1
|
Luo Y, Liang H. Developmental-status-aware transcriptional decomposition establishes a cell state panorama of human cancers. Genome Med 2024; 16:124. [PMID: 39468667 PMCID: PMC11514945 DOI: 10.1186/s13073-024-01393-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cancer cells evolve under unique functional adaptations that unlock transcriptional programs embedded in adult stem and progenitor-like cells for progression, metastasis, and therapeutic resistance. However, it remains challenging to quantify the stemness-aware cell state of a tumor based on its gene expression profile. METHODS We develop a developmental-status-aware transcriptional decomposition strategy using single-cell RNA-sequencing-derived tissue-specific fetal and adult cell signatures as anchors. We apply our method to various biological contexts, including developing human organs, adult human tissues, experimentally induced differentiation cultures, and bulk human tumors, to benchmark its performance and to reveal novel biology of entangled developmental signaling in oncogenic processes. RESULTS Our strategy successfully captures complex dynamics in developmental tissue bulks, reveals remarkable cellular heterogeneity in adult tissues, and resolves the ambiguity of cell identities in in vitro transformations. Applying it to large patient cohorts of bulk RNA-seq, we identify clinically relevant cell-of-origin patterns and observe that decomposed fetal cell signals significantly increase in tumors versus normal tissues and metastases versus primary tumors. Across cancer types, the inferred fetal-state strength outperforms published stemness indices in predicting patient survival and confers substantially improved predictive power for therapeutic responses. CONCLUSIONS Our study not only provides a general approach to quantifying developmental-status-aware cell states of bulk samples but also constructs an information-rich, biologically interpretable, cell-state panorama of human cancers, enabling diverse translational applications.
Collapse
Affiliation(s)
- Yikai Luo
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Han Liang
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Jurcau MC, Jurcau A, Cristian A, Hogea VO, Diaconu RG, Nunkoo VS. Inflammaging and Brain Aging. Int J Mol Sci 2024; 25:10535. [PMID: 39408862 PMCID: PMC11476611 DOI: 10.3390/ijms251910535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Progress made by the medical community in increasing lifespans comes with the costs of increasing the incidence and prevalence of age-related diseases, neurodegenerative ones included. Aging is associated with a series of morphological changes at the tissue and cellular levels in the brain, as well as impairments in signaling pathways and gene transcription, which lead to synaptic dysfunction and cognitive decline. Although we are not able to pinpoint the exact differences between healthy aging and neurodegeneration, research increasingly highlights the involvement of neuroinflammation and chronic systemic inflammation (inflammaging) in the development of age-associated impairments via a series of pathogenic cascades, triggered by dysfunctions of the circadian clock, gut dysbiosis, immunosenescence, or impaired cholinergic signaling. In addition, gender differences in the susceptibility and course of neurodegeneration that appear to be mediated by glial cells emphasize the need for future research in this area and an individualized therapeutic approach. Although rejuvenation research is still in its very early infancy, accumulated knowledge on the various signaling pathways involved in promoting cellular senescence opens the perspective of interfering with these pathways and preventing or delaying senescence.
Collapse
Affiliation(s)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Vlad Octavian Hogea
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
3
|
Wilson S, Christiaens D, Yun H, Uus A, Cordero-Grande L, Karolis V, Price A, Deprez M, Tournier JD, Rutherford M, Grant E, Hajnal JV, Edwards AD, Arichi T, O'Muircheartaigh J, Im K. Dynamic changes in subplate and cortical plate microstructure at the onset of cortical folding in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.16.562524. [PMID: 38979235 PMCID: PMC11230247 DOI: 10.1101/2023.10.16.562524] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cortical gyrification takes place predominantly during the second to third trimester, alongside other fundamental developmental processes, such as the development of white matter connections, lamination of the cortex and formation of neural circuits. The mechanistic biology that drives the formation cortical folding patterns remains an open question in neuroscience. In our previous work, we modelled the in utero diffusion signal to quantify the maturation of microstructure in transient fetal compartments, identifying patterns of change in diffusion metrics that reflect critical neurobiological transitions occurring in the second to third trimester. In this work, we apply the same modelling approach to explore whether microstructural maturation of these compartments is correlated with the process of gyrification. We quantify the relationship between sulcal depth and tissue anisotropy within the cortical plate (CP) and underlying subplate (SP), key transient fetal compartments often implicated in mechanistic hypotheses about the onset of gyrification. Using in utero high angular resolution multi-shell diffusion-weighted imaging (HARDI) from the Developing Human Connectome Project (dHCP), our analysis reveals that the anisotropic, tissue component of the diffusion signal in the SP and CP decreases immediately prior to the formation of sulcal pits in the fetal brain. By back-projecting a map of folded brain regions onto the unfolded brain, we find evidence for cytoarchitectural differences between gyral and sulcal areas in the late second trimester, suggesting that regional variation in the microstructure of transient fetal compartments precedes, and thus may have a mechanistic function, in the onset of cortical folding in the developing human brain.
Collapse
Affiliation(s)
- Siân Wilson
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Daan Christiaens
- Department of Electrical Engineering, Katholieke Universiteit Leuven, Belgium
| | - Hyukjin Yun
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alena Uus
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
- Department of Biomedical Engineering, School Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, United Kingdom
| | | | - Vyacheslav Karolis
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
| | - Anthony Price
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
| | - Maria Deprez
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
- Department of Biomedical Engineering, School Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, United Kingdom
| | - Jacques-Donald Tournier
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
- Department of Biomedical Engineering, School Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, United Kingdom
| | - Mary Rutherford
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
| | - Ellen Grant
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph V Hajnal
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
- Department of Biomedical Engineering, School Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, United Kingdom
| | - A David Edwards
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
| | - Tomoki Arichi
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, United Kingdom
- Children's Neurosciences, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, United Kingdom
| | - Jonathan O'Muircheartaigh
- Research Department of Early Life Imaging, Kings College London, London, United Kingdom
- Department of Forensic and Neurodevelopmental Sciences, King's College London, United Kingdom
| | - Kiho Im
- Fetal-Neonatal Neuroimaging & Developmental Science Center, Boston Children's Hospital, Boston, Massachusetts, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Kwak T, Park SH, Lee S, Shin Y, Yoon KJ, Cho SW, Park JC, Yang SH, Cho H, Im HI, Ahn SJ, Sun W, Yang JH. Guidelines for Manufacturing and Application of Organoids: Brain. Int J Stem Cells 2024; 17:158-181. [PMID: 38777830 PMCID: PMC11170118 DOI: 10.15283/ijsc24056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
This study offers a comprehensive overview of brain organoids for researchers. It combines expert opinions with technical summaries on organoid definitions, characteristics, culture methods, and quality control. This approach aims to enhance the utilization of brain organoids in research. Brain organoids, as three-dimensional human cell models mimicking the nervous system, hold immense promise for studying the human brain. They offer advantages over traditional methods, replicating anatomical structures, physiological features, and complex neuronal networks. Additionally, brain organoids can model nervous system development and interactions between cell types and the microenvironment. By providing a foundation for utilizing the most human-relevant tissue models, this work empowers researchers to overcome limitations of two-dimensional cultures and conduct advanced disease modeling research.
Collapse
Affiliation(s)
| | - Si-Hyung Park
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | | | | | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
- Organoid Standards Initiative
| | - Seung-Woo Cho
- Organoid Standards Initiative
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Jong-Chan Park
- Organoid Standards Initiative
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Seung-Ho Yang
- Organoid Standards Initiative
- Department of Neurosurgery, St. Vincent’s Hospital, The Catholic University of Korea, Suwon, Korea
| | - Heeyeong Cho
- Organoid Standards Initiative
- Center for Rare Disease Therapeutic Technology, Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Heh-In Im
- Organoid Standards Initiative
- Behavioral and Molecular Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Sun-Ju Ahn
- Organoid Standards Initiative
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
- Organoid Standards Initiative
| | - Ji Hun Yang
- Next & Bio Inc., Seoul, Korea
- Organoid Standards Initiative
| |
Collapse
|
5
|
Dixit T. A synthesis of coevolution across levels of biological organization. Evolution 2024; 78:211-220. [PMID: 38085659 DOI: 10.1093/evolut/qpad082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/16/2023] [Accepted: 04/28/2023] [Indexed: 02/03/2024]
Abstract
In evolutionary ecology, coevolution is typically defined as reciprocal evolution of interacting species. However, outside the context of interacting species, the term "coevolution" is also used at levels of biological organization within species (e.g., between males and females, between cells, and between genes or proteins). Furthermore, although evolution is typically defined as "genetic change over time", coevolution need not involve genetic changes in the interacting parties, since cultures can also evolve. In this review, I propose that coevolution be defined more broadly as "reciprocal adaptive evolution at any level of biological organisation". The classification of reciprocal evolution at all levels of biological organization as coevolution would maintain consistency in terminology. More importantly, the broader definition should facilitate greater integration of coevolution research across disciplines. For example, principles usually discussed only in the context of coevolution between species or coevolution between genes (e.g., tight and diffuse coevolution, and compensatory coevolution, respectively) could be more readily applied to new fields. The application of coevolutionary principles to new contexts could also provide benefits to society, for instance in deducing the dynamics of coevolution between cancer cells and cells of the human immune system.
Collapse
Affiliation(s)
- Tanmay Dixit
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
- DST-NRF Centre of Excellence at the FitzPatrick Institute of African Ornithology, University of Cape Town, Rondebosch, Cape Town, South Africa
| |
Collapse
|
6
|
Royall LN, Machado D, Jessberger S, Denoth-Lippuner A. Asymmetric inheritance of centrosomes maintains stem cell properties in human neural progenitor cells. eLife 2023; 12:e83157. [PMID: 37882444 PMCID: PMC10629821 DOI: 10.7554/elife.83157] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/25/2023] [Indexed: 10/27/2023] Open
Abstract
During human forebrain development, neural progenitor cells (NPCs) in the ventricular zone (VZ) undergo asymmetric cell divisions to produce a self-renewed progenitor cell, maintaining the potential to go through additional rounds of cell divisions, and differentiating daughter cells, populating the developing cortex. Previous work in the embryonic rodent brain suggested that the preferential inheritance of the pre-existing (older) centrosome to the self-renewed progenitor cell is required to maintain stem cell properties, ensuring proper neurogenesis. If asymmetric segregation of centrosomes occurs in NPCs of the developing human brain, which depends on unique molecular regulators and species-specific cellular composition, remains unknown. Using a novel, recombination-induced tag exchange-based genetic tool to birthdate and track the segregation of centrosomes over multiple cell divisions in human embryonic stem cell-derived regionalised forebrain organoids, we show the preferential inheritance of the older mother centrosome towards self-renewed NPCs. Aberration of asymmetric segregation of centrosomes by genetic manipulation of the centrosomal, microtubule-associated protein Ninein alters fate decisions of NPCs and their maintenance in the VZ of human cortical organoids. Thus, the data described here use a novel genetic approach to birthdate centrosomes in human cells and identify asymmetric inheritance of centrosomes as a mechanism to maintain self-renewal properties and to ensure proper neurogenesis in human NPCs.
