductive medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Jiangxia Li
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qiji Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiangang Gao
- School of Laboratory Animal Science, Shandong First Medical University, Jinan, Shandong 250117, China
| |
Collapse
|
79
|
Camargo Ortega G, Götz M. Centrosome heterogeneity in stem cells regulates cell diversity. Trends Cell Biol 2022; 32:707-719. [PMID: 35750615 DOI: 10.1016/j.tcb.2022.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 11/27/2022]
Abstract
Stem cells are at the source of creating cellular diversity. Multiple mechanisms, including basic cell biological processes, regulate their fate. The centrosome is at the core of many stem cell functions and recent work highlights the association of distinct proteins at the centrosome in stem cell differentiation. As showcased by a novel centrosome protein regulating neural stem cell differentiation, it is timely to review the heterogeneity of the centrosome at protein and RNA levels and how this impacts their function in stem and progenitor cells. Together with evidence for heterogeneity of other organelles so far considered as similar between cells, we call for exploring the cell type-specific composition of organelles as a way to expand protein function in development with relevance to regenerative medicine.
Collapse
Affiliation(s)
- Germán Camargo Ortega
- Department of Biosystems Science and Engineering, ETH, Zurich, 4058 Basel, Switzerland.
| | - Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, 82152 Planegg-Martinsried, Germany; Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, 82152 Planegg-Martinsried, Germany; 4 SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
80
|
Leibovitz Z, Lerman-Sagie T, Haddad L. Fetal Brain Development: Regulating Processes and Related Malformations. Life (Basel) 2022; 12:life12060809. [PMID: 35743840 PMCID: PMC9224903 DOI: 10.3390/life12060809] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/16/2022] Open
Abstract
This paper describes the contemporary state of knowledge regarding processes that regulate normal development of the embryonic–fetal central nervous system (CNS). The processes are described according to the developmental timetable: dorsal induction, ventral induction, neurogenesis, neuronal migration, post-migration neuronal development, and cortical organization. We review the current literature on CNS malformations associated with these regulating processes. We specifically address neural tube defects, holoprosencephaly, malformations of cortical development (including microcephaly, megalencephaly, lissencephaly, cobblestone malformations, gray matter heterotopia, and polymicrogyria), disorders of the corpus callosum, and posterior fossa malformations. Fetal ventriculomegaly, which frequently accompanies these disorders, is also reviewed. Each malformation is described with reference to the etiology, genetic causes, prenatal sonographic imaging, associated anomalies, differential diagnosis, complimentary diagnostic studies, clinical interventions, neurodevelopmental outcome, and life quality.
Collapse
Affiliation(s)
- Zvi Leibovitz
- Obstetrics-Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Fetal Neurology Clinic, Wolfson Medical Center, Holon and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 5822012, Israel;
- Obstetrics-Gynecology Ultrasound Unit, Bnai-Zion Medical Center, Rappaport Faculty of Medicine, The Technion, Haifa 31048, Israel;
- Correspondence:
| | - Tally Lerman-Sagie
- Obstetrics-Gynecology Ultrasound Unit, Department of Obstetrics and Gynecology, Fetal Neurology Clinic, Wolfson Medical Center, Holon and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 5822012, Israel;
- Pediatric Neurology Unit, Wolfson Medical Center, Holon and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 5822012, Israel
| | - Leila Haddad
- Obstetrics-Gynecology Ultrasound Unit, Bnai-Zion Medical Center, Rappaport Faculty of Medicine, The Technion, Haifa 31048, Israel;
| |
Collapse
|
81
|
Wang Y, Cui H, Esworthy T, Mei D, Wang Y, Zhang LG. Emerging 4D Printing Strategies for Next-Generation Tissue Regeneration and Medical Devices. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109198. [PMID: 34951494 DOI: 10.1002/adma.202109198] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/17/2021] [Indexed: 06/14/2023]
Abstract
The rapid development of 3D printing has led to considerable progress in the field of biomedical engineering. Notably, 4D printing provides a potential strategy to achieve a time-dependent physical change within tissue scaffolds or replicate the dynamic biological behaviors of native tissues for smart tissue regeneration and the fabrication of medical devices. The fabricated stimulus-responsive structures can offer dynamic, reprogrammable deformation or actuation to mimic complex physical, biochemical, and mechanical processes of native tissues. Although there is notable progress made in the development of the 4D printing approach for various biomedical applications, its more broad-scale adoption for clinical use and tissue engineering purposes is complicated by a notable limitation of printable smart materials and the simplistic nature of achievable responses possible with current sources of stimulation. In this review, the recent progress made in the field of 4D printing by discussing the various printing mechanisms that are achieved with great emphasis on smart ink mechanisms of 4D actuation, construct structural design, and printing technologies, is highlighted. Recent 4D printing studies which focus on the applications of tissue/organ regeneration and medical devices are then summarized. Finally, the current challenges and future perspectives of 4D printing are also discussed.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Fluid Power and Mechatronics Systems, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, 20052, USA
