1
|
Mwachaka PM, Gichangi P, Abdelmalek A, Odula P, Ogeng’o J. Maternal dietary folate imbalance alters cerebellar astrocyte morphology and density in offspring. IBRO Neurosci Rep 2025; 18:78-87. [PMID: 39844943 PMCID: PMC11751541 DOI: 10.1016/j.ibneur.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background Maternal folate usage is essential for neurodevelopment, but its effects on cerebellar structure are unclear. Cerebellum undergoes a protracted period of development, making it sensitive to maternal nutritional imbalances. Astrocytes are necessary for cerebellar cortex structure and function. This study examined the impact of varying maternal dietary folate levels on the morphology and density of cerebellar astrocytes in rat offspring. Materials and methods Twelve adult female rats (Rattus norvegicus) were randomly allocated to one of four premixed food groups: standard (2 mg/kg), folate-deficient (0 mg/kg), folate-supplemented (8 mg/kg), or folate supra-supplemented (40 mg/kg). The rats began their diets 14 days before mating and continued throughout pregnancy and lactation. On postnatal day 35, five pups from each group were sacrificed and their cerebellums were processed for immunohistochemical examination. The cerebellar astrocytes were labelled with an antibody against Glial Fibrillary Acid Protein (GFAP). Results The offspring of the folate-deficient diet group exhibited few Bergmann and granule layer astrocytes. The Bergmann radial glial processes in this group were thinner, discontinuous, poorly organised, and had unclear end feet compared to controls. Conversely, the folate-supplemented group showed a predominance of well-organized Bergmann glia astrocytes with distinct, thicker, and densely packed processes, ending in clear conical pial foot processes. In the supra-supplemented group, there was evidence of astrogliosis in the form of large granule layer astrocytes with extended cytoplasmic projections. The Bergmann glia in this group were fewer and more varied in distribution and morphology. Some locations had many astrocytic processes, whereas others had none. Some processes were discontinuous and tortuous. The proportion of cerebellar cortical GFAP immunoreactive cells in folate-deficient diet, controls, folate-supplemented, and folate supra-supplemented groups were 2.09 ± 0.06 %, 4.69 ± 0.12 %, 10.14 ± 0.67 %, and 23.12 ± 3.48 %, respectively (p < 0.001). Conclusions These findings imply that both folate deficiency and excess supplementation in pregnancy can impair normal cerebellar astrocyte development, highlighting the importance of balanced folate levels during pregnancy for optimal neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Philip Maseghe Mwachaka
- Department of Human Anatomy and Medical Physiology, Faculty of Health Sciences, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Peter Gichangi
- Department of Human Anatomy and Medical Physiology, Faculty of Health Sciences, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Adel Abdelmalek
- Department of Human Anatomy and Medical Physiology, Faculty of Health Sciences, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Paul Odula
- Department of Human Anatomy and Medical Physiology, Faculty of Health Sciences, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Julius Ogeng’o
- Department of Human Anatomy and Medical Physiology, Faculty of Health Sciences, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| |
Collapse
|
2
|
Pakula A, El Nagar S, Bayin NS, Christensen JB, Stephen DN, Reid AJ, Koche R, Joyner AL. An increase in reactive oxygen species underlies neonatal cerebellum repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.14.618368. [PMID: 39464104 PMCID: PMC11507802 DOI: 10.1101/2024.10.14.618368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The neonatal mouse cerebellum shows remarkable regenerative potential upon injury at birth, wherein a subset of Nestin-expressing progenitors (NEPs) undergoes adaptive reprogramming to replenish granule cell progenitors that die. Here, we investigate how the microenvironment of the injured cerebellum changes upon injury and contributes to the regenerative potential of normally gliogenic - NEPs and their adaptive reprogramming. Single cell transcriptomic and bulk chromatin accessibility analyses of the NEPs from injured neonatal cerebella compared to controls show a temporary increase in cellular processes involved in responding to reactive oxygen species (ROS), a known damage-associated molecular pattern. Analysis of ROS levels in cerebellar tissue confirm a transient increased one day after injury at postanal day 1, overlapping with the peak cell death in the cerebellum. In a transgenic mouse line that ubiquitously overexpresses human mitochondrial catalase (mCAT), ROS is reduced 1 day after injury to the granule cell progenitors, and we demonstrate that several steps in the regenerative process of NEPs are curtailed leading to reduced cerebellar growth. We also provide preliminary evidence that microglia are involved in one step of adaptive reprogramming by regulating NEP replenishment of the granule cell precursors. Collectively, our results highlight that changes in the tissue microenvironment regulate multiple steps in adaptative reprogramming of NEPs upon death of cerebellar granule cell progenitors at birth, highlighting the instructive roles of microenvironmental signals during regeneration of the neonatal brain.