Collapse
Affiliation(s)
- Lars N Royall
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of ZurichZurichSwitzerland
| | - Diana Machado
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of ZurichZurichSwitzerland
| | - Sebastian Jessberger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of ZurichZurichSwitzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning (AdaBD), University of ZurichZurichSwitzerland
| | - Annina Denoth-Lippuner
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of ZurichZurichSwitzerland
| |
Collapse
|
7
|
Chavoshnejad P, Vallejo L, Zhang S, Guo Y, Dai W, Zhang T, Razavi MJ. Mechanical hierarchy in the formation and modulation of cortical folding patterns. Sci Rep 2023; 13:13177. [PMID: 37580340 PMCID: PMC10425471 DOI: 10.1038/s41598-023-40086-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023] Open
Abstract
The important mechanical parameters and their hierarchy in the growth and folding of the human brain have not been thoroughly understood. In this study, we developed a multiscale mechanical model to investigate how the interplay between initial geometrical undulations, differential tangential growth in the cortical plate, and axonal connectivity form and regulate the folding patterns of the human brain in a hierarchical order. To do so, different growth scenarios with bilayer spherical models that features initial undulations on the cortex and uniform or heterogeneous distribution of axonal fibers in the white matter were developed, statistically analyzed, and validated by the imaging observations. The results showed that the differential tangential growth is the inducer of cortical folding, and in a hierarchal order, high-amplitude initial undulations on the surface and axonal fibers in the substrate regulate the folding patterns and determine the location of gyri and sulci. The locations with dense axonal fibers after folding settle in gyri rather than sulci. The statistical results also indicated that there is a strong correlation between the location of positive (outward) and negative (inward) initial undulations and the locations of gyri and sulci after folding, respectively. In addition, the locations of 3-hinge gyral folds are strongly correlated with the initial positive undulations and locations of dense axonal fibers. As another finding, it was revealed that there is a correlation between the density of axonal fibers and local gyrification index, which has been observed in imaging studies but not yet fundamentally explained. This study is the first step in understanding the linkage between abnormal gyrification (surface morphology) and disruption in connectivity that has been observed in some brain disorders such as Autism Spectrum Disorder. Moreover, the findings of the study directly contribute to the concept of the regularity and variability of folding patterns in individual human brains.
Collapse
Affiliation(s)
- Poorya Chavoshnejad
- Department of Mechanical Engineering, Binghamton University, Binghamton, NY, 13902, USA
| | - Liam Vallejo
- Department of Mechanical Engineering, Binghamton University, Binghamton, NY, 13902, USA
| | - Songyao Zhang
- Brain Decoding Research Center and School of Automation, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Yanchen Guo
- Department of Computer Science, Binghamton University, Binghamton, NY, USA
| | - Weiying Dai
- Department of Computer Science, Binghamton University, Binghamton, NY, USA
| | - Tuo Zhang
- Brain Decoding Research Center and School of Automation, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Mir Jalil Razavi
- Department of Mechanical Engineering, Binghamton University, Binghamton, NY, 13902, USA.
| |
Collapse
|
8
|
Kim H, Lee K, Shim YM, Kim EE, Kim SK, Phi JH, Park CK, Choi SH, Park SH. Epigenetic Alteration of H3K27me3 as a Possible Oncogenic Mechanism of Central Neurocytoma. J Transl Med 2023; 103:100159. [PMID: 37088465 DOI: 10.1016/j.labinv.2023.100159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/25/2023] Open
Abstract
Central neurocytoma (CN) is a low-grade neuronal tumor that mainly arises from the lateral ventricle (LV). This tumor remains poorly understood in the sense that no driver gene aberrations have been identified thus far. We investigated immunomarkers in fetal and adult brains and 45 supratentorial periventricular tumors to characterize the biomarkers, cell of origin, and tumorigenesis of CN. All CNs occurred in the LV. A minority involved the third ventricle, but none involved the fourth ventricle. As expected, next-generation sequencing performed using a brain-tumor-targeted gene panel in 7 CNs and whole exome sequencing in 5 CNs showed no driver mutations. Immunohistochemically, CNs were robustly positive for FGFR3 (100%), SSTR2 (92%), TTF-1 (Nkx2.1) (88%), GLUT-1 (84%), and L1CAM (76%), in addition to the well-known markers of CN, synaptophysin (100%) and NeuN (96%). TTF-1 was also positive in subependymal giant cell astrocytomas (100%, 5/5) and the pituicyte tumor family, including pituicytoma and spindle cell oncocytoma (100%, 5/5). Interestingly, 1 case of LV subependymoma (20%, 1/5) was positive for TTF-1, but all LV ependymomas were negative (0/5 positive). Because TTF-1-positive cells were detected in the medial ganglionic eminence around the foramen of Monro of the fetal brain and in the subventricular zone of the LV of the adult brain, CN may arise from subventricular TTF-1-positive cells undergoing neuronal differentiation. H3K27me3 loss was observed in all CNs and one case (20%) of LV subependymoma, suggesting that chromatin remodeling complexes or epigenetic alterations may be involved in the tumorigenesis of all CNs and some ST-subependymomas. Further studies are required to determine the exact tumorigenic mechanism of CN.
Collapse
Affiliation(s)
- Hyunhee Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwanghoon Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu-Mi Shim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eric Eunshik Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ki Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Hoon Phi
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Fernández V, Borrell V. Developmental mechanisms of gyrification. Curr Opin Neurobiol 2023; 80:102711. [DOI: 10.1016/j.conb.2023.102711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 03/31/2023]
|
10
|
Morales A, Andrews MG. Approaches to investigating metabolism in human neurodevelopment using organoids: insights from intestinal and cancer studies. Development 2022; 149:dev200506. [PMID: 36255366 PMCID: PMC9720749 DOI: 10.1242/dev.200506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Interrogating the impact of metabolism during development is important for understanding cellular and tissue formation, organ and systemic homeostasis, and dysregulation in disease states. To evaluate the vital functions metabolism coordinates during human brain development and disease, pluripotent stem cell-derived models, such as organoids, provide tractable access to neurodevelopmental processes. Despite many strengths of neural organoid models, the extent of their replication of endogenous metabolic programs is currently unclear and requires direct investigation. Studies in intestinal and cancer organoids that functionally evaluate dynamic bioenergetic changes provide a framework that can be adapted for the study of neural metabolism. Validation of in vitro models remains a significant challenge; investigation using in vivo models and primary tissue samples is required to improve our in vitro model systems and, concomitantly, improve our understanding of human development.
Collapse
Affiliation(s)
- Alexandria Morales
- Schoolof Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA
- Biomedical Engineering Graduate Program, Arizona State University, Tempe, AZ 85281, USA
| | - Madeline G. Andrews
- Schoolof Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
11
|
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
|
12
|
Whiteley JT, Fernandes S, Sharma A, Mendes APD, Racha V, Benassi SK, Marchetto MC. Reaching into the toolbox: Stem cell models to study neuropsychiatric disorders. Stem Cell Reports 2022; 17:187-210. [PMID: 35063127 PMCID: PMC8828548 DOI: 10.1016/j.stemcr.2021.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Recent advances in genetics, molecular biology, and stem cell biology have accelerated our understanding of neuropsychiatric disorders, like autism spectrum disorder (ASD), major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia (SZ). This progress highlights the incredible complexity of both the human brain and mental illnesses from the biochemical to the cellular level. Contributing to the complexity of neuropsychiatric disorders are their polygenic nature, cellular and brain region interconnectivity, and dysregulation of human-specific neurodevelopmental processes. Here, we discuss available tools, including CRISPR-Cas9, and the applications of these tools to develop cell-based two-dimensional (2D) models and 3D brain organoid models that better represent and unravel the intricacies of neuropsychiatric disorder pathophysiology.
Collapse
Affiliation(s)
- Jack T Whiteley
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Doctoral Program in Neurobiology and Behavior, Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, L7-028, MC 9872, New York, NY 10027, USA
| | - Sarah Fernandes
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Biological Sciences, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Amandeep Sharma
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ana Paula D Mendes
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Vipula Racha
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Simone K Benassi
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maria C Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Anthropology, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA.
| |
Collapse
|
13
|
Prodromidou K, Matsas R. Evolving features of human cortical development and the emerging roles of non-coding RNAs in neural progenitor cell diversity and function. Cell Mol Life Sci 2021; 79:56. [PMID: 34921638 PMCID: PMC11071749 DOI: 10.1007/s00018-021-04063-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 10/19/2022]
Abstract
The human cerebral cortex is a uniquely complex structure encompassing an unparalleled diversity of neuronal types and subtypes. These arise during development through a series of evolutionary conserved processes, such as progenitor cell proliferation, migration and differentiation, incorporating human-associated adaptations including a protracted neurogenesis and the emergence of novel highly heterogeneous progenitor populations. Disentangling the unique features of human cortical development involves elucidation of the intricate developmental cell transitions orchestrated by progressive molecular events. Crucially, developmental timing controls the fine balance between cell cycle progression/exit and the neurogenic competence of precursor cells, which undergo morphological transitions coupled to transcriptome-defined temporal states. Recent advances in bulk and single-cell transcriptomic technologies suggest that alongside protein-coding genes, non-coding RNAs exert important regulatory roles in these processes. Interestingly, a considerable number of novel long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have appeared in human and non-human primates suggesting an evolutionary role in shaping cortical development. Here, we present an overview of human cortical development and highlight the marked diversification and complexity of human neuronal progenitors. We further discuss how lncRNAs and miRNAs constitute critical components of the extended epigenetic regulatory network defining intermediate states of progenitors and controlling cell cycle dynamics and fate choices with spatiotemporal precision, during human neurodevelopment.
Collapse
Affiliation(s)
- Kanella Prodromidou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521, Athens, Greece.
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521, Athens, Greece
| |
Collapse
|
14
|
Abstract
The human brain is characterized by the large size and intricate folding of its cerebral cortex, which are fundamental for our higher cognitive function and frequently altered in pathological dysfunction. Cortex folding is not unique to humans, nor even to primates, but is common across mammals. Cortical growth and folding are the result of complex developmental processes that involve neural stem and progenitor cells and their cellular lineages, the migration and differentiation of neurons, and the genetic programs that regulate and fine-tune these processes. All these factors combined generate mechanical stress and strain on the developing neural tissue, which ultimately drives orderly cortical deformation and folding. In this review we examine and summarize the current knowledge on the molecular, cellular, histogenic and mechanical mechanisms that are involved in and influence folding of the cerebral cortex, and how they emerged and changed during mammalian evolution. We discuss the main types of pathological malformations of human cortex folding, their specific developmental origin, and how investigating their genetic causes has illuminated our understanding of key events involved. We close our review by presenting the state-of-the-art animal and in vitro models of cortex folding that are currently used to study these devastating developmental brain disorders in children, and what are the main challenges that remain ahead of us to fully understand brain folding.
Collapse
Affiliation(s)
- Lucia Del Valle Anton
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| | - Victor Borrell
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| |
Collapse
|
15
|
Akeret K, Vasella F, Staartjes VE, Velz J, Müller T, Neidert MC, Weller M, Regli L, Serra C, Krayenbühl N. Anatomical phenotyping and staging of brain tumours. Brain 2021; 145:1162-1176. [PMID: 34554211 DOI: 10.1093/brain/awab352] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/25/2021] [Accepted: 08/21/2021] [Indexed: 11/14/2022] Open
Abstract
Unlike other tumors, the anatomical extent of brain tumors is not objectified and quantified through staging. Staging systems are based on understanding the anatomical sequence of tumor progression and its relationship to histopathological dedifferentiation and survival. The aim of this study was to describe the spatiotemporal phenotype of the most frequent brain tumor entities, to assess the association of anatomical tumor features with survival probability and to develop a staging system for WHO grade 2 and 3 gliomas and glioblastoma. Anatomical phenotyping was performed on a consecutive cohort of 1000 patients with first diagnosis of a primary or secondary brain tumor. Tumor probability in different topographic, phylogenetic and ontogenetic parcellation units was assessed on preoperative MRI through normalization of the relative tumor prevalence to the relative volume of the respective structure. We analyzed the spatiotemporal tumor dynamics by cross-referencing preoperative against preceding and subsequent MRIs of the respective patient. The association between anatomical phenotype and outcome defined prognostically critical anatomical tumor features at diagnosis. Based on a hypothesized sequence of anatomical tumor progression, we developed a three-level staging system for WHO grade 2 and 3 gliomas and glioblastoma. This staging system was validated internally in the original cohort and externally in an independent cohort of 300 consecutive patients. While primary central nervous system lymphoma showed highest probability along white matter tracts, metastases enriched along terminal arterial flow areas. Neuroepithelial tumors mapped along all sectors of the ventriculocortical axis, while adjacent units were spared, consistent with a transpallial behavior within phylo-ontogenetic radial units. Their topographic pattern correlated with morphogenetic processes of convergence and divergence of radial units during phylo- and ontogenesis. While a ventriculofugal growth dominated in neuroepithelial tumors, a gradual deviation from this neuroepithelial spatiotemporal behavior was found with progressive histopathological dedifferentiation. The proposed three-level staging system for WHO grade 2 and 3 gliomas and glioblastoma correlated with the degree of histological dedifferentiation and proved accurate in terms of survival upon both internal and external validation. In conclusion, this study identified specific spatiotemporal phenotypes in brain tumors through topographic probability and growth pattern assessment. The association of anatomical tumor features with survival defined critical steps in the anatomical sequence of neuroepithelial tumor progression, based on which a staging system for WHO grade 2 and 3 gliomas and glioblastoma was developed and validated.