| | - Deqing Mei
- State Key Laboratory of Fluid Power and Mechatronics Systems, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yancheng Wang
- State Key Laboratory of Fluid Power and Mechatronics Systems, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Electrical and Computer Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Biomedical Engineering, The George Washington University, Washington, DC, 20052, USA
- Department of Medicine, The George Washington University, Washington, DC, 20052, USA
| |
Collapse
|
82
|
Demirci N, Holland MA. Cortical thickness systematically varies with curvature and depth in healthy human brains. Hum Brain Mapp 2022; 43:2064-2084. [PMID: 35098606 PMCID: PMC8933257 DOI: 10.1002/hbm.25776] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/30/2021] [Accepted: 01/05/2022] [Indexed: 12/30/2022] Open
Abstract
Cortical thickness varies throughout the cortex in a systematic way. However, it is challenging to investigate the patterns of cortical thickness due to the intricate geometry of the cortex. The cortex has a folded nature both in radial and tangential directions which forms not only gyri and sulci but also tangential folds and intersections. In this article, cortical curvature and depth are used to characterize the spatial distribution of the cortical thickness with much higher resolution than conventional regional atlases. To do this, a computational pipeline was developed that is capable of calculating a variety of quantitative measures such as surface area, cortical thickness, curvature (mean curvature, Gaussian curvature, shape index, intrinsic curvature index, and folding index), and sulcal depth. By analyzing 501 neurotypical adult human subjects from the ABIDE-I dataset, we show that cortex has a very organized structure and cortical thickness is strongly correlated with local shape. Our results indicate that cortical thickness consistently increases along the gyral-sulcal spectrum from concave to convex shape, encompassing the saddle shape along the way. Additionally, tangential folds influence cortical thickness in a similar way as gyral and sulcal folds; outer folds are consistently thicker than inner.
Collapse
Affiliation(s)
- Nagehan Demirci
- Bioengineering Graduate ProgramUniversity of Notre DameNotre DameIndianaUSA
| | - Maria A. Holland
- Bioengineering Graduate ProgramUniversity of Notre DameNotre DameIndianaUSA
- Department of Aerospace and Mechanical EngineeringUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
83
|
Shinmyo Y, Hamabe-Horiike T, Saito K, Kawasaki H. Investigation of the Mechanisms Underlying the Development and Evolution of the Cerebral Cortex Using Gyrencephalic Ferrets. Front Cell Dev Biol 2022; 10:847159. [PMID: 35386196 PMCID: PMC8977464 DOI: 10.3389/fcell.2022.847159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex has changed significantly during evolution. As a result of the increase in the number of neurons and glial cells in the cerebral cortex, its size has markedly expanded. Moreover, folds, called gyri and sulci, appeared on its surface, and its neuronal circuits have become much more complicated. Although these changes during evolution are considered to have been crucial for the acquisition of higher brain functions, the mechanisms underlying the development and evolution of the cerebral cortex of mammals are still unclear. This is, at least partially, because it is difficult to investigate these mechanisms using mice only. Therefore, genetic manipulation techniques for the cerebral cortex of gyrencephalic carnivore ferrets were developed recently. Furthermore, gene knockout was achieved in the ferret cerebral cortex using the CRISPR/Cas9 system. These techniques enabled molecular investigations using the ferret cerebral cortex. In this review, we will summarize recent findings regarding the mechanisms underlying the development and evolution of the mammalian cerebral cortex, mainly focusing on research using ferrets.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
84
|
Hajdu C, Kumar P, Horváth D, Tóth Á. Pattern selection of directionally oriented chitosan tubes. J Chem Phys 2022; 156:134902. [PMID: 35395898 DOI: 10.1063/5.0087961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The growth of viscoelastic curved materials, inspired by biological systems, may give rise to various complex structures. One of the simplest ways to control the pattern formation is to vary the orientation of the reaction vessel while keeping all other experimental conditions constant. Here, we report the self-organization of soft chitosan tubes by injecting acidic chitosan sol into a pool of sodium hydroxide solution, where the adhesive force between the gel and container keeps the tubules on the bottom of the reactor. The horizontal growth of the tubular structure undergoes spontaneous symmetry breaking, where instabilities develop on the surface of the chitosan tubules. Transformation of folds into wrinkles and finally to a smooth tube takes place by varying the orientation of the container. In addition to characterizing the evolving structures, we have also shown that the linear growth rate of the tube scales with the tilt angle of the container from the horizontal.