Collapse
|
3
|
Marullo C, Croci L, Giupponi I, Rivoletti C, Zuffetti S, Bettegazzi B, Cremona O, Giunti P, Ambrosi A, Casoni F, Consalez GG, Codazzi F. Altered Ca2+ responses and antioxidant properties in Friedreich's ataxia-like cerebellar astrocytes. J Cell Sci 2025; 138:jcs263446. [PMID: 39648860 PMCID: PMC11828468 DOI: 10.1242/jcs.263446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
Friedreich's ataxia (FRDA) is a neurodegenerative disorder characterized by severe neurological signs, affecting the peripheral and central nervous system, caused by reduced frataxin protein (FXN) levels. Although several studies have highlighted cellular dysfunctions in neurons, there is limited information on the effects of FXN depletion in astrocytes and on the potential non-cell autonomous mechanisms affecting neurons in FRDA. In this study, we generated a model of FRDA cerebellar astrocytes to unveil phenotypic alterations that might contribute to cerebellar atrophy. We treated primary cerebellar astrocytes with an RNA interference-based approach, to achieve a reduction of FXN comparable to that observed in individuals with FRDA. These FRDA-like astrocytes display some typical features of the disease, such as an increase of oxidative stress and a depletion of glutathione content. Moreover, FRDA-like astrocytes exhibit decreased Ca2+ responses to purinergic stimuli. Our findings shed light on cellular changes caused by FXN downregulation in cerebellar astrocytes, likely impairing their complex interaction with neurons. The potentially impaired ability to provide neuronal cells with glutathione or to release neuromodulators in a Ca2+-dependent manner could affect neuronal function, contributing to neurodegeneration.
Collapse
Affiliation(s)
- Chiara Marullo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Laura Croci
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Iris Giupponi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudia Rivoletti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sofia Zuffetti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Barbara Bettegazzi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ottavio Cremona
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Paola Giunti
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Alessandro Ambrosi
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Filippo Casoni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Gian Giacomo Consalez
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Franca Codazzi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
4
|
Lozano Casasbuenas D, Kortebi I, Gora C, Scott EY, Gomes C, Oliveira MS, Sharma T, Daniele E, Olfat A, Gibbs R, Yuzwa SA, Gilbert EA, Küry P, Wheeler AR, Lévesque M, Faiz M. The laminar position, morphology, and gene expression profiles of cortical astrocytes are influenced by time of birth from ventricular/subventricular progenitors. Glia 2024; 72:1693-1706. [PMID: 38852127 DOI: 10.1002/glia.24578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Astrocytes that reside in superficial (SL) and deep cortical layers have distinct molecular profiles and morphologies, which may underlie specific functions. Here, we demonstrate that the production of SL and deep layer (DL) astrocyte populations from neural progenitor cells in the mouse is temporally regulated. Lineage tracking following in utero and postnatal electroporation with PiggyBac (PB) EGFP and birth dating with EdU and FlashTag, showed that apical progenitors produce astrocytes during late embryogenesis (E16.5) that are biased to the SL, while postnatally labeled (P0) astrocytes are biased to the DL. In contrast, astrocytes born during the predominantly neurogenic window (E14.5) showed a random distribution in the SL and DL. Of interest, E13.5 astrocytes birth dated at E13.5 with EdU showed a lower layer bias, while FT labeling of apical progenitors showed no bias. Finally, examination of the morphologies of "biased" E16.5- and P0-labeled astrocytes demonstrated that E16.5-labeled astrocytes exhibit different morphologies in different layers, while P0-labeled astrocytes do not. Differences based on time of birth are also observed in the molecular profiles of E16.5 versus P0-labeled astrocytes. Altogether, these results suggest that the morphological, molecular, and positional diversity of cortical astrocytes is related to their time of birth from ventricular/subventricular zone progenitors.