Collapse
Affiliation(s)
- Kevin Akeret
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Flavio Vasella
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland.,Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Victor E Staartjes
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Julia Velz
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Timothy Müller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Marian Christoph Neidert
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Carlo Serra
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Niklaus Krayenbühl
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland.,Division of Pediatric Neurosurgery, University Children's Hospital, 8032 Zurich, Switzerland
| |
Collapse
|
16
|
Agboola OS, Hu X, Shan Z, Wu Y, Lei L. Brain organoid: a 3D technology for investigating cellular composition and interactions in human neurological development and disease models in vitro. Stem Cell Res Ther 2021; 12:430. [PMID: 34332630 PMCID: PMC8325286 DOI: 10.1186/s13287-021-02369-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/03/2021] [Indexed: 01/01/2023] Open
Abstract
Abstract The study of human brain physiology, including cellular interactions in normal and disease conditions, has been a challenge due to its complexity and unavailability. Induced pluripotent stem cell (iPSC) study is indispensable in the study of the pathophysiology of neurological disorders. Nevertheless, monolayer systems lack the cytoarchitecture necessary for cellular interactions and neurological disease modeling. Brain organoids generated from human pluripotent stem cells supply an ideal environment to model both cellular interactions and pathophysiology of the human brain. This review article discusses the composition and interactions among neural lineage and non-central nervous system cell types in brain organoids, current studies, and future perspectives in brain organoid research. Ultimately, the promise of brain organoids is to unveil previously inaccessible features of neurobiology that emerge from complex cellular interactions and to improve our mechanistic understanding of neural development and diseases. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02369-8.
Collapse
Affiliation(s)
- Oluwafemi Solomon Agboola
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China
| | - Xinglin Hu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China
| | - Zhiyan Shan
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China
| | - Yanshuang Wu
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China.
| | - Lei Lei
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, 194 Xuefu Rd, Nangang District, Heilongjiang Province, Harbin, 150081, People's Republic of China. .,Key Laboratory of Preservative of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China.
| |
Collapse
|
17
|
Schnabl J, Litz MPH, Schneider C, PenkoffLidbeck N, Bashiruddin S, Schwartz MS, Alligood K, Devoto SH, Barresi MJF. Characterizing the diverse cells that associate with the developing commissures of the zebrafish forebrain. Dev Neurobiol 2021; 81:671-695. [PMID: 33314626 DOI: 10.1002/dneu.22801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/20/2020] [Accepted: 12/08/2020] [Indexed: 01/02/2023]
Abstract
During embryonic development of bilaterally symmetrical organisms, neurons send axons across the midline at specific points to connect the two halves of the nervous system with a commissure. Little is known about the cells at the midline that facilitate this tightly regulated process. We exploit the conserved process of vertebrate embryonic development in the zebrafish model system to elucidate the identity of cells at the midline that may facilitate postoptic (POC) and anterior commissure (AC) development. We have discovered that three different gfap+ astroglial cell morphologies persist in contact with pathfinding axons throughout commissure formation. Similarly, olig2+ progenitor cells occupy delineated portions of the postoptic and anterior commissures where they act as multipotent, neural progenitors. Moreover, we conclude that both gfap+ and olig2+ progenitor cells give rise to neuronal populations in both the telencephalon and diencephalon; however, these varied cell populations showed significant developmental timing differences between the telencephalon and diencephalon. Lastly, we also showed that fli1a+ mesenchymal cells migrate along the presumptive commissure regions before and during midline axon crossing. Furthermore, following commissure maturation, specific blood vessels formed at the midline of the POC and immediately ventral and parallel to the AC. This comprehensive account of the cellular populations that correlate with the timing and position of commissural axon pathfinding has supported the conceptual modeling and identification of the early forebrain architecture that may be necessary for proper commissure development.
Collapse
Affiliation(s)
- Jake Schnabl
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Mackenzie P H Litz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caitlin Schneider
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,McGill University, Montreal, QC, Canada
| | | | - Sarah Bashiruddin
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Family Medicine Assoc, Westfield, MA, USA
| | - Morgan S Schwartz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kristin Alligood
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Farmers Conservation Alliance, Hood River, OR, USA
| | | | - Michael J F Barresi
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA.,Department of Biological Sciences, Smith College, Northampton, MA, USA
| |
Collapse
|
18
|
Jiang X, Zhang T, Zhang S, Kendrick KM, Liu T. Fundamental functional differences between gyri and sulci: implications for brain function, cognition, and behavior. PSYCHORADIOLOGY 2021; 1:23-41. [PMID: 38665307 PMCID: PMC10939337 DOI: 10.1093/psyrad/kkab002] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/24/2021] [Accepted: 02/02/2021] [Indexed: 04/28/2024]
Abstract
Folding of the cerebral cortex is a prominent characteristic of mammalian brains. Alterations or deficits in cortical folding are strongly correlated with abnormal brain function, cognition, and behavior. Therefore, a precise mapping between the anatomy and function of the brain is critical to our understanding of the mechanisms of brain structural architecture in both health and diseases. Gyri and sulci, the standard nomenclature for cortical anatomy, serve as building blocks to make up complex folding patterns, providing a window to decipher cortical anatomy and its relation with brain functions. Huge efforts have been devoted to this research topic from a variety of disciplines including genetics, cell biology, anatomy, neuroimaging, and neurology, as well as involving computational approaches based on machine learning and artificial intelligence algorithms. However, despite increasing progress, our understanding of the functional anatomy of gyro-sulcal patterns is still in its infancy. In this review, we present the current state of this field and provide our perspectives of the methodologies and conclusions concerning functional differentiation between gyri and sulci, as well as the supporting information from genetic, cell biology, and brain structure research. In particular, we will further present a proposed framework for attempting to interpret the dynamic mechanisms of the functional interplay between gyri and sulci. Hopefully, this review will provide a comprehensive summary of anatomo-functional relationships in the cortical gyro-sulcal system together with a consideration of how these contribute to brain function, cognition, and behavior, as well as to mental disorders.
Collapse
Affiliation(s)
- Xi Jiang
- School of Life Science and Technology, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Tuo Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an 710129, China
| | - Shu Zhang
- Center for Brain and Brain-Inspired Computing Research, School of Computer Science, Northwestern Polytechnical University, Xi'an 710129, China
| | - Keith M Kendrick
- School of Life Science and Technology, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Tianming Liu
- Cortical Architecture Imaging and Discovery Laboratory, Department of Computer Science and Bioimaging Research Center, The University of Georgia, Athens, GA 30605, USA
| |
Collapse
|
19
|
Figueres-Oñate M, Sánchez-González R, López-Mascaraque L. Deciphering neural heterogeneity through cell lineage tracing. Cell Mol Life Sci 2021; 78:1971-1982. [PMID: 33151389 PMCID: PMC7966193 DOI: 10.1007/s00018-020-03689-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/10/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
Understanding how an adult brain reaches an appropriate size and cell composition from a pool of progenitors that proliferates and differentiates is a key question in Developmental Neurobiology. Not only the control of final size but also, the proper arrangement of cells of different embryonic origins is fundamental in this process. Each neural progenitor has to produce a precise number of sibling cells that establish clones, and all these clones will come together to form the functional adult nervous system. Lineage cell tracing is a complex and challenging process that aims to reconstruct the offspring that arise from a single progenitor cell. This tracing can be achieved through strategies based on genetically modified organisms, using either genetic tracers, transfected viral vectors or DNA constructs, and even single-cell sequencing. Combining different reporter proteins and the use of transgenic mice revolutionized clonal analysis more than a decade ago and now, the availability of novel genome editing tools and single-cell sequencing techniques has vastly improved the capacity of lineage tracing to decipher progenitor potential. This review brings together the strategies used to study cell lineages in the brain and the role they have played in our understanding of the functional clonal relationships among neural cells. In addition, future perspectives regarding the study of cell heterogeneity and the ontogeny of different cell lineages will also be addressed.
Collapse
Affiliation(s)
- María Figueres-Oñate
- Department of Molecular, Cellular and Development Neurobiology, Instituto Cajal-CSIC, 28002, Madrid, Spain
- Max Planck Research Unit for Neurogenetics, 60438, Frankfurt am Main, Germany
| | - Rebeca Sánchez-González
- Department of Molecular, Cellular and Development Neurobiology, Instituto Cajal-CSIC, 28002, Madrid, Spain
| | - Laura López-Mascaraque
- Department of Molecular, Cellular and Development Neurobiology, Instituto Cajal-CSIC, 28002, Madrid, Spain.
| |
Collapse
|
20
|
Abstract
The mammalian cerebral cortex is the pinnacle of brain evolution, reaching its maximum complexity in terms of neuron number, diversity and functional circuitry. The emergence of this outstanding complexity begins during embryonic development, when a limited number of neural stem and progenitor cells manage to generate myriads of neurons in the appropriate numbers, types and proportions, in a process called neurogenesis. Here we review the current knowledge on the regulation of cortical neurogenesis, beginning with a description of the types of progenitor cells and their lineage relationships. This is followed by a review of the determinants of neuron fate, the molecular and genetic regulatory mechanisms, and considerations on the evolution of cortical neurogenesis in vertebrates leading to humans. We finish with an overview on how dysregulation of neurogenesis is a leading cause of human brain malformations and functional disabilities.
Collapse
Affiliation(s)
- Ana Villalba
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Magdalena Götz
- Institute for Stem Cell Research, Helmholtz Zentrum München & Biomedical Center, Ludwig-Maximilians Universitaet, Planegg-Martinsried, Germany
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain.
| |
Collapse
|
21
|
Amin S, Borrell V. The Extracellular Matrix in the Evolution of Cortical Development and Folding. Front Cell Dev Biol 2020; 8:604448. [PMID: 33344456 PMCID: PMC7744631 DOI: 10.3389/fcell.2020.604448] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/12/2020] [Indexed: 02/02/2023] Open
Abstract
The evolution of the mammalian cerebral cortex leading to humans involved a remarkable sophistication of developmental mechanisms. Specific adaptations of progenitor cell proliferation and neuronal migration mechanisms have been proposed to play major roles in this evolution of neocortical development. One of the central elements influencing neocortex development is the extracellular matrix (ECM). The ECM provides both a structural framework during tissue formation and to present signaling molecules to cells, which directly influences cell behavior and movement. Here we review recent advances in the understanding of the role of ECM molecules on progenitor cell proliferation and neuronal migration, and how these contribute to cerebral cortex expansion and folding. We discuss how transcriptomic studies in human, ferret and mouse identify components of ECM as being candidate key players in cortex expansion during development and evolution. Then we focus on recent functional studies showing that ECM components regulate cortical progenitor cell proliferation, neuron migration and the mechanical properties of the developing cortex. Finally, we discuss how these features differ between lissencephalic and gyrencephalic species, and how the molecular evolution of ECM components and their expression profiles may have been fundamental in the emergence and evolution of cortex folding across mammalian phylogeny.