Collapse
Affiliation(s)
- Cintia Hajdu
- Department of Physical Chemistry and Materials Science, University of Szeged, Rerrich Béla tér 1., Szeged H-6720, Hungary
| | - Pawan Kumar
- Department of Physical Chemistry and Materials Science, University of Szeged, Rerrich Béla tér 1., Szeged H-6720, Hungary
| | - Dezső Horváth
- Department of Applied and Environmental Chemistry, University of Szeged, Rerrich Béla tér 1., Szeged H-6720, Hungary
| | - Ágota Tóth
- Department of Physical Chemistry and Materials Science, University of Szeged, Rerrich Béla tér 1., Szeged H-6720, Hungary
| |
Collapse
|
85
|
Wang W, Su L, Ji F, Zhang D, Wang Y, Zhao J, Jiao RD, Zhang M, Huang E, Jiang H, Zhang J, Jiao J. The human FOXM1 homolog promotes basal progenitor cell proliferation and cortical folding in mouse. EMBO Rep 2022; 23:e53602. [PMID: 34935271 PMCID: PMC8892259 DOI: 10.15252/embr.202153602] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 11/29/2021] [Accepted: 12/13/2021] [Indexed: 11/09/2022] Open
Abstract
Cortical expansion and folding are key processes in human brain development and evolution and are considered to be principal elements of intellectual ability. How cortical folding has evolved and is induced during embryo development is not well understood. Here, we show that the expression of human FOXM1 promotes basal progenitor cell proliferation and induces cortical thickening and folding in mice. Human-specific protein sequences further promote the generation of basal progenitor cells. Human FOXM1 increases the proliferation of neural progenitors by binding to the Lin28a promoter and increasing Lin28a expression. Furthermore, overexpression of LIN28A rescues the proliferation of human FOXM1 knockout neural progenitor cells. Together, our findings demonstrate that a human gene can increase the number of basal progenitor cells in mice, leading to brain size increase and gyrification, and may thus contribute to evolutionary brain development and cortical expansion.
Collapse
Affiliation(s)
- Wenwen Wang
- School of Life SciencesUniversity of Science and Technology of ChinaHefeiChina,State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Libo Su
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Fen Ji
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Dongming Zhang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Yanyan Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Jinyue Zhao
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | | | - Mengtian Zhang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina
| | - Enyu Huang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical UniversityZhanjiangChina
| | - Hong Jiang
- Department of PhysiologyShandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: PhysiologySchool of Basic MedicineMedical CollegeQingdao UniversityQingdaoChina
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical UniversityZhanjiangChina
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina,University of Chinese Academy of SciencesBeijingChina,Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina,Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| |
Collapse
|
86
|
Alenyá M, Wang X, Lefévre J, Auzias G, Fouquet B, Eixarch E, Rousseau F, Camara O. Computational pipeline for the generation and validation of patient-specific mechanical models of brain development. BRAIN MULTIPHYSICS 2022. [DOI: 10.1016/j.brain.2022.100045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
87
|
Tomasello U, Klingler E, Niquille M, Mule N, Santinha AJ, de Vevey L, Prados J, Platt RJ, Borrell V, Jabaudon D, Dayer A. miR-137 and miR-122, two outer subventricular zone non-coding RNAs, regulate basal progenitor expansion and neuronal differentiation. Cell Rep 2022; 38:110381. [PMID: 35172154 PMCID: PMC8864305 DOI: 10.1016/j.celrep.2022.110381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 01/24/2022] [Indexed: 12/29/2022] Open
Abstract
Cortical expansion in primate brains relies on enlargement of germinal zones during a prolonged developmental period. Although most mammals have two cortical germinal zones, the ventricular zone (VZ) and subventricular zone (SVZ), gyrencephalic species display an additional germinal zone, the outer subventricular zone (oSVZ), which increases the number and diversity of neurons generated during corticogenesis. How the oSVZ emerged during evolution is poorly understood, but recent studies suggest a role for non-coding RNAs, which allow tight genetic program regulation during development. Here, using in vivo functional genetics, single-cell RNA sequencing, live imaging, and electrophysiology to assess progenitor and neuronal properties in mice, we identify two oSVZ-expressed microRNAs (miRNAs), miR-137 and miR-122, which regulate key cellular features of cortical expansion. miR-137 promotes basal progenitor self-replication and superficial layer neuron fate, whereas miR-122 decreases the pace of neuronal differentiation. These findings support a cell-type-specific role of miRNA-mediated gene expression in cortical expansion. oSVZ-expressed microRNAs 137 and 122 promote superficial layer identity of neurons miR-137 promotes basal progenitor proliferation and layer 2/3 neuron generation miR-122 slows down neuronal differentiation pace
Collapse
Affiliation(s)
- Ugo Tomasello
- Department of Basic Neurosciences, University of Geneva, 1205 Geneva, Switzerland; Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Esther Klingler
- Department of Basic Neurosciences, University of Geneva, 1205 Geneva, Switzerland
| | - Mathieu Niquille
- Department of Basic Neurosciences, University of Geneva, 1205 Geneva, Switzerland; Department of Psychiatry, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Nandkishor Mule
- Department of Basic Neurosciences, University of Geneva, 1205 Geneva, Switzerland
| | - Antonio J Santinha
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Laura de Vevey
- Department of Basic Neurosciences, University of Geneva, 1205 Geneva, Switzerland
| | - Julien Prados
- Department of Psychiatry, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Randall J Platt
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Victor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, 1205 Geneva, Switzerland; Clinic of Neurology, Geneva University Hospital, 1205 Geneva, Switzerland.