Collapse
Affiliation(s)
- Daniela Lozano Casasbuenas
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ines Kortebi
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Charles Gora
- Department of Psychiatry and Neurosciences, Université Laval, Québec, Canada; CERVO Brain Research Center, Québec, Canada
| | - Erica Y Scott
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Celeste Gomes
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Markley Silva Oliveira
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Tanvi Sharma
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Emerson Daniele
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Arman Olfat
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Gibbs
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Emily A Gilbert
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Küry
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Aaron R Wheeler
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Université Laval, Québec, Canada; CERVO Brain Research Center, Québec, Canada
| | - Maryam Faiz
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Yu H, Liu Y, Xu F, Fu Y, Yang M, Ding L, Wu Y, Tang F, Qiao J, Wen L. A human fetal cerebellar map of the late second trimester reveals developmental molecular characteristics and abnormality in trisomy 21. Cell Rep 2024; 43:114586. [PMID: 39137113 DOI: 10.1016/j.celrep.2024.114586] [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: 02/06/2024] [Revised: 05/29/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Our understanding of human fetal cerebellum development during the late second trimester, a critical period for the generation of astrocytes, oligodendrocytes, and unipolar brush cells (UBCs), remains limited. Here, we performed single-cell RNA sequencing (scRNA-seq) in human fetal cerebellum samples from gestational weeks (GWs) 18-25. We find that proliferating UBC progenitors distribute in the subventricular zone of the rhombic lip (RLSVZ) near white matter (WM), forming a layer structure. We also delineate two trajectories from astrogenic radial glia (ARGs) to Bergmann glial progenitors (BGPs) and recognize oligodendrogenic radial glia (ORGs) as one source of primitive oligodendrocyte progenitor cells (PriOPCs). Additionally, our scRNA-seq analysis of the trisomy 21 fetal cerebellum at this stage reveals abnormal upregulated genes in pathways such as the cell adhesion pathway and focal adhesion pathway, which potentially promote neuronal differentiation. Overall, our research provides valuable insights into normal and abnormal development of the human fetal cerebellum.
Collapse
Affiliation(s)
- Hongmin Yu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Yun Liu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China
| | - Fanqing Xu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yuanyuan Fu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Ming Yang
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Ling Ding
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yixuan Wu
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China
| | - Jie Qiao
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China.
| | - Lu Wen
- Biomedical Pioneering Innovation Center, Department of Obstetrics and Gynecology, Academy for Advanced Interdisciplinary Studies, Third Hospital, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Changping Laboratory, Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China.
| |
Collapse
|
6
|
Schilling K. Revisiting the development of cerebellar inhibitory interneurons in the light of single-cell genetic analyses. Histochem Cell Biol 2024; 161:5-27. [PMID: 37940705 PMCID: PMC10794478 DOI: 10.1007/s00418-023-02251-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 11/10/2023]
Abstract
The present review aims to provide a short update of our understanding of the inhibitory interneurons of the cerebellum. While these cells constitute but a minority of all cerebellar neurons, their functional significance is increasingly being recognized. For one, inhibitory interneurons of the cerebellar cortex are now known to constitute a clearly more diverse group than their traditional grouping as stellate, basket, and Golgi cells suggests, and this diversity is now substantiated by single-cell genetic data. The past decade or so has also provided important information about interneurons in cerebellar nuclei. Significantly, developmental studies have revealed that the specification and formation of cerebellar inhibitory interneurons fundamentally differ from, say, the cortical interneurons, and define a mode of diversification critically dependent on spatiotemporally patterned external signals. Last, but not least, in the past years, dysfunction of cerebellar inhibitory interneurons could also be linked with clinically defined deficits. I hope that this review, however fragmentary, may stimulate interest and help focus research towards understanding the cerebellum.