Collapse
Affiliation(s)
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d’Alacant, Spain
| |
Collapse
|
22
|
Huang JY, Krebs BB, Miskus ML, Russell ML, Duffy EP, Graf JM, Lu HC. Enhanced FGFR3 activity in postmitotic principal neurons during brain development results in cortical dysplasia and axonal tract abnormality. Sci Rep 2020; 10:18508. [PMID: 33116259 PMCID: PMC7595096 DOI: 10.1038/s41598-020-75537-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Abnormal levels of fibroblast growth factors (FGFs) and FGF receptors (FGFRs) have been detected in various neurological disorders. The potent impact of FGF-FGFR in multiple embryonic developmental processes makes it challenging to elucidate their roles in postmitotic neurons. Taking an alternative approach to examine the impact of aberrant FGFR function on glutamatergic neurons, we generated a FGFR gain-of-function (GOF) transgenic mouse, which expresses constitutively activated FGFR3 (FGFR3K650E) in postmitotic glutamatergic neurons. We found that GOF disrupts mitosis of radial-glia neural progenitors (RGCs), inside-out radial migration of post-mitotic glutamatergic neurons, and axonal tract projections. In particular, late-born CUX1-positive neurons are widely dispersed throughout the GOF cortex. Such a cortical migration deficit is likely caused, at least in part, by a significant reduction of the radial processes projecting from RGCs. RNA-sequencing analysis of the GOF embryonic cortex reveals significant alterations in several pathways involved in cell cycle regulation and axonal pathfinding. Collectively, our data suggest that FGFR3 GOF in postmitotic neurons not only alters axonal growth of postmitotic neurons but also impairs RGC neurogenesis and radial glia processes.
Collapse
Affiliation(s)
- Jui-Yen Huang
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| | - Bruna Baumgarten Krebs
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN, 47405, USA
| | - Marisha Lynn Miskus
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - May Lin Russell
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Eamonn Patrick Duffy
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Jason Michael Graf
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
23
|
Ferent J, Zaidi D, Francis F. Extracellular Control of Radial Glia Proliferation and Scaffolding During Cortical Development and Pathology. Front Cell Dev Biol 2020; 8:578341. [PMID: 33178693 PMCID: PMC7596222 DOI: 10.3389/fcell.2020.578341] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/08/2020] [Indexed: 01/14/2023] Open
Abstract
During the development of the cortex, newly generated neurons migrate long-distances in the expanding tissue to reach their final positions. Pyramidal neurons are produced from dorsal progenitors, e.g., radial glia (RGs) in the ventricular zone, and then migrate along RG processes basally toward the cortex. These neurons are hence dependent upon RG extensions to support their migration from apical to basal regions. Several studies have investigated how intracellular determinants are required for RG polarity and subsequent formation and maintenance of their processes. Fewer studies have identified the influence of the extracellular environment on this architecture. This review will focus on extracellular factors which influence RG morphology and pyramidal neuronal migration during normal development and their perturbations in pathology. During cortical development, RGs are present in different strategic positions: apical RGs (aRGs) have their cell bodies located in the ventricular zone with an apical process contacting the ventricle, while they also have a basal process extending radially to reach the pial surface of the cortex. This particular conformation allows aRGs to be exposed to long range and short range signaling cues, whereas basal RGs (bRGs, also known as outer RGs, oRGs) have their cell bodies located throughout the cortical wall, limiting their access to ventricular factors. Long range signals impacting aRGs include secreted molecules present in the embryonic cerebrospinal fluid (e.g., Neuregulin, EGF, FGF, Wnt, BMP). Secreted molecules also contribute to the extracellular matrix (fibronectin, laminin, reelin). Classical short range factors include cell to cell signaling, adhesion molecules and mechano-transduction mechanisms (e.g., TAG1, Notch, cadherins, mechanical tension). Changes in one or several of these components influencing the RG extracellular environment can disrupt the development or maintenance of RG architecture on which neuronal migration relies, leading to a range of cortical malformations. First, we will detail the known long range signaling cues impacting RG. Then, we will review how short range cell contacts are also important to instruct the RG framework. Understanding how RG processes are structured by their environment to maintain and support radial migration is a critical part of the investigation of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Julien Ferent
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Donia Zaidi
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Fiona Francis
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| |
Collapse
|
24
|
Iwashita M, Nomura T, Suetsugu T, Matsuzaki F, Kojima S, Kosodo Y. Comparative Analysis of Brain Stiffness Among Amniotes Using Glyoxal Fixation and Atomic Force Microscopy. Front Cell Dev Biol 2020; 8:574619. [PMID: 33043008 PMCID: PMC7517470 DOI: 10.3389/fcell.2020.574619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Brain structures are diverse among species despite the essential molecular machinery of neurogenesis being common. Recent studies have indicated that differences in the mechanical properties of tissue may result in the dynamic deformation of brain structure, such as folding. However, little is known about the correlation between mechanical properties and species-specific brain structures. To address this point, a comparative analysis of mechanical properties using several animals is required. For a systematic measurement of the brain stiffness of remotely maintained animals, we developed a novel strategy of tissue-stiffness measurement using glyoxal as a fixative combined with atomic force microscopy. A comparison of embryonic and juvenile mouse and songbird brain tissue revealed that glyoxal fixation can maintain brain structure as well as paraformaldehyde (PFA) fixation. Notably, brain tissue fixed by glyoxal remained much softer than PFA-fixed brains, and it can maintain the relative stiffness profiles of various brain regions. Based on this method, we found that the homologous brain regions between mice and songbirds exhibited different stiffness patterns. We also measured brain stiffness in other amniotes (chick, turtle, and ferret) following glyoxal fixation. We found stage-dependent and species-specific stiffness in pallia among amniotes. The embryonic chick and matured turtle pallia showed gradually increasing stiffness along the apico-basal tissue axis, the lowest region at the most apical region, while the ferret pallium exhibited a catenary pattern, that is, higher in the ventricular zone, the inner subventricular zone, and the cortical plate and the lowest in the outer subventricular zone. These results indicate that species-specific microenvironments with distinct mechanical properties emerging during development might contribute to the formation of brain structures with unique morphology.
Collapse
Affiliation(s)
| | - Tadashi Nomura
- Developmental Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taeko Suetsugu
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | | | | | | |
Collapse
|
25
|
Baburamani AA, Vontell RT, Uus A, Pietsch M, Patkee PA, Wyatt-Ashmead J, Chin-Smith EC, Supramaniam VG, Donald Tournier J, Deprez M, Rutherford MA. Assessment of radial glia in the frontal lobe of fetuses with Down syndrome. Acta Neuropathol Commun 2020; 8:141. [PMID: 32819430 PMCID: PMC7441567 DOI: 10.1186/s40478-020-01015-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
Down syndrome (DS) occurs with triplication of human chromosome 21 and is associated with deviations in cortical development evidenced by simplified gyral appearance and reduced cortical surface area. Radial glia are neuronal and glial progenitors that also create a scaffolding structure essential for migrating neurons to reach cortical targets and therefore play a critical role in cortical development. The aim of this study was to characterise radial glial expression pattern and morphology in the frontal lobe of the developing human fetal brain with DS and age-matched controls. Secondly, we investigated whether microstructural information from in vivo magnetic resonance imaging (MRI) could reflect histological findings from human brain tissue samples. Immunohistochemistry was performed on paraffin-embedded human post-mortem brain tissue from nine fetuses and neonates with DS (15-39 gestational weeks (GW)) and nine euploid age-matched brains (18-39 GW). Radial glia markers CRYAB, HOPX, SOX2, GFAP and Vimentin were assessed in the Ventricular Zone, Subventricular Zone and Intermediate Zone. In vivo diffusion MRI was used to assess microstructure in these regions in one DS (21 GW) and one control (22 GW) fetal brain. We found a significant reduction in radial glial progenitor SOX2 and subtle deviations in radial glia expression (GFAP and Vimentin) prior to 24 GW in DS. In vivo, fetal MRI demonstrates underlying radial projections consistent with immunohistopathology. Radial glial alterations may contribute to the subsequent simplified gyral patterns and decreased cortical volumes observed in the DS brain. Recent advances in fetal MRI acquisition and analysis could provide non-invasive imaging-based biomarkers of early developmental deviations.
Collapse
Affiliation(s)
- Ana A. Baburamani
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - Regina T. Vontell
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
- University of Miami Brain Endowment Bank, Miami, FL 33136 USA
| | - Alena Uus
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - Maximilian Pietsch
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - Prachi A. Patkee
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - Jo Wyatt-Ashmead
- Neuropathology and Pediatric-Perinatal Pathology Service [NaPPPS], Holly Springs, MS 38635 USA
| | - Evonne C. Chin-Smith
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - Veena G. Supramaniam
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - J. Donald Tournier
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - Maria Deprez
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| | - Mary A. Rutherford
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King’s College London, London, SE1 7EH UK
| |
Collapse
|
26
|
Najas S, Pijuan I, Esteve-Codina A, Usieto S, Martinez JD, Zwijsen A, Arbonés ML, Martí E, Le Dréau G. A SMAD1/5-YAP signalling module drives radial glia self-amplification and growth of the developing cerebral cortex. Development 2020; 147:dev.187005. [PMID: 32541003 DOI: 10.1242/dev.187005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/02/2020] [Indexed: 12/19/2022]
Abstract
The growth and evolutionary expansion of the cerebral cortex are defined by the spatial-temporal production of neurons, which itself depends on the decision of radial glial cells (RGCs) to self-amplify or to switch to neurogenic divisions. The mechanisms regulating these RGC fate decisions are still incompletely understood. Here, we describe a novel and evolutionarily conserved role of the canonical BMP transcription factors SMAD1/5 in controlling neurogenesis and growth during corticogenesis. Reducing the expression of both SMAD1 and SMAD5 in neural progenitors at early mouse cortical development caused microcephaly and an increased production of early-born cortical neurons at the expense of late-born ones, which correlated with the premature differentiation and depletion of the pool of cortical progenitors. Gain- and loss-of-function experiments performed during early cortical neurogenesis in the chick revealed that SMAD1/5 activity supports self-amplifying RGC divisions and restrains the neurogenic ones. Furthermore, we demonstrate that SMAD1/5 stimulate RGC self-amplification through the positive post-transcriptional regulation of the Hippo signalling effector YAP. We anticipate this SMAD1/5-YAP signalling module to be fundamental in controlling growth and evolution of the amniote cerebral cortex.
Collapse
Affiliation(s)
- Sonia Najas
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Isabel Pijuan
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 4, 08028 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Susana Usieto
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| | - Juan D Martinez
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| | - An Zwijsen
- Department of Cardiovascular Sciences, KU Leuven, Leuven 3000, Belgium
| | - Maria L Arbonés
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Elisa Martí
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| | - Gwenvael Le Dréau
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| |
Collapse
|
27
|
Cárdenas A, Borrell V. Molecular and cellular evolution of corticogenesis in amniotes. Cell Mol Life Sci 2020; 77:1435-1460. [PMID: 31563997 PMCID: PMC11104948 DOI: 10.1007/s00018-019-03315-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/03/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
The cerebral cortex varies dramatically in size and complexity between amniotes due to differences in neuron number and composition. These differences emerge during embryonic development as a result of variations in neurogenesis, which are thought to recapitulate modifications occurred during evolution that culminated in the human neocortex. Here, we review work from the last few decades leading to our current understanding of the evolution of neurogenesis and size of the cerebral cortex. Focused on specific examples across vertebrate and amniote phylogeny, we discuss developmental mechanisms regulating the emergence, lineage, complexification and fate of cortical germinal layers and progenitor cell types. At the cellular level, we discuss the fundamental impact of basal progenitor cells and the advent of indirect neurogenesis on the increased number and diversity of cortical neurons and layers in mammals, and on cortex folding. Finally, we discuss recent work that unveils genetic and molecular mechanisms underlying this progressive expansion and increased complexity of the amniote cerebral cortex during evolution, with a particular focus on those leading to human-specific features. Whereas new genes important in human brain development emerged the recent hominid lineage, regulation of the patterns and levels of activity of highly conserved signaling pathways are beginning to emerge as mechanisms of central importance in the evolutionary increase in cortical size and complexity across amniotes.