| | - Alexandre Dayer
- Department of Basic Neurosciences, University of Geneva, 1205 Geneva, Switzerland; Department of Psychiatry, Geneva University Hospital, 1205 Geneva, Switzerland
| |
Collapse
|
88
|
Jash S, Sharma S. Pathogenic Infections during Pregnancy and the Consequences for Fetal Brain Development. Pathogens 2022; 11:pathogens11020193. [PMID: 35215136 PMCID: PMC8877441 DOI: 10.3390/pathogens11020193] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Pathogens comprised of viruses, bacteria, gut microbiome, and parasites are a leading cause of ever-emerging diseases in humans. Studying pathogens for their ability to cause diseases is a topic of critical discussion among scientists and pharmaceutical centers for effective drug development that diagnose, treat, and prevent infection-associated disorders. Pathogens impact health either directly by invading the host or by eliciting an acute inflammatory immune response. This paradigm of inflammatory immune responses is even more consequential in people who may be immunocompromised. In this regard, pregnancy offers an altered immunity scenario, which may allow the onset of severe diseases. Viruses, such as Influenza, HIV, and now SARS-CoV-2, associated with the COVID-19 pandemic, raise new concerns for maternal and fetal/neonatal health. Intrauterine bacterial and parasitic infections are also known to impact pregnancy outcomes and neonatal health. More importantly, viral and bacterial infections during pregnancy have been identified as a common contributor to fetal brain development defects. Infection-mediated inflammatory uterine immune milieu is thought to be the main trigger for causing poor fetal brain development, resulting in long-term cognitive impairments. The concept of in utero programming of childhood and adult disorders has revolutionized the field of neurodevelopment and its associated complications. Recent findings in mice and humans clearly support the idea that uterine immunity during pregnancy controls the health trajectory of the child and considerably influences the cognitive function and mental health. In this review, we focus on the in utero programming of autism spectrum disorders (ASD) and assess the effects of pathogens on the onset of ASD-like symptoms.
Collapse
|
89
|
Lee WS, Baldassari S, Stephenson SEM, Lockhart PJ, Baulac S, Leventer RJ. Cortical Dysplasia and the mTOR Pathway: How the Study of Human Brain Tissue Has Led to Insights into Epileptogenesis. Int J Mol Sci 2022; 23:1344. [PMID: 35163267 PMCID: PMC8835853 DOI: 10.3390/ijms23031344] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 02/01/2023] Open
Abstract
Type II focal cortical dysplasia (FCD) is a neuropathological entity characterised by cortical dyslamination with the presence of dysmorphic neurons only (FCDIIA) or the presence of both dysmorphic neurons and balloon cells (FCDIIB). The year 2021 marks the 50th anniversary of the recognition of FCD as a cause of drug resistant epilepsy, and it is now the most common reason for epilepsy surgery. The causes of FCD remained unknown until relatively recently. The study of resected human FCD tissue using novel genomic technologies has led to remarkable advances in understanding the genetic basis of FCD. Mechanistic parallels have emerged between these non-neoplastic lesions and neoplastic disorders of cell growth and differentiation, especially through perturbations of the mammalian target of rapamycin (mTOR) signalling pathway. This narrative review presents the advances through which the aetiology of FCDII has been elucidated in chronological order, from recognition of an association between FCD and the mTOR pathway to the identification of somatic mosaicism within FCD tissue. We discuss the role of a two-hit mechanism, highlight current challenges and future directions in detecting somatic mosaicism in brain and discuss how knowledge of FCD may inform novel precision treatments of these focal epileptogenic malformations of human cortical development.
Collapse
Affiliation(s)
- Wei Shern Lee
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Parkville 3052, Australia; (W.S.L.); (S.E.M.S.); (P.J.L.)
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Sara Baldassari
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France;
| | - Sarah E. M. Stephenson
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Parkville 3052, Australia; (W.S.L.); (S.E.M.S.); (P.J.L.)
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Paul J. Lockhart
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Parkville 3052, Australia; (W.S.L.); (S.E.M.S.); (P.J.L.)
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Stéphanie Baulac
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France;
| | - Richard J. Leventer
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Department of Neurology, The Royal Children’s Hospital, Parkville 3052, Australia
| |
Collapse
|
90
|
Ossola C, Kalebic N. Roots of the Malformations of Cortical Development in the Cell Biology of Neural Progenitor Cells. Front Neurosci 2022; 15:817218. [PMID: 35069108 PMCID: PMC8766818 DOI: 10.3389/fnins.2021.817218] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
The cerebral cortex is a structure that underlies various brain functions, including cognition and language. Mammalian cerebral cortex starts developing during the embryonic period with the neural progenitor cells generating neurons. Newborn neurons migrate along progenitors’ radial processes from the site of their origin in the germinal zones to the cortical plate, where they mature and integrate in the forming circuitry. Cell biological features of neural progenitors, such as the location and timing of their mitoses, together with their characteristic morphologies, can directly or indirectly regulate the abundance and the identity of their neuronal progeny. Alterations in the complex and delicate process of cerebral cortex development can lead to malformations of cortical development (MCDs). They include various structural abnormalities that affect the size, thickness and/or folding pattern of the developing cortex. Their clinical manifestations can entail a neurodevelopmental disorder, such as epilepsy, developmental delay, intellectual disability, or autism spectrum disorder. The recent advancements of molecular and neuroimaging techniques, along with the development of appropriate in vitro and in vivo model systems, have enabled the assessment of the genetic and environmental causes of MCDs. Here we broadly review the cell biological characteristics of neural progenitor cells and focus on those features whose perturbations have been linked to MCDs.