Collapse
Affiliation(s)
- Karl Schilling
- Anatomisches Institut - Anatomie und Zellbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Nussallee 10, 53115, Bonn, Germany.
| |
Collapse
|
7
|
Rigoldi L, Mallamaci A. Assessing Neuronogenic Versus Astrogenic Bias of Neural Stem Cells Via In Vitro Clonal Assay. Methods Mol Biol 2024; 2736:95-103. [PMID: 37284943 DOI: 10.1007/7651_2023_481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Within the developing cerebral cortex, neural stem cells (NSCs) give rise to neurons and glial cells, according to complex spatio-temporal trajectories. In this respect, a key issue is how NSCs are committed to different neural lineages in time and space. Clonal assays are a powerful tool to address this issue. Here we describe an easy clonal assay protocol employable to dissect NSCs lineage commitment and molecular mechanisms underlying it. NSCs of distinctive spatio-temporal origin, and/or having undergone different molecular manipulations, are plated at low density and allowed to differentiate for a few days. Then, systematic immunoprofiling of the resulting clones allows to quantify commitment of their NSC ancestors to neuronal and astroglial fates.
Collapse
Affiliation(s)
- Laura Rigoldi
- Laboratory of Cerebral Cortex Development, Department of Neuroscience, SISSA, Trieste, Italy
| | - Antonello Mallamaci
- Laboratory of Cerebral Cortex Development, Department of Neuroscience, SISSA, Trieste, Italy.
| |
Collapse
|
8
|
Sepp M, Leiss K, Murat F, Okonechnikov K, Joshi P, Leushkin E, Spänig L, Mbengue N, Schneider C, Schmidt J, Trost N, Schauer M, Khaitovich P, Lisgo S, Palkovits M, Giere P, Kutscher LM, Anders S, Cardoso-Moreira M, Sarropoulos I, Pfister SM, Kaessmann H. Cellular development and evolution of the mammalian cerebellum. Nature 2024; 625:788-796. [PMID: 38029793 PMCID: PMC10808058 DOI: 10.1038/s41586-023-06884-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
The expansion of the neocortex, a hallmark of mammalian evolution1,2, was accompanied by an increase in cerebellar neuron numbers3. However, little is known about the evolution of the cellular programmes underlying the development of the cerebellum in mammals. In this study we generated single-nucleus RNA-sequencing data for around 400,000 cells to trace the development of the cerebellum from early neurogenesis to adulthood in human, mouse and the marsupial opossum. We established a consensus classification of the cellular diversity in the developing mammalian cerebellum and validated it by spatial mapping in the fetal human cerebellum. Our cross-species analyses revealed largely conserved developmental dynamics of cell-type generation, except for Purkinje cells, for which we observed an expansion of early-born subtypes in the human lineage. Global transcriptome profiles, conserved cell-state markers and gene-expression trajectories across neuronal differentiation show that cerebellar cell-type-defining programmes have been overall preserved for at least 160 million years. However, we also identified many orthologous genes that gained or lost expression in cerebellar neural cell types in one of the species or evolved new expression trajectories during neuronal differentiation, indicating widespread gene repurposing at the cell-type level. In sum, our study unveils shared and lineage-specific gene-expression programmes governing the development of cerebellar cells and expands our understanding of mammalian brain evolution.
Collapse
Affiliation(s)
- Mari Sepp
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| | - Kevin Leiss
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| | - Florent Murat
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
- INRAE, LPGP, Rennes, France
| | - Konstantin Okonechnikov
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Piyush Joshi
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Group, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Evgeny Leushkin
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Lisa Spänig
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Group, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Noe Mbengue
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Céline Schneider
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Julia Schmidt
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Nils Trost
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Maria Schauer
- Museum für Naturkunde Berlin, Leibniz Institute for Evolution and Biodiversity Science, Berlin, Germany
| | - Philipp Khaitovich
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Steven Lisgo
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Miklós Palkovits
- Human Brain Tissue Bank, Semmelweis University, Budapest, Hungary
| | - Peter Giere
- Museum für Naturkunde Berlin, Leibniz Institute for Evolution and Biodiversity Science, Berlin, Germany
| | - Lena M Kutscher
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Group, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Simon Anders
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
- BioQuant, Heidelberg University, Heidelberg, Germany
| | | | - Ioannis Sarropoulos
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Wellcome Sanger Institute, Cambridge, UK.