Collapse
Affiliation(s)
- Adrián Cárdenas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernández, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernández, 03550, Sant Joan d'Alacant, Alicante, Spain.
| |
Collapse
|
28
|
Charvet CJ, Das A, Song JW, Tindal-Burgess DJ, Kabaria P, Dai G, Kane T, Takahashi E. High Angular Resolution Diffusion MRI Reveals Conserved and Deviant Programs in the Paths that Guide Human Cortical Circuitry. Cereb Cortex 2020; 30:1447-1464. [PMID: 31667494 PMCID: PMC7132938 DOI: 10.1093/cercor/bhz178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/13/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
Diffusion magnetic resonance (MR) tractography represents a novel opportunity to investigate conserved and deviant developmental programs between humans and other species such as mice. To that end, we acquired high angular resolution diffusion MR scans of mice [embryonic day (E) 10.5 to postnatal week 4] and human brains [gestational week (GW) 17-30] at successive stages of fetal development to investigate potential evolutionary changes in radial organization and emerging pathways between humans and mice. We compare radial glial development as well as commissural development (e.g., corpus callosum), primarily because our findings can be integrated with previous work. We also compare corpus callosal growth trajectories across primates (i.e., humans and rhesus macaques) and rodents (i.e., mice). One major finding is that the developing cortex of humans is predominated by pathways likely associated with a radial glial organization at GW 17-20, which is not as evident in age-matched mice (E 16.5, 17.5). Another finding is that, early in development, the corpus callosum follows a similar developmental timetable in primates (i.e., macaques and humans) as in mice. However, the corpus callosum grows for an extended period of time in primates compared with rodents. Taken together, these findings highlight deviant developmental programs underlying the emergence of cortical pathways in the human brain.
Collapse
Affiliation(s)
| | - Avilash Das
- Medical Sciences in the College of Arts and Sciences, Boston University, Boston, MA 02215, USA
- Division of Newborn Medicine, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Fetal-Neonatal Brain Imaging and Developmental Science Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Jae W Song
- Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Priya Kabaria
- Department of Behavioral Neuroscience, Northeastern University, Boston, MA 02115, USA
| | - Guangping Dai
- Science Center, Wellesley College, Wellesley, MA 02481, USA
| | - Tara Kane
- Department of Behavioral Neuroscience, Northeastern University, Boston, MA 02115, USA
| | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Fetal-Neonatal Brain Imaging and Developmental Science Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02215, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
29
|
Coolen M, Labusch M, Mannioui A, Bally-Cuif L. Mosaic Heterochrony in Neural Progenitors Sustains Accelerated Brain Growth and Neurogenesis in the Juvenile Killifish N. furzeri. Curr Biol 2020; 30:736-745.e4. [PMID: 32004451 PMCID: PMC7040570 DOI: 10.1016/j.cub.2019.12.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/16/2019] [Accepted: 12/13/2019] [Indexed: 12/19/2022]
Abstract
Although developmental mechanisms driving an increase in brain size during vertebrate evolution are actively studied, we know less about evolutionary strategies allowing accelerated brain growth. In zebrafish and other vertebrates studied to date, apical radial glia (RG) constitute the primary neurogenic progenitor population throughout life [1]; thus, RG activity is a determining factor of growth speed. Here, we ask whether enhanced RG activity is the mechanism selected to drive explosive growth, in adaptation to an ephemeral habitat. In post-hatching larvae of the turquoise killifish, which display drastic developmental acceleration, we show that the dorsal telencephalon (pallium) grows three times faster than in zebrafish. Rather than resulting from enhanced RG activity, we demonstrate that pallial growth is the product of a second type of progenitors (that we term NGPs for non-glial progenitors) that actively sustains neurogenesis and germinal zone self-renewal. Intriguingly, NGPs appear to retain, at larval stages, features of early embryonic progenitors. In parallel, RGs enter premature quiescence and express markers of astroglial function. Altogether, we propose that mosaic heterochrony within the neural progenitor population might permit rapid pallial growth by safeguarding both continued neurogenesis and astroglial function. Two types of apical progenitors exist in the pallium of the fast-growing killifish Killifish pallial RGs enter precociously into an adult-like quiescent state NGPs, both self-renewing and neurogenic, resemble early neuroepithelial progenitors Mosaic heterochrony among progenitors sustains rapid killifish pallial growth
Collapse
Affiliation(s)
- Marion Coolen
- Zebrafish Neurogenetics Unit, Developmental & Stem Cell Biology Department, Institut Pasteur, UMR3738, CNRS, 25 rue du Dr Roux, 75015 Paris, France.
| | - Miriam Labusch
- Zebrafish Neurogenetics Unit, Developmental & Stem Cell Biology Department, Institut Pasteur, UMR3738, CNRS, 25 rue du Dr Roux, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Abdelkrim Mannioui
- Institut de Biologie Paris-Seine (IBPS), Aquatic Facility, Sorbonne Université, 7 quai Saint Bernard, 75005 Paris, France
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Unit, Developmental & Stem Cell Biology Department, Institut Pasteur, UMR3738, CNRS, 25 rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
30
|
Signs of Reduced Basal Progenitor Levels and Cortical Neurogenesis in Human Fetuses with Open Spina Bifida at 11-15 Weeks of Gestation. J Neurosci 2020; 40:1766-1777. [PMID: 31953373 DOI: 10.1523/jneurosci.0192-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 12/06/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022] Open
Abstract
Open spina bifida (OSB) is one of the most prevalent congenital malformations of the CNS that often leads to severe disabilities. Previous studies reported the volume and thickness of the neocortex to be altered in children and adolescents diagnosed with OSB. Until now, the onset and the underlying cause of the atypical neocortex organization in OSB patients remain largely unknown. To examine the effects of OSB on fetal neocortex development, we analyzed human fetuses of both sexes diagnosed with OSB between 11 and 15 weeks of gestation by immunofluorescence for established neuronal and neural progenitor marker proteins and compared the results with healthy controls of the same, or very similar, gestational age. Our data indicate that neocortex development in OSB fetuses is altered as early as 11 weeks of gestation. We observed a marked reduction in the radial thickness of the OSB neocortex, which appears to be attributable to a massive decrease in the number of deep- and upper-layer neurons per field, and found a marked reduction in the number of basal progenitors (BPs) per field in the OSB neocortex, consistent with an impairment of cortical neurogenesis underlying the neuronal decrease in OSB fetuses. Moreover, our data suggest that the decrease in BP number in the OSB neocortex may be associated with BPs spending a lesser proportion of their cell cycle in M-phase. Together, our findings expand our understanding of the pathophysiology of OSB and support the need for an early fetal therapy (i.e., in the first trimester of pregnancy).SIGNIFICANCE STATEMENT Open spina bifida (OSB) is one of the most prevalent congenital malformations of the CNS. This study provides novel data on neocortex development of human OSB fetuses. Our data indicate that neocortex development in OSB fetuses is altered as early as 11 weeks of gestation. We observed a marked reduction in the radial thickness of the OSB neocortex, which appears to be attributable a decrease in the number of deep- and upper-layer neurons per field, and found a marked reduction in the number of basal progenitors per field, indicating that impaired neurogenesis underlies the neuronal decrease in OSB fetuses. Our findings support the need for an early fetal therapy and expand our understanding of the pathophysiology of OSB.
Collapse
|
31
|
Wang X, Ma Y, Xu R, Ma J, Zhang H, Qi S, Xu J, Qin X, Zhang H, Liu C, Chen J, Li B, Yang H, Saijilafu. c‐Myc controls the fate of neural progenitor cells during cerebral cortex development. J Cell Physiol 2019; 235:4011-4021. [PMID: 31625158 DOI: 10.1002/jcp.29297] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 09/30/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Xiu‐Li Wang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Yan‐Xia Ma
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Ren‐Jie Xu
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
- Department of Orthopaedics Suzhou Municipal Hospital/The Affiliated Hospital of Nanjing Medical University Suzhou Jiangsu China
| | - Jin‐Jin Ma
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Hong‐Cheng Zhang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Shi‐Bin Qi
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Jin‐Hui Xu
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Xu‐Zhen Qin
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Hao‐Nan Zhang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Chang‐Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology Chinese Academy of Science Beijing China
- Savaid Medical School University of Chinese Academy of Sciences Beijing China
| | - Jian‐Quan Chen
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Bin Li
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Hui‐Lin Yang
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| | - Saijilafu
- Department of Orthopaedics The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University Suzhou Jiangsu China
| |
Collapse
|
32
|
Castaneyra-Ruiz L, Morales DM, McAllister JP, Brody SL, Isaacs AM, Strahle JM, Dahiya SM, Limbrick DD. Blood Exposure Causes Ventricular Zone Disruption and Glial Activation In Vitro. J Neuropathol Exp Neurol 2019; 77:803-813. [PMID: 30032242 DOI: 10.1093/jnen/nly058] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Intraventricular hemorrhage (IVH) is the most common cause of pediatric hydrocephalus in North America but remains poorly understood. Cell junction-mediated ventricular zone (VZ) disruption and astrogliosis are associated with the pathogenesis of congenital, nonhemorrhagic hydrocephalus. Recently, our group demonstrated that VZ disruption is also present in preterm infants with IVH. On the basis of this observation, we hypothesized that blood triggers the loss of VZ cell junction integrity and related cytopathology. In order to test this hypothesis, we developed an in vitro model of IVH by applying syngeneic blood to cultured VZ cells obtained from newborn mice. Following blood treatment, cells were assayed for N-cadherin-dependent adherens junctions, ciliated ependymal cells, and markers of glial activation using immunohistochemistry and immunoblotting. After 24-48 hours of exposure to blood, VZ cell junctions were disrupted as determined by a significant reduction in N-cadherin expression (p < 0.05). This was also associated with significant decrease in multiciliated cells and increase in glial fibrillary acid protein-expressing cells (p < 0.05). These observations suggest that, in vitro, blood triggers VZ cell loss and glial activation in a pattern that mirrors the cytopathology of human IVH and supports the relevance of this in vitro model to define injury mechanisms.
Collapse
Affiliation(s)
- Leandro Castaneyra-Ruiz
- Department of Neurological Surgery, Washington University School of Medicine and the St. Louis Children's Hospital, St. Louis, Missouri
| | - Diego M Morales
- Department of Neurological Surgery, Washington University School of Medicine and the St. Louis Children's Hospital, St. Louis, Missouri
| | - James P McAllister
- Department of Neurological Surgery, Washington University School of Medicine and the St. Louis Children's Hospital, St. Louis, Missouri
| | | | | | - Jennifer M Strahle
- Department of Neurological Surgery, Washington University School of Medicine and the St. Louis Children's Hospital, St. Louis, Missouri.,Department of Pediatrics
| | - Sonika M Dahiya
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - David D Limbrick
- Department of Neurological Surgery, Washington University School of Medicine and the St. Louis Children's Hospital, St. Louis, Missouri.,Department of Pediatrics
| |
Collapse
|
33
|
Zhang L, Zhang X, Zhang Y, Xu N, Wang J, Zhu Y, Xia C. Brn4 promotes the differentiation of radial glial cells into neurons by inhibiting CtBP2. Life Sci 2019; 254:116866. [PMID: 31518606 DOI: 10.1016/j.lfs.2019.116866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 10/26/2022]
Abstract
Neural stem cells (NSCs) are pluripotent cells that are capable of differentiating into neurons and considered as the most promising cell source for cell replacement therapy. However, the difficulty in inducing neuronal differentiation and maturation from NSCs is a major challenge for their clinical application. Clarifying the molecular mechanisms underlying the neuronal differentiation of NSCs can provide a basis for expanding their uses. Brain 4 (Brn4) is a member of the POU domain family of transcription factors and can induce the neuronal differentiation of NSCs, but its precise function in NSCs is unclear. To address this question, in this study we isolated and expanded radial glial cells (RGCs), a type of NSC, from the cerebral cortex of 14-day embryonic rats and used lentivirus carrying the human Brn4 gene to overexpress Brn4 in these cells. This induced the differentiation of RGCs into neurons and inhibited the expression of C-terminal binding protein 2 (CtBP2), a transcriptional co-repressor. CtBP2 overexpression in RGCs suppressed their differentiation into neurons, whereas CtBP2 knockdown had the opposite effect. These results indicated that Brn4 promoted the neuronal differentiation of NSCs via inhibition of CtBP2 and is a potential tool for generating neurons in cell replacement therapy of neurodegenerative diseases and brain injury.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Anatomy and Cytoneurobiology Unit, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xinhua Zhang
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Ye Zhang
- Department of Anatomy and Cytoneurobiology Unit, Medical College of Soochow University, Suzhou, Jiangsu 215123, China
| | - Naijuan Xu
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jue Wang
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Yuanyuan Zhu
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Chunlin Xia
- Department of Anatomy and Cytoneurobiology Unit, Medical College of Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
34
|
Lv X, Ren SQ, Zhang XJ, Shen Z, Ghosh T, Xianyu A, Gao P, Li Z, Lin S, Yu Y, Zhang Q, Groszer M, Shi SH. TBR2 coordinates neurogenesis expansion and precise microcircuit organization via Protocadherin 19 in the mammalian cortex. Nat Commun 2019; 10:3946. [PMID: 31477701 PMCID: PMC6718393 DOI: 10.1038/s41467-019-11854-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/06/2019] [Indexed: 11/09/2022] Open
Abstract
Cerebral cortex expansion is a hallmark of mammalian brain evolution; yet, how increased neurogenesis is coordinated with structural and functional development remains largely unclear. The T-box protein TBR2/EOMES is preferentially enriched in intermediate progenitors and supports cortical neurogenesis expansion. Here we show that TBR2 regulates fine-scale spatial and circuit organization of excitatory neurons in addition to enhancing neurogenesis in the mouse cortex. TBR2 removal leads to a significant reduction in neuronal, but not glial, output of individual radial glial progenitors as revealed by mosaic analysis with double markers. Moreover, in the absence of TBR2, clonally related excitatory neurons become more laterally dispersed and their preferential synapse development is impaired. Interestingly, TBR2 directly regulates the expression of Protocadherin 19 (PCDH19), and simultaneous PCDH19 expression rescues neurogenesis and neuronal organization defects caused by TBR2 removal. Together, these results suggest that TBR2 coordinates neurogenesis expansion and precise microcircuit assembly via PCDH19 in the mammalian cortex.