Collapse
|
91
|
Makhdoom EUH, Anwar H, Baig SM, Hussain G. Whole exome sequencing identifies a novel mutation in ASPM and ultra-rare mutation in CDK5RAP2 causing Primary microcephaly in consanguineous Pakistani families. Pak J Med Sci 2022; 38:84-89. [PMID: 35035405 PMCID: PMC8713189 DOI: 10.12669/pjms.38.1.4464] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/29/2021] [Accepted: 10/29/2021] [Indexed: 12/04/2022] Open
Abstract
Background & Objectives: Primary Microcephaly (MCPH) is a rare neurogenetic disease, manifesting congenitally reduced head circumference and non-progressive intellectual disability (ID). To date, twenty-eight genes with biallelic mutations have been reported for this disorder. The study aimed for molecular genetic characterization of Pakistani families segregating MCPH. Methods: We studied two unrelated consanguineous families (family A and B) presenting >2 patients with diagnostic symptoms of MCPH, born to asymptomatic parents. We employed whole-exome sequencing (WES) of probands to find putative causal mutations. The candidate variants were further confirmed and analyzed for co-segregation by Sanger sequencing of all available members of each family. This study was conducted at Government College University, Faisalabad, Pakistan, and Cologne Center for Genomics (CCG), University of Cologne, Germany; during 2017-2020. Results: We identified a novel homozygous variant c.10097_10098delGA, p.(Gly3366Glufs*19) in exon 26 of ASPM gene in family A which presents with moderate intellectual disability, speech impairment, visual abnormalities, seizures, and ptyalism. Family B was found to segregate nonsense, homozygous variant c.448C>T p.(Arg150*) in CDK5RAP2. The patients also exhibited mild to severe seizures without ptyalism that has not been previously reported in patients with mutations in the CDK5RAP2 gene. Conclusion: We report a novel mutation in ASPM and ultra-rare mutation in the CDK5RAP2 gene, both causing primary microcephaly. The study expands the mutational spectrum of the ASPM gene to 212, and also adds to the clinical spectrum of CDK5RAP2 mutations. It also demonstrated the utility of WES in the investigation and genetic diagnosis of genetically heterogeneous disorders like MCPH. These findings would aid in diagnostic and preventive strategies including carrier screening, cascade testing, and genetic counselling.
Collapse
Affiliation(s)
- Ehtisham Ul Haq Makhdoom
- Ehtisham ul Haq Makhdoom (MPhil), Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, 38000, Faisalabad, Pakistan. Human Molecular Genetics Laboratory, Health Biotechnology Division, NIBGE College, PIEAS, 38000, Faisalabad, Pakistan
| | - Haseeb Anwar
- Haseeb Anwar (PhD), Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, 38000, Faisalabad, Pakistan
| | - Shahid Mahmood Baig
- Shahid Mahmood Baig (PhD), Department of Biological and Biomedical Sciences, The Aga Khan University, 74000, Karachi, Pakistan. Pakistan Science Foundation, Constitution Avenue, 44000, Islamabad, Pakistan. Human Molecular Genetics Laboratory, Health Biotechnology Division, NIBGE College, PIEAS, 38000, Faisalabad, Pakistan
| | - Ghulam Hussain
- Ghulam Hussain (PhD), Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, 38000, Faisalabad, Pakistan
| |
Collapse
|
92
|
Chinnappa K, Cárdenas A, Prieto-Colomina A, Villalba A, Márquez-Galera Á, Soler R, Nomura Y, Llorens E, Tomasello U, López-Atalaya JP, Borrell V. Secondary loss of miR-3607 reduced cortical progenitor amplification during rodent evolution. SCIENCE ADVANCES 2022; 8:eabj4010. [PMID: 35020425 PMCID: PMC8754304 DOI: 10.1126/sciadv.abj4010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The evolutionary expansion and folding of the mammalian cerebral cortex resulted from amplification of progenitor cells during embryonic development. This process was reversed in the rodent lineage after splitting from primates, leading to smaller and smooth brains. Genetic mechanisms underlying this secondary loss in rodent evolution remain unknown. We show that microRNA miR-3607 is expressed embryonically in the large cortex of primates and ferret, distant from the primate-rodent lineage, but not in mouse. Experimental expression of miR-3607 in embryonic mouse cortex led to increased Wnt/β-catenin signaling, amplification of radial glia cells (RGCs), and expansion of the ventricular zone (VZ), via blocking the β-catenin inhibitor APC (adenomatous polyposis coli). Accordingly, loss of endogenous miR-3607 in ferret reduced RGC proliferation, while overexpression in human cerebral organoids promoted VZ expansion. Our results identify a gene selected for secondary loss during mammalian evolution to limit RGC amplification and, potentially, cortex size in rodents.