| | - Stefan M Pfister
- Hopp-Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| | - Henrik Kaessmann
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
9
|
Hong W, Gong P, Pan X, Ren Z, Liu Y, Qi G, Li JL, Sun W, Ge WP, Zhang CL, Duan S, Qin S. Temporal-spatial Generation of Astrocytes in the Developing Diencephalon. Neurosci Bull 2024; 40:1-16. [PMID: 37843774 PMCID: PMC10774245 DOI: 10.1007/s12264-023-01131-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 06/24/2023] [Indexed: 10/17/2023] Open
Abstract
Astrocytes are the largest glial population in the mammalian brain. However, we have a minimal understanding of astrocyte development, especially fate specification in different regions of the brain. Through lineage tracing of the progenitors of the third ventricle (3V) wall via in-utero electroporation in the embryonic mouse brain, we show the fate specification and migration pattern of astrocytes derived from radial glia along the 3V wall. Unexpectedly, radial glia located in different regions along the 3V wall of the diencephalon produce distinct cell types: radial glia in the upper region produce astrocytes and those in the lower region produce neurons in the diencephalon. With genetic fate mapping analysis, we reveal that the first population of astrocytes appears along the zona incerta in the diencephalon. Astrogenesis occurs at an early time point in the dorsal region relative to that in the ventral region of the developing diencephalon. With transcriptomic analysis of the region-specific 3V wall and lateral ventricle (LV) wall, we identified cohorts of differentially-expressed genes in the dorsal 3V wall compared to the ventral 3V wall and LV wall that may regulate astrogenesis in the dorsal diencephalon. Together, these results demonstrate that the generation of astrocytes shows a spatiotemporal pattern in the developing mouse diencephalon.
Collapse
Affiliation(s)
- Wentong Hong
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Pifang Gong
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xinjie Pan
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Zhonggan Ren
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yitong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Guibo Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jun-Liszt Li
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Wenzhi Sun
- Chinese Institute for Brain Research, Beijing, 102206, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390-9148, USA
| | - Shumin Duan
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Okonechnikov K, Joshi P, Sepp M, Leiss K, Sarropoulos I, Murat F, Sill M, Beck P, Chan KCH, Korshunov A, Sah F, Deng MY, Sturm D, DeSisto J, Donson AM, Foreman NK, Green AL, Robinson G, Orr BA, Gao Q, Darrow E, Hadley JL, Northcott PA, Gojo J, Kawauchi D, Hovestadt V, Filbin MG, von Deimling A, Zuckermann M, Pajtler KW, Kool M, Jones DTW, Jäger N, Kutscher LM, Kaessmann H, Pfister SM. Mapping pediatric brain tumors to their origins in the developing cerebellum. Neuro Oncol 2023; 25:1895-1909. [PMID: 37534924 PMCID: PMC10547518 DOI: 10.1093/neuonc/noad124] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND Distinguishing the cellular origins of childhood brain tumors is key for understanding tumor initiation and identifying lineage-restricted, tumor-specific therapeutic targets. Previous strategies to map the cell-of-origin typically involved comparing human tumors to murine embryonal tissues, which is potentially limited due to species-specific differences. The aim of this study was to unravel the cellular origins of the 3 most common pediatric brain tumors, ependymoma, pilocytic astrocytoma, and medulloblastoma, using a developing human cerebellar atlas. METHODS We used a single-nucleus atlas of the normal developing human cerebellum consisting of 176 645 cells as a reference for an in-depth comparison to 4416 bulk and single-cell transcriptome tumor datasets, using gene set variation analysis, correlation, and single-cell matching techniques. RESULTS We find that the astroglial cerebellar lineage is potentially the origin for posterior fossa ependymomas. We propose that infratentorial pilocytic astrocytomas originate from the oligodendrocyte lineage and MHC II genes are specifically enriched in these tumors. We confirm that SHH and Group 3/4 medulloblastomas originate from the granule cell and unipolar brush cell lineages. Radiation-induced gliomas stem from cerebellar glial lineages and demonstrate distinct origins from the primary medulloblastoma. We identify tumor genes that are expressed in the cerebellar lineage of origin, and genes that are tumor specific; both gene sets represent promising therapeutic targets for future study. CONCLUSION Based on our results, individual cells within a tumor may resemble different cell types along a restricted developmental lineage. Therefore, we suggest that tumors can arise from multiple cellular states along the cerebellar "lineage of origin."