Collapse
Affiliation(s)
- Xiaohui Lv
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Si-Qiang Ren
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center of Biological Structures, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xin-Jun Zhang
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Zhongfu Shen
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center of Biological Structures, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tanay Ghosh
- Inserm, UMR-S839, Sorbonne Université, Institut du Fer à Moulin, Paris, 75005, France.,Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Anjin Xianyu
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Graduate Program in Biophysics, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Peng Gao
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Graduate Program in Neuroscience, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Zhizhong Li
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Susan Lin
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Graduate Program in Neuroscience, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Yang Yu
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Qiangqiang Zhang
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Matthias Groszer
- Inserm, UMR-S839, Sorbonne Université, Institut du Fer à Moulin, Paris, 75005, France
| | - Song-Hai Shi
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA. .,IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Sciences, Beijing Frontier Research Center of Biological Structures, School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,Graduate Program in Biophysics, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA. .,Graduate Program in Neuroscience, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
35
|
Chen P, Zhang T, Yuan Z, Shen B, Chen L. Expression of the RNA methyltransferase Nsun5 is essential for developing cerebral cortex. Mol Brain 2019; 12:74. [PMID: 31462248 PMCID: PMC6714381 DOI: 10.1186/s13041-019-0496-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/21/2019] [Indexed: 11/10/2022] Open
Abstract
Nsun5 gene, encoding a cytosine-5 RNA methyltransferase, is deleted in about 95% patients with Williams-Beuren syndrome (WBS). WBS is a neurodevelopmental disorder and characterized by cognitive disorder. We generated single-gene Nsun5 knockout (Nsun5-KO) mice and reported that the Nsun5 deletion leads to deficit in spatial cognition. This study focused on investigating the influence of Nsun5 deficiency in the development of cerebral cortex. In comparison with wild-type littermates, the cortical thickness in postnatal day 10 Nsun5-KO mice was obviously reduced with an abnormal laminar organization, and the processes of pyramidal cells were shorter and finer. Nsun5 was selectively expressed in radial glial cells (RGCs) of cerebral cortex from embryonic day (E) 12.5 to E16.5, but not in intermediate progenitor cells (IPCs) or neocortical neurons. The Nsun5 deletion did not alter proliferation of RGCs or differentiation of RGCs into IPCs. Notably, the ablation of Nsun5 disrupted the growth of radial glial scaffolds, thus numerous basal processes of RGCs failed to reach pial basement membrane. Level of cell polarity regulator Cdc42 protein in radial glial scaffolds of E14.5 Nsun5-KO mice was reduced, but the level of Cdc42 mRNA was unchanged. The dysfunction of glial scaffolds impeded the radial migration of upper-layer and deeper-layer neurons to cause their subcortical accumulation and apoptosis, resulting in an obvious thinness of the cortical plate in E18.5 Nsun5-KO mice. These findings establish a critical role of Nsun5 in development of cerebral cortex through regulating radial glial scaffolds of RGCs to control migration of neocortical neurons.
Collapse
Affiliation(s)
- Peipei Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China.,Department of Physiology, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China
| | - Tingting Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China.,Department of Physiology, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China
| | - Zihao Yuan
- Department of Physiology, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China
| | - Bin Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China.
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China. .,Department of Physiology, Nanjing Medical University, Tianyuan East Road 818, Nanjing, China.
| |
Collapse
|
36
|
Penisson M, Ladewig J, Belvindrah R, Francis F. Genes and Mechanisms Involved in the Generation and Amplification of Basal Radial Glial Cells. Front Cell Neurosci 2019; 13:381. [PMID: 31481878 PMCID: PMC6710321 DOI: 10.3389/fncel.2019.00381] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022] Open
Abstract
The development of the cerebral cortex relies on different types of progenitor cell. Among them, the recently described basal radial glial cell (bRG) is suggested to be of critical importance for the development of the brain in gyrencephalic species. These cells are highly numerous in primate and ferret brains, compared to lissencephalic species such as the mouse in which they are few in number. Their somata are located in basal subventricular zones in gyrencephalic brains and they generally possess a basal process extending to the pial surface. They sometimes also have an apical process directed toward the ventricular surface, similar to apical radial glial cells (aRGs) from which they are derived, and whose somata are found more apically in the ventricular zone. bRGs share similarities with aRGs in terms of gene expression (SOX2, PAX6, and NESTIN), whilst also expressing a range of more specific genes (such as HOPX). In primate brains, bRGs can divide multiple times, self-renewing and/or generating intermediate progenitors and neurons. They display a highly specific cytokinesis behavior termed mitotic somal translocation. We focus here on recently identified molecular mechanisms associated with the generation and amplification of bRGs, including bRG-like cells in the rodent. These include signaling pathways such as the FGF-MAPK cascade, SHH, PTEN/AKT, PDGF pathways, and proteins such as INSM, GPSM2, ASPM, TRNP1, ARHGAP11B, PAX6, and HIF1α. A number of these proteins were identified through transcriptome comparisons in human aRGs vs. bRGs, and validated by modifying their activities or expression levels in the mouse. This latter experiment often revealed enhanced bRG-like cell production, even in some cases generating folds (gyri) on the surface of the mouse cortex. We compare the features of the identified cells and methods used to characterize them in each model. These important data converge to indicate pathways essential for the production and expansion of bRGs, which may help us understand cortical development in health and disease.
Collapse
Affiliation(s)
- Maxime Penisson
- Inserm, Institut du Fer à Moulin, Sorbonne Université, Paris, France.,Inserm UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Julia Ladewig
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research (gGmbH), Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Richard Belvindrah
- Inserm, Institut du Fer à Moulin, Sorbonne Université, Paris, France.,Inserm UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Fiona Francis
- Inserm, Institut du Fer à Moulin, Sorbonne Université, Paris, France.,Inserm UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
37
|
Tennant KG, Lindsley SR, Kirigiti MA, True C, Kievit P. Central and Peripheral Administration of Fibroblast Growth Factor 1 Improves Pancreatic Islet Insulin Secretion in Diabetic Mouse Models. Diabetes 2019; 68:1462-1472. [PMID: 31048370 PMCID: PMC6609981 DOI: 10.2337/db18-1175] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/15/2019] [Indexed: 12/19/2022]
Abstract
Fibroblast growth factor 1 (FGF1) has been shown to reverse hyperglycemia in diabetic rodent models through peripheral and central administration routes. Previous studies demonstrated that insulin is required for central and peripheral FGF1 metabolic improvements; however, it is unknown if FGF1 targets insulin secretion at the islet level. Here we show for the first time that FGF1 increases islet insulin secretion in diabetic mouse models. FGF1 was administered via a single intracerebroventricular or multiple subcutaneous injections to leptin receptor-deficient (db/db), diet-induced obese, and control mice; pancreatic islets were isolated 7 days later for analysis of insulin secretion. Central and peripheral FGF1 significantly lowered blood glucose in vivo and increased ex vivo islet insulin secretion from diabetic, but not control, mice. FGF1 injections to the cisterna magna mimicked intracerebroventricular outcomes, pointing to a novel therapeutic potential. Central effects of FGF1 appeared dependent on reductions in food intake, whereas peripheral FGF1 had acute actions on islet function prior to significant changes in food intake or blood glucose. Additionally, peripheral, but not central, FGF1 increased islet β-cell density, suggesting that peripheral FGF1 may induce long-term changes in islet structure and function that are not present with central treatment.
Collapse
Affiliation(s)
- Katherine G Tennant
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR
| | - Sarah R Lindsley
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR
| | - Melissa A Kirigiti
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR
| | - Cadence True
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR
| |
Collapse
|
38
|
Kyrousi C, Cappello S. Using brain organoids to study human neurodevelopment, evolution and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e347. [PMID: 31071759 DOI: 10.1002/wdev.347] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/18/2019] [Accepted: 04/07/2019] [Indexed: 01/12/2023]
Abstract
The brain is one of the most complex organs, responsible for the advanced intellectual and cognitive ability of humans. Although primates are to some extent capable of performing cognitive tasks, their abilities are less evolved. One of the reasons for this is the vast differences in the brain of humans compared to other mammals, in terms of shape, size and complexity. Such differences make the study of human brain development fascinating. Interestingly, the cerebral cortex is by far the most complex brain region resulting from its selective evolution within mammals over millions of years. Unraveling the molecular and cellular mechanisms regulating brain development, as well as the evolutionary differences seen across species and the need to understand human brain disorders, are some of the reasons why scientists are interested in improving their current knowledge on human corticogenesis. Toward this end, several animal models including primates have been used, however, these models are limited in their extent to recapitulate human-specific features. Recent technological achievements in the field of stem cell research, which have enabled the generation of human models of corticogenesis, called brain or cerebral organoids, are of great importance. This review focuses on the main cellular and molecular features of human corticogenesis and the use of brain organoids to study it. We will discuss the key differences between cortical development in human and nonhuman mammals, the technological applications of brain organoids and the different aspects of cortical development in normal and pathological conditions, which can be modeled using brain organoids. This article is categorized under: Comparative Development and Evolution > Regulation of Organ Diversity Nervous System Development > Vertebrates: General Principles.
Collapse
Affiliation(s)
- Christina Kyrousi
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Silvia Cappello
- Department of Developmental Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
39
|
Kostović I, Sedmak G, Judaš M. Neural histology and neurogenesis of the human fetal and infant brain. Neuroimage 2018; 188:743-773. [PMID: 30594683 DOI: 10.1016/j.neuroimage.2018.12.043] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 01/11/2023] Open
Abstract
The human brain develops slowly and over a long period of time which lasts for almost three decades. This enables good spatio-temporal resolution of histogenetic and neurogenetic events as well as an appropriate and clinically relevant timing of these events. In order to successfully apply in vivo neuroimaging data, in analyzing both the normal brain development and the neurodevelopmental origin of major neurological and mental disorders, it is important to correlate these neuroimaging data with the existing data on morphogenetic, histogenetic and neurogenetic events. Furthermore, when performing such correlation, the genetic, genomic, and molecular biology data on phenotypic specification of developing brain regions, areas and neurons should also be included. In this review, we focus on early developmental periods (form 8 postconceptional weeks to the second postnatal year) and describe the microstructural organization and neural circuitry elements of the fetal and early postnatal human cerebrum.