Collapse
|
93
|
Cortical morphology and illness insight in patients with schizophrenia. Eur Arch Psychiatry Clin Neurosci 2022; 272:985-995. [PMID: 34518921 PMCID: PMC9388450 DOI: 10.1007/s00406-021-01328-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/27/2021] [Indexed: 11/07/2022]
Abstract
Insight into illness in schizophrenia (SZ) patients has a major impact on treatment adherence and outcome. Previous studies have linked distinct deviations of brain structure to illness insight, specifically in frontoparietal and subcortical regions. Some of these abnormalities are thought to reflect aberrant cortical development. In this study, we used cross-sectional data to examine associations between illness insight and two cortical surface markers that are known to follow distinct neurodevelopmental trajectories, i.e. cortical gyrification (CG) and thickness (CT). CG and CT was investigated in SZ patients (n = 82) and healthy controls (HC, n = 48) using 3 T structural magnetic resonance imaging. Illness insight in SZ patients was measured using the OSSTI scale, an instrument that provides information on two distinct dimensions of illness insight, i.e. treatment adherence (OSSTI-A) and identification of disease-related symptoms (OSSTI-I). CT and CG were computed using the Computational Anatomy Toolbox (CAT12). Whole-brain and regions-of-interest (ROI)-based analyses were performed. SZ patients showed higher CG in anterior cingulate, superior frontal and temporal gyrus and reduced CG in insular and superior frontal cortex when compared to HC. SZ patients showed decreased CT in pre- and paracentral, occipital, cingulate, frontoparietal and temporal regions. Illness insight in SZ patients was significantly associated with both CG and CT in the left inferior parietal lobule (OSSTI-A) and the right precentral gyrus (CG/OSSTI-A, CT/OSSTI-I). The data support a multi-parametric neuronal model with both pre- and postnatal brain developmental factors having an impact on illness insight in patients with SZ.
Collapse
|
94
|
Melliou S, Sangster KT, Djuric U, Diamandis P. The promise of organoids for unraveling the proteomic landscape of the developing human brain. Mol Psychiatry 2022; 27:73-80. [PMID: 34703024 DOI: 10.1038/s41380-021-01354-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022]
Abstract
Cerebral organoids offer an opportunity to bioengineer experimental avatars of the developing human brain and have already begun garnering relevant insights into complex neurobiological processes and disease. Thus far, investigations into their heterogeneous cellular composition and developmental trajectories have been largely limited to transcriptional readouts. Recent advances in global proteomic technologies have enabled a new range of techniques to explore dynamic and non-overlapping spatiotemporal protein-level programs operational in these humanoid neural structures. Here we discuss these early protein-based studies and their potentially essential role for unraveling critical secreted paracrine signals, processes with poor proteogenomic correlations, or neurodevelopmental proteins requiring post-translational modification for biological activity. Integrating emerging proteomic tools with these faithful human-derived neurodevelopmental models could transform our understanding of complex neural cell phenotypes and neurobiological processes, not exclusively driven by transcriptional regulation. These insights, less accessible by exclusive RNA-based approaches, could reveal new knowledge into human brain development and guide improvements in neural regenerative medicine efforts.
Collapse
Affiliation(s)
- Sofia Melliou
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Kevin T Sangster
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Ugljesa Djuric
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Phedias Diamandis
- Princess Margaret Cancer Centre, 101 College Street, Toronto, ON, M5G 1L7, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Laboratory Medicine Program, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
95
|
van der Meer D, Kaufmann T, Shadrin AA, Makowski C, Frei O, Roelfs D, Monereo-Sánchez J, Linden DEJ, Rokicki J, Alnæs D, de Leeuw C, Thompson WK, Loughnan R, Fan CC, Westlye LT, Andreassen OA, Dale AM. The genetic architecture of human cortical folding. SCIENCE ADVANCES 2021; 7:eabj9446. [PMID: 34910505 PMCID: PMC8673767 DOI: 10.1126/sciadv.abj9446] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/27/2021] [Indexed: 05/04/2023]
Abstract
The folding of the human cerebral cortex is a highly genetically regulated process that allows for a much larger surface area to fit into the cranial vault and optimizes functional organization. Sulcal depth is a robust yet understudied measure of localized folding, previously associated with multiple neurodevelopmental disorders. Here, we report the first genome-wide association study of sulcal depth. Through the multivariate omnibus statistical test (MOSTest) applied to vertex-wise measures from 33,748 U.K. Biobank participants (mean age, 64.3 years; 52.0% female), we identified 856 genome-wide significant loci (P < 5 × 10−8). Comparisons with cortical thickness and surface area indicated that sulcal depth has higher locus yield, heritability, and effective sample size. There was a large amount of genetic overlap between these traits, with gene-based analyses indicating strong associations with neurodevelopmental processes. Our findings demonstrate sulcal depth is a promising neuroimaging phenotype that may enhance our understanding of cortical morphology.