Collapse
Affiliation(s)
- I Kostović
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Centre of Excellence for Basic, Clinical and Translational Neuroscience, Šalata 12, 10000, Zagreb, Croatia.
| | - G Sedmak
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Centre of Excellence for Basic, Clinical and Translational Neuroscience, Šalata 12, 10000, Zagreb, Croatia.
| | - M Judaš
- University of Zagreb School of Medicine, Croatian Institute for Brain Research, Centre of Excellence for Basic, Clinical and Translational Neuroscience, Šalata 12, 10000, Zagreb, Croatia.
| |
Collapse
|
40
|
Žunić Išasegi I, Radoš M, Krsnik Ž, Radoš M, Benjak V, Kostović I. Interactive histogenesis of axonal strata and proliferative zones in the human fetal cerebral wall. Brain Struct Funct 2018; 223:3919-3943. [PMID: 30094607 PMCID: PMC6267252 DOI: 10.1007/s00429-018-1721-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022]
Abstract
Development of the cerebral wall is characterized by partially overlapping histogenetic events. However, little is known with regards to when, where, and how growing axonal pathways interact with progenitor cell lineages in the proliferative zones of the human fetal cerebrum. We analyzed the developmental continuity and spatial distribution of the axonal sagittal strata (SS) and their relationship with proliferative zones in a series of human brains (8-40 post-conceptional weeks; PCW) by comparing histological, histochemical, and immunocytochemical data with magnetic resonance imaging (MRI). Between 8.5 and 11 PCW, thalamocortical fibers from the intermediate zone (IZ) were initially dispersed throughout the subventricular zone (SVZ), while sizeable axonal "invasion" occurred between 12.5 and 15 PCW followed by callosal fibers which "delaminated" the ventricular zone-inner SVZ from the outer SVZ (OSVZ). During midgestation, the SS extensively invaded the OSVZ, separating cell bands, and a new multilaminar axonal-cellular compartment (MACC) was formed. Preterm period reveals increased complexity of the MACC in terms of glial architecture and the thinning of proliferative bands. The addition of associative fibers and the formation of the centrum semiovale separated the SS from the subplate. In vivo MRI of the occipital SS indicates a "triplet" structure of alternating hypointense and hyperintense bands. Our results highlighted the developmental continuity of sagittally oriented "corridors" of projection, commissural and associative fibers, and histogenetic interaction with progenitors, neurons, and glia. Histogenetical changes in the MACC, and consequently, delineation of the SS on MRI, may serve as a relevant indicator of white matter microstructural integrity in the developing brain.
Collapse
Affiliation(s)
- Iris Žunić Išasegi
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Milan Radoš
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Marko Radoš
- Department of Radiology, Clinical Hospital Center Zagreb, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Vesna Benjak
- Department of Pediatrics, Clinical Hospital Center Zagreb, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, Centar of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, School of Medicine, Zagreb, Croatia.
| |
Collapse
|
41
|
Garcia KE, Kroenke CD, Bayly PV. Mechanics of cortical folding: stress, growth and stability. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2017.0321. [PMID: 30249772 DOI: 10.1098/rstb.2017.0321] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2018] [Indexed: 12/17/2022] Open
Abstract
Cortical folding, or gyrification, coincides with several important developmental processes. The folded shape of the human brain allows the cerebral cortex, the thin outer layer of neurons and their associated projections, to attain a large surface area relative to brain volume. Abnormal cortical folding has been associated with severe neurological, cognitive and behavioural disorders, such as epilepsy, autism and schizophrenia. However, despite decades of study, the mechanical forces that lead to cortical folding remain incompletely understood. Leading hypotheses have focused on the roles of (i) tangential growth of the outer cortex, (ii) spatio-temporal patterns in the birth and migration of neurons, and (iii) internal tension in axons. Recent experimental studies have illuminated not only the fundamental cellular and molecular processes underlying cortical development, but also the stress state, mechanical properties and spatio-temporal patterns of growth in the developing brain. The combination of mathematical modelling and physical measurements has allowed researchers to evaluate hypothesized mechanisms of folding, to determine whether each is consistent with physical laws. This review summarizes what physical scientists have learned from models and recent experimental observations, in the context of recent neurobiological discoveries regarding cortical development. Here, we highlight evidence of a combined mechanism, in which spatio-temporal patterns bias the locations of primary folds (i), but tangential growth of the cortical plate induces mechanical instability (ii) to propagate primary and higher-order folds.This article is part of the Theo Murphy meeting issue 'Mechanics of development'.
Collapse
Affiliation(s)
- K E Garcia
- Mechanical Engineering and Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA.,Engineering, University of Southern Indiana, Evansville, IN, USA
| | - C D Kroenke
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - P V Bayly
- Mechanical Engineering and Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| |
Collapse
|
42
|
Sauerland C, Menzies BR, Glatzle M, Seeger J, Renfree MB, Fietz SA. The Basal Radial Glia Occurs in Marsupials and Underlies the Evolution of an Expanded Neocortex in Therian Mammals. Cereb Cortex 2018; 28:145-157. [PMID: 29253253 DOI: 10.1093/cercor/bhw360] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
A hallmark of mammalian brain evolution is the emergence of the neocortex, which has expanded in all mammalian infraclasses (Eutheria, Marsupialia, Monotremata). In eutherians, neocortical neurons derive from distinct neural stem and progenitor cells (NPCs). However, precise data on the presence and abundance of the NPCs, especially of basal radial glia (bRG), in the neocortex of marsupials are lacking. This study characterized and quantified the NPCs in the developing neocortex of a marsupial, the tammar wallaby (Macropus eugenii). Our data demonstrate that its neocortex is characterized by high NPC diversity. Importantly, we show that bRG exist at high relative abundance in the tammar indicating that this cell type is not specific to the eutherian neocortex and that similar mechanisms may underlie the formation of an expanded neocortex in eutherian and marsupial mammals. We also show that bRG are likely to have been present in the therian ancestor, so did not emerge independently in the eutherian and marsupial lineages. Moreover, our data support the concept that changes in multiple parameters contribute to neocortex expansion and demonstrate the importance of bRG and other NPCs for the development and expansion of the mammalian neocortex.
Collapse
Affiliation(s)
- Christine Sauerland
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| | - Brandon R Menzies
- School of BioSciences, The University of Melbourne, Victoria 3010, Melbourne, Australia
| | - Megan Glatzle
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| | - Johannes Seeger
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Victoria 3010, Melbourne, Australia
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
43
|
Cárdenas A, Villalba A, de Juan Romero C, Picó E, Kyrousi C, Tzika AC, Tessier-Lavigne M, Ma L, Drukker M, Cappello S, Borrell V. Evolution of Cortical Neurogenesis in Amniotes Controlled by Robo Signaling Levels. Cell 2018; 174:590-606.e21. [PMID: 29961574 PMCID: PMC6063992 DOI: 10.1016/j.cell.2018.06.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/24/2018] [Accepted: 06/01/2018] [Indexed: 11/29/2022]
Abstract
Cerebral cortex size differs dramatically between reptiles, birds, and mammals, owing to developmental differences in neuron production. In mammals, signaling pathways regulating neurogenesis have been identified, but genetic differences behind their evolution across amniotes remain unknown. We show that direct neurogenesis from radial glia cells, with limited neuron production, dominates the avian, reptilian, and mammalian paleocortex, whereas in the evolutionarily recent mammalian neocortex, most neurogenesis is indirect via basal progenitors. Gain- and loss-of-function experiments in mouse, chick, and snake embryos and in human cerebral organoids demonstrate that high Slit/Robo and low Dll1 signaling, via Jag1 and Jag2, are necessary and sufficient to drive direct neurogenesis. Attenuating Robo signaling and enhancing Dll1 in snakes and birds recapitulates the formation of basal progenitors and promotes indirect neurogenesis. Our study identifies modulation in activity levels of conserved signaling pathways as a primary mechanism driving the expansion and increased complexity of the mammalian neocortex during amniote evolution.
Collapse
Affiliation(s)
- Adrián Cárdenas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Ana Villalba
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Camino de Juan Romero
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Esther Picó
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Christina Kyrousi
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Athanasia C Tzika
- Department Genetics and Evolution, University of Geneva, 1205 Geneva, Switzerland; SIB Swiss Institute of Bioinformatics, 1211 Geneva, Switzerland
| | | | - Le Ma
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Micha Drukker
- Institute of Stem Cell Research and the Induced Pluripotent Stem Cell Core Facility, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Silvia Cappello
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain.
| |
Collapse
|
44
|
Nganou G, Silva CG, Gladwyn-Ng I, Engel D, Coumans B, Delgado-Escueta AV, Tanaka M, Nguyen L, Grisar T, de Nijs L, Lakaye B. Importin-8 Modulates Division of Apical Progenitors, Dendritogenesis and Tangential Migration During Development of Mouse Cortex. Front Mol Neurosci 2018; 11:234. [PMID: 30042658 PMCID: PMC6048241 DOI: 10.3389/fnmol.2018.00234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/13/2018] [Indexed: 01/18/2023] Open
Abstract
The building of the brain is a multistep process that requires the coordinate expression of thousands of genes and an intense nucleocytoplasmic transport of RNA and proteins. This transport is mediated by karyopherins that comprise importins and exportins. Here, we investigated the role of the ß-importin, importin-8 (IPO8) during mouse cerebral corticogenesis as several of its cargoes have been shown to be essential during this process. First, we showed that Ipo8 mRNA is expressed in mouse brain at various embryonic ages with a clear signal in the sub-ventricular/ventricular zone (SVZ/VZ), the cerebral cortical plate (CP) and the ganglionic eminences. We found that acute knockdown of IPO8 in cortical progenitors reduced both their proliferation and cell cycle exit leading to the increase in apical progenitor pool without influencing the number of basal progenitors (BPs). Projection neurons ultimately reached their appropriate cerebral cortical layer, but their dendritogenesis was specifically affected, resulting in neurons with reduced dendrite complexity. IPO8 knockdown also slowed the migration of cortical interneurons. Together, our data demonstrate that IPO8 contribute to the coordination of several critical steps of cerebral cortex development. These results suggest that the impairment of IPO8 function might be associated with some diseases of neuronal migration defects.
Collapse
Affiliation(s)
- Gerry Nganou
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| | - Carla G Silva
- GIGA-Neurosciences, University of Liege, Liege, Belgium
| | | | | | - Bernard Coumans
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| | - Antonio V Delgado-Escueta
- GENESS International Consortium, Los Angeles, CA, United States.,Epilepsy Genetics/Genomics Lab, Neurology and Research Services, VA Greater Los Angeles Healthcare System (VA GLAHS), University of California, Los Angeles, Los Angeles, CA, United States
| | - Miyabi Tanaka
- GENESS International Consortium, Los Angeles, CA, United States.,Epilepsy Genetics/Genomics Lab, Neurology and Research Services, VA Greater Los Angeles Healthcare System (VA GLAHS), University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Thierry Grisar
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| | - Laurence de Nijs
- GENESS International Consortium, Los Angeles, CA, United States.,MHeNS, Maastricht University, Maastricht, Netherlands
| | - Bernard Lakaye
- GIGA-Neurosciences, University of Liege, Liege, Belgium.,GENESS International Consortium, Los Angeles, CA, United States
| |
Collapse
|
45
|
Römer S, Bender H, Knabe W, Zimmermann E, Rübsamen R, Seeger J, Fietz SA. Neural Progenitors in the Developing Neocortex of the Northern Tree Shrew ( Tupaia belangeri) Show a Closer Relationship to Gyrencephalic Primates Than to Lissencephalic Rodents. Front Neuroanat 2018; 12:29. [PMID: 29725291 PMCID: PMC5917011 DOI: 10.3389/fnana.2018.00029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/03/2018] [Indexed: 01/03/2023] Open
Abstract
The neocortex is the most complex part of the mammalian brain and as such it has undergone tremendous expansion during evolution, especially in primates. The majority of neocortical neurons originate from distinct neural stem and progenitor cells (NPCs) located in the ventricular and subventricular zone (SVZ). Previous studies revealed that the SVZ thickness as well as the abundance and distribution of NPCs, especially that of basal radial glia (bRG), differ markedly between the lissencephalic rodent and gyrencephalic primate neocortex. The northern tree shrew (Tupaia belangeri) is a rat-sized mammal with a high brain to body mass ratio, which stands phylogenetically mid-way between rodents and primates. Our study provides – for the first time – detailed data on the presence, abundance and distribution of bRG and other distinct NPCs in the developing neocortex of the northern tree shrew (Tupaia belangeri). We show that the developing tree shrew neocortex is characterized by an expanded SVZ, a high abundance of Pax6+ NPCs in the SVZ, and a relatively high percentage of bRG at peak of upper-layer neurogenesis. We further demonstrate that key features of tree shrew neocortex development, e.g., the presence, abundance and distribution of distinct NPCs, are closer related to those of gyrencephalic primates than to those of ferret and lissencephalic rodents. Together, our study provides novel insight into the evolution of bRG and other distinct NPCs in the neocortex development of Euarchontoglires and introduces the tree shrew as a potential novel model organism in the area of human brain development and developmental disorders.