Collapse
Affiliation(s)
- Dennis van der Meer
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Tobias Kaufmann
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Alexey A. Shadrin
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Carolina Makowski
- Center for Multimodal Imaging and Genetics, University of California at San Diego, La Jolla, CA 92037, USA
| | - Oleksandr Frei
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Daniel Roelfs
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jennifer Monereo-Sánchez
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - David E. J. Linden
- School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Jaroslav Rokicki
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Dag Alnæs
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Christiaan de Leeuw
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Wesley K. Thompson
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, La Jolla, CA 92037, USA
| | - Robert Loughnan
- Center for Multimodal Imaging and Genetics, University of California at San Diego, La Jolla, CA 92037, USA
| | - Chun Chieh Fan
- Center for Multimodal Imaging and Genetics, University of California at San Diego, La Jolla, CA 92037, USA
| | - Lars T. Westlye
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Anders M. Dale
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Center for Multimodal Imaging and Genetics, University of California at San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
96
|
He Z, Du L, Huang Y, Jiang X, Lv J, Guo L, Zhang S, Zhang T. Gyral Hinges Account for the Highest Cost and the Highest Communication Capacity in a Corticocortical Network. Cereb Cortex 2021; 32:3359-3376. [PMID: 34875041 DOI: 10.1093/cercor/bhab420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 12/11/2022] Open
Abstract
Prior studies reported the global structure of brain networks exhibits the "small-world" and "rich-world" attributes. However, the underlying structural and functional architecture highlighted by these graph theory findings hasn't been explicitly related to the morphology of the cortex. This could be attributed to the lower resolution of used folding patterns, such as gyro-sulcal patterns. By defining a novel gyral folding pattern, termed gyral hinge (GH), which is the conjunction of ordinary gyri from multiple directions, we found GHs possess the highest length and cost in the white matter fiber connective network, and the shortest paths in the network tend to travel through GHs in their middle part. Based on these findings, we would hypothesize GHs could reside in the centers of a network core, thereby accounting for the highest cost and the highest communication capacity in a corticocortical network. The following results further support our hypothesis: 1) GHs possess stronger functional network integration capacity. 2) Higher cost is found on the connection with GHs to hinges and GHs to GHs. 3) Moving GHs introduces higher extra network cost. Our findings and hypotheses could reveal a profound relationship among the cortical folding patterns, axonal wiring architectures, and brain functions.
Collapse
Affiliation(s)
- Zhibin He
- School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Lei Du
- School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ying Huang
- School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Xi Jiang
- School of Life Science and Technology, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jinglei Lv
- School of Biomedical Engineering, Sydney Imaging, Brain and Mind Centre, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Lei Guo
- School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shu Zhang
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tuo Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
97
|
Wilsch-Bräuninger M, Huttner WB. Primary Cilia and Centrosomes in Neocortex Development. Front Neurosci 2021; 15:755867. [PMID: 34744618 PMCID: PMC8566538 DOI: 10.3389/fnins.2021.755867] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
During mammalian brain development, neural stem and progenitor cells generate the neurons for the six-layered neocortex. The proliferative capacity of the different types of progenitor cells within the germinal zones of the developing neocortex is a major determinant for the number of neurons generated. Furthermore, the various modes of progenitor cell divisions, for which the orientation of the mitotic spindle of progenitor cells has a pivotal role, are a key parameter to ensure the appropriate size and proper cytoarchitecture of the neocortex. Here, we review the roles of primary cilia and centrosomes of progenitor cells in these processes during neocortical development. We specifically focus on the apical progenitor cells in the ventricular zone. In particular, we address the alternating, dual role of the mother centriole (i) as a component of one of the spindle poles during mitosis, and (ii) as the basal body of the primary cilium in interphase, which is pivotal for the fate of apical progenitor cells and their proliferative capacity. We also discuss the interactions of these organelles with the microtubule and actin cytoskeleton, and with junctional complexes. Centriolar appendages have a specific role in this interaction with the cell cortex and the plasma membrane. Another topic of this review is the specific molecular composition of the ciliary membrane and the membrane vesicle traffic to the primary cilium of apical progenitors, which underlie the ciliary signaling during neocortical development; this signaling itself, however, is not covered in depth here. We also discuss the recently emerging evidence regarding the composition and roles of primary cilia and centrosomes in basal progenitors, a class of progenitors thought to be of particular importance for neocortex expansion in development and evolution. While the tight interplay between primary cilia and centrosomes makes it difficult to allocate independent roles to either organelle, mutations in genes encoding ciliary and/or centrosome proteins indicate that both are necessary for the formation of a properly sized and functioning neocortex during development. Human neocortical malformations, like microcephaly, underpin the importance of primary cilia/centrosome-related processes in neocortical development and provide fundamental insight into the underlying mechanisms involved.