Collapse
Affiliation(s)
- Sebastian Römer
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| | - Hannah Bender
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| | - Wolfgang Knabe
- Prosektur Anatomie, Medizinische Fakultät, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Elke Zimmermann
- Institute of Zoology, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Rudolf Rübsamen
- Institute of Biology, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
| | - Johannes Seeger
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
46
|
Psychiatry in a Dish: Stem Cells and Brain Organoids Modeling Autism Spectrum Disorders. Biol Psychiatry 2018; 83:558-568. [PMID: 29295738 DOI: 10.1016/j.biopsych.2017.11.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 12/23/2022]
Abstract
Autism spectrum disorders are a group of pervasive neurodevelopmental conditions with heterogeneous etiology, characterized by deficits in social cognition, communication, and behavioral flexibility. Despite an increasing scientific effort to find the pathophysiological explanations for the disease, the neurobiological links remain unclear. A large amount of evidence suggests that pathological processes taking place in early embryonic neurodevelopment might be responsible for later manifestation of autistic symptoms. This dysfunctional development includes altered maturation/differentiation processes, disturbances in cell-cell communication, and an unbalanced ratio between certain neuronal populations. All those processes are highly dependent on the interconnectivity and three-dimensional organizations of the brain. Moreover, in order to gain a deeper understanding of the complex neurobiology of autism spectrum disorders, valid disease models are pivotal. Induced pluripotent stem cells could potentially help to elucidate the complex mechanisms of the disease and lead to the development of more effective individualized treatment. The induced pluripotent stem cells approach allows comparison between the development of various cellular phenotypes generated from cell lines of patients and healthy individuals. A newly advanced organoid technology makes it possible to create three-dimensional in vitro models of brain development and structural interconnectivity, based on induced pluripotent stem cells derived from the respective individuals. The biggest challenge for modeling psychiatric diseases in vitro is finding and establishing the link between cellular and molecular findings with the clinical symptoms, and this review aims to give an overview over the feasibility and applicability of this new tissue engineering tool in psychiatry.
Collapse
|
47
|
Helm C, Karl A, Beckers P, Kaul-Strehlow S, Ulbricht E, Kourtesis I, Kuhrt H, Hausen H, Bartolomaeus T, Reichenbach A, Bleidorn C. Early evolution of radial glial cells in Bilateria. Proc Biol Sci 2018; 284:rspb.2017.0743. [PMID: 28724733 PMCID: PMC5543218 DOI: 10.1098/rspb.2017.0743] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/13/2017] [Indexed: 12/13/2022] Open
Abstract
Bilaterians usually possess a central nervous system, composed of neurons and supportive cells called glial cells. Whereas neuronal cells are highly comparable in all these animals, glial cells apparently differ, and in deuterostomes, radial glial cells are found. These particular secretory glial cells may represent the archetype of all (macro) glial cells and have not been reported from protostomes so far. This has caused controversial discussions of whether glial cells represent a homologous bilaterian characteristic or whether they (and thus, centralized nervous systems) evolved convergently in the two main clades of bilaterians. By using histology, transmission electron microscopy, immunolabelling and whole-mount in situ hybridization, we show here that protostomes also possess radial glia-like cells, which are very likely to be homologous to those of deuterostomes. Moreover, our antibody staining indicates that the secretory character of radial glial cells is maintained throughout their various evolutionary adaptations. This implies an early evolution of radial glial cells in the last common ancestor of Protostomia and Deuterostomia. Furthermore, it suggests that an intraepidermal nervous system—composed of sensory cells, neurons and radial glial cells—was probably the plesiomorphic condition in the bilaterian ancestor.
Collapse
Affiliation(s)
- Conrad Helm
- Sars International Center for Marine Molecular Biology, University of Bergen, 5008 Bergen, Norway
| | - Anett Karl
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany.,Translational Center for Regenerative Medicine, University of Leipzig, 04103 Leipzig, Germany.,Carl-Ludwig-Institute for Physiology, University of Leipzig, 04103 Leipzig, Germany
| | - Patrick Beckers
- Institute of Evolutionary Biology and Ecology, University of Bonn, 53121 Bonn, Germany
| | | | - Elke Ulbricht
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ioannis Kourtesis
- Sars International Center for Marine Molecular Biology, University of Bergen, 5008 Bergen, Norway
| | - Heidrun Kuhrt
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Harald Hausen
- Sars International Center for Marine Molecular Biology, University of Bergen, 5008 Bergen, Norway
| | - Thomas Bartolomaeus
- Institute of Evolutionary Biology and Ecology, University of Bonn, 53121 Bonn, Germany
| | - Andreas Reichenbach
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Christoph Bleidorn
- Museo Nacional de Ciencias Naturales, Spanish National Research Council (CSIC), 28006 Madrid, Spain
| |
Collapse
|
48
|
Bayliss J, Mukherjee P, Lu C, Jain SU, Chung C, Martinez D, Sabari B, Margol AS, Panwalkar P, Parolia A, Pekmezci M, McEachin RC, Cieslik M, Tamrazi B, Garcia BA, La Rocca G, Santi M, Lewis PW, Hawkins C, Melnick A, David Allis C, Thompson CB, Chinnaiyan AM, Judkins AR, Venneti S. Lowered H3K27me3 and DNA hypomethylation define poorly prognostic pediatric posterior fossa ependymomas. Sci Transl Med 2017; 8:366ra161. [PMID: 27881822 DOI: 10.1126/scitranslmed.aah6904] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 08/02/2016] [Accepted: 10/19/2016] [Indexed: 12/12/2022]
Abstract
Childhood posterior fossa (PF) ependymomas cause substantial morbidity and mortality. These tumors lack recurrent genetic mutations, but a subset of these ependymomas exhibits CpG island (CpGi) hypermethylation [PF group A (PFA)], implicating epigenetic alterations in their pathogenesis. Further, histological grade does not reliably predict prognosis, highlighting the importance of developing more robust prognostic markers. We discovered global H3K27me3 reduction in a subset of these tumors (PF-ve ependymomas) analogous to H3K27M mutant gliomas. PF-ve tumors exhibited many clinical and biological similarities with PFA ependymomas. Genomic H3K27me3 distribution showed an inverse relationship with CpGi methylation, suggesting that CpGi hypermethylation drives low H3K27me3 in PF-ve ependymomas. Despite CpGi hypermethylation and global H3K27me3 reduction, these tumors showed DNA hypomethylation in the rest of the genome and exhibited increased H3K27me3 genomic enrichment at limited genomic loci similar to H3K27M mutant gliomas. Combined integrative analysis of PF-ve ependymomas with H3K27M gliomas uncovered common epigenetic deregulation of select factors that control radial glial biology, and PF radial glia in early human development exhibited reduced H3K27me3. Finally, H3K27me3 immunostaining served as a biomarker of poor prognosis and delineated radiologically invasive tumors, suggesting that reduced H3K27me3 may be a prognostic indicator in PF ependymomas.
Collapse
Affiliation(s)
- Jill Bayliss
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Piali Mukherjee
- Epigenomics Core Facility, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Chao Lu
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY 10065, USA
| | - Siddhant U Jain
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53715, USA
| | - Chan Chung
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Daniel Martinez
- Department of Pathology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin Sabari
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY 10065, USA
| | - Ashley S Margol
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Pooja Panwalkar
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA.,Michigan Center for Translational Pathology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48104, USA
| | - Melike Pekmezci
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Richard C McEachin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48104, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA.,Michigan Center for Translational Pathology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48104, USA
| | - Benita Tamrazi
- Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Benjamin A Garcia
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gaspare La Rocca
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mariarita Santi
- Department of Pathology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter W Lewis
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53715, USA
| | - Cynthia Hawkins
- Arthur and Sonia Labatt Brain Tumour Research Centre and Division of Pathology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Faculty of Medicine, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ari Melnick
- Epigenomics Core Facility, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - C David Allis
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY 10065, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA.,Michigan Center for Translational Pathology, Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48104, USA
| | - Alexander R Judkins
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA.
| | - Sriram Venneti
- Department of Pathology, University of Michigan, Ann Arbor, MI 48104, USA.
| |
Collapse
|
49
|
McIntosh R, Norris J, Clarke JD, Alexandre P. Spatial distribution and characterization of non-apical progenitors in the zebrafish embryo central nervous system. Open Biol 2017; 7:rsob.160312. [PMID: 28148823 PMCID: PMC5356445 DOI: 10.1098/rsob.160312] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 12/21/2016] [Indexed: 11/24/2022] Open
Abstract
Studies of non-apical progenitors (NAPs) have been largely limited to the developing mammalian cortex. They are postulated to generate the increase in neuron numbers that underlie mammalian brain expansion. Recently, NAPs have also been reported in the retina and central nervous system of non-mammalian species; in the latter, however, they remain poorly characterized. Here, we characterize NAP location along the zebrafish central nervous system during embryonic development, and determine their cellular and molecular characteristics and renewal capacity. We identified a small population of NAPs in the spinal cord, hindbrain and telencephalon of zebrafish embryos. Live-imaging analysis revealed at least two types of mitotic behaviour in the telencephalon: one NAP subtype retains the apical attachment during division, while another divides in a subapical position disconnected from the apical surface. All NAPs observed in spinal cord lost apical contact prior to mitoses. These NAPs express HuC and produce two neurons from a single division. Manipulation of Notch activity reveals that neurons and NAPs in the spinal cord use similar regulatory mechanisms. This work suggests that the majority of spinal NAPs in zebrafish share characteristics with basal progenitors in mammalian brains.
Collapse
Affiliation(s)
- Rebecca McIntosh
- Developmental Biology and Cancer Department, UCL Institute of Child Health, London WC1N 1EH, UK.,Division of Surgery and Interventional Science, University College London, London, UK
| | - Joseph Norris
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Jon D Clarke
- Department of Developmental Neurobiology, King's College London, London SE1 1UL, UK
| | - Paula Alexandre
- Developmental Biology and Cancer Department, UCL Institute of Child Health, London WC1N 1EH, UK
| |
Collapse
|
50
|
Goffinet AM. The evolution of cortical development: the synapsid-diapsid divergence. Development 2017; 144:4061-4077. [PMID: 29138289 DOI: 10.1242/dev.153908] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cerebral cortex covers the rostral part of the brain and, in higher mammals and particularly humans, plays a key role in cognition and consciousness. It is populated with neuronal cell bodies distributed in radially organized layers. Understanding the common and lineage-specific molecular mechanisms that orchestrate cortical development and evolution are key issues in neurobiology. During evolution, the cortex appeared in stem amniotes and evolved divergently in two main branches of the phylogenetic tree: the synapsids (which led to present day mammals) and the diapsids (reptiles and birds). Comparative studies in organisms that belong to those two branches have identified some common principles of cortical development and organization that are possibly inherited from stem amniotes and regulated by similar molecular mechanisms. These comparisons have also highlighted certain essential features of mammalian cortices that are absent or different in diapsids and that probably evolved after the synapsid-diapsid divergence. Chief among these is the size and multi-laminar organization of the mammalian cortex, and the propensity to increase its area by folding. Here, I review recent data on cortical neurogenesis, neuronal migration and cortical layer formation and folding in this evolutionary perspective, and highlight important unanswered questions for future investigation.
Collapse
Affiliation(s)
- Andre M Goffinet
- University of Louvain, Avenue Mounier, 73 Box B1.73.16, B1200 Brussels, Belgium
| |
Collapse
|