Collapse
Affiliation(s)
| | - Wieland B Huttner
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
98
|
Darayi M, Hoffman ME, Sayut J, Wang S, Demirci N, Consolini J, Holland MA. Computational models of cortical folding: A review of common approaches. J Biomech 2021; 139:110851. [PMID: 34802706 DOI: 10.1016/j.jbiomech.2021.110851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/09/2021] [Accepted: 10/26/2021] [Indexed: 11/29/2022]
Abstract
The process of gyrification, by which the brain develops the intricate pattern of gyral hills and sulcal valleys, is the result of interactions between biological and mechanical processes during brain development. Researchers have developed a vast array of computational models in order to investigate cortical folding. This review aims to summarize these studies, focusing on five essential elements of the brain that affect development and gyrification and how they are represented in computational models: (i) the constraints of skull, meninges, and cerebrospinal fluid; (ii) heterogeneity of cortical layers and regions; (iii) anisotropic behavior of subcortical fiber tracts; (iv) material properties of brain tissue; and (v) the complex geometry of the brain. Finally, we highlight areas of need for future simulations of brain development.
Collapse
Affiliation(s)
- Mohsen Darayi
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Mia E Hoffman
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - John Sayut
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shuolun Wang
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Nagehan Demirci
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jack Consolini
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Maria A Holland
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
99
|
Keuken MC, Alkemade A, Stevenson N, Innes RJ, Forstmann BU. Structure-function similarities in deep brain stimulation targets cross-species. Neurosci Biobehav Rev 2021; 131:1127-1135. [PMID: 34715147 DOI: 10.1016/j.neubiorev.2021.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 11/24/2022]
Abstract
Deep Brain Stimulation (DBS) is an effective neurosurgical treatment to alleviate motor symptoms of advanced Parkinson's disease. Due to its potential, DBS usage is rapidly expanding to target a large number of brain regions to treat a wide range of diseases and neuropsychiatric disorders. The identification and validation of new target regions heavily rely on the insights gained from rodent and primate models. Here we present a large-scale automatic meta-analysis in which the structure-function associations within and between species are compared for 21 DBS targets in humans. The results indicate that the structure-function association for the majority of the 21 included subcortical areas were conserved cross-species. A subset of structures showed overlapping functional association. This can potentially be attributed to shared brain networks and might explain why multiple brain areas are targeted for the same disease or neuropsychiatric disorder.
Collapse
Affiliation(s)
- Max C Keuken
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Nieuwe Achtergracht 129B, Postbus 15926, 1001 NK, Amsterdam, The Netherlands.
| | - Anneke Alkemade
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Nieuwe Achtergracht 129B, Postbus 15926, 1001 NK, Amsterdam, The Netherlands
| | - Niek Stevenson
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Nieuwe Achtergracht 129B, Postbus 15926, 1001 NK, Amsterdam, The Netherlands
| | - Reilly J Innes
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Nieuwe Achtergracht 129B, Postbus 15926, 1001 NK, Amsterdam, The Netherlands; Newcastle Cognition Lab, University of Newcastle, Callaghan, NSW, Australia
| | - Birte U Forstmann
- Integrative Model-based Cognitive Neuroscience Research Unit, University of Amsterdam, Nieuwe Achtergracht 129B, Postbus 15926, 1001 NK, Amsterdam, The Netherlands
| |
Collapse
|
100
|
High-coverage metabolomics uncovers microbiota-driven biochemical landscape of interorgan transport and gut-brain communication in mice. Nat Commun 2021; 12:6000. [PMID: 34667167 PMCID: PMC8526691 DOI: 10.1038/s41467-021-26209-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 09/06/2021] [Indexed: 01/09/2023] Open
Abstract
The mammalian gut harbors a complex and dynamic microbial ecosystem: the microbiota. While emerging studies support that microbiota regulates brain function with a few molecular cues suggested, the overall biochemical landscape of the “microbiota-gut-brain axis” remains largely unclear. Here we use high-coverage metabolomics to comparatively profile feces, blood sera, and cerebral cortical brain tissues of germ-free C57BL/6 mice and their age-matched conventionally raised counterparts. Results revealed for all three matrices metabolomic signatures owing to microbiota, yielding hundreds of identified metabolites including 533 altered for feces, 231 for sera, and 58 for brain with numerous significantly enriched pathways involving aromatic amino acids and neurotransmitters. Multicompartmental comparative analyses single out microbiota-derived metabolites potentially implicated in interorgan transport and the gut-brain axis, as exemplified by indoxyl sulfate and trimethylamine-N-oxide. Gender-specific characteristics of these landscapes are discussed. Our findings may be valuable for future research probing microbial influences on host metabolism and gut-brain communication. The gut microbiota harbours neuroactive potential with links to neurological disorders. Here, the authors apply global metabolomics with an integrated annotation strategy to comparatively profile fecal, blood serum and cerebral cortical brain tissues of eight-week-old germ-free mice vs. age-matched specific-pathogen-free mice, providing a snapshot of the metabolome status linked to the gut-brain axis.
Collapse
|