Collapse
Affiliation(s)
- Konstantin Okonechnikov
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Piyush Joshi
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mari Sepp
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Kevin Leiss
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Ioannis Sarropoulos
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Florent Murat
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
- INRAE, LPGP, Rennes, France
| | | | - Pengbo Beck
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Kenneth Chun-Ho Chan
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Andrey Korshunov
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Sah
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Maximilian Y Deng
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominik Sturm
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - John DeSisto
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Andrew M Donson
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nicholas K Foreman
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
- Children’s Hospital Colorado, Aurora, CO, USA
| | - Adam L Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
- Children’s Hospital Colorado, Aurora, CO, USA
| | - Giles Robinson
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Brent A Orr
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Qingsong Gao
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Emily Darrow
- Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jennifer L Hadley
- Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Paul A Northcott
- Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Johannes Gojo
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of Neuropathology, NN Burdenko Neurosurgical Institute, Moscow, Russia
| | - Daisuke Kawauchi
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Volker Hovestadt
- Department of Pediatric Oncology, Dana-Farber Boston Children’s Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of Harvard and MIT, Cambridge, USA
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber Boston Children’s Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of Harvard and MIT, Cambridge, USA
| | - Andreas von Deimling
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc Zuckermann
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Kristian W Pajtler
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcel Kool
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, the Netherlands
| | - David T W Jones
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Natalie Jäger
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Lena M Kutscher
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henrik Kaessmann
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children’s Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Mockenhaupt K, Tyc KM, McQuiston A, Gonsiewski AK, Zarei-Kheirabadi M, Hariprashad A, Biswas DD, Gupta AS, Olex AL, Singh SK, Waters MR, Dupree JL, Dozmorov MG, Kordula T. Yin Yang 1 controls cerebellar astrocyte maturation. Glia 2023; 71:2437-2455. [PMID: 37417428 PMCID: PMC10529878 DOI: 10.1002/glia.24434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 07/08/2023]
Abstract
Diverse subpopulations of astrocytes tile different brain regions to accommodate local requirements of neurons and associated neuronal circuits. Nevertheless, molecular mechanisms governing astrocyte diversity remain mostly unknown. We explored the role of a zinc finger transcription factor Yin Yang 1 (YY1) that is expressed in astrocytes. We found that specific deletion of YY1 from astrocytes causes severe motor deficits in mice, induces Bergmann gliosis, and results in simultaneous loss of GFAP expression in velate and fibrous cerebellar astrocytes. Single cell RNA-seq analysis showed that YY1 exerts specific effects on gene expression in subpopulations of cerebellar astrocytes. We found that although YY1 is dispensable for the initial stages of astrocyte development, it regulates subtype-specific gene expression during astrocyte maturation. Moreover, YY1 is continuously needed to maintain mature astrocytes in the adult cerebellum. Our findings suggest that YY1 plays critical roles regulating cerebellar astrocyte maturation during development and maintaining a mature phenotype of astrocytes in the adult cerebellum.
Collapse
Affiliation(s)
- Karli Mockenhaupt
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Katarzyna M. Tyc
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
- Massey Cancer Center Bioinformatics Shared Resource Core, Virginia Commonwealth University, Richmond, Virginia
| | - Adam McQuiston
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Alexandra K. Gonsiewski
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Masoumeh Zarei-Kheirabadi
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Avani Hariprashad
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Debolina D. Biswas
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Angela S. Gupta
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Amy L. Olex
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia
| | - Sandeep K. Singh
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Michael R. Waters
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Jeff L. Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
- Research Service, Central Virginia VA Health Care System, Richmond, Virginia
| | - Mikhail G. Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
- The Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
12
|
Santo M, Rigoldi L, Falcone C, Tuccillo M, Calabrese M, Martínez-Cerdeño V, Mallamaci A. Spatial control of astrogenesis progression by cortical arealization genes. Cereb Cortex 2023; 33:3107-3123. [PMID: 35818636 DOI: 10.1093/cercor/bhac264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Sizes of neuronal, astroglial and oligodendroglial complements forming the neonatal cerebral cortex largely depend on rates at which pallial stem cells give rise to lineage-committed progenitors and the latter ones progress to mature cell types. Here, we investigated the spatial articulation of pallial stem cells' (SCs) commitment to astrogenesis as well as the progression of committed astroglial progenitors (APs) to differentiated astrocytes, by clonal and kinetic profiling of pallial precursors. We found that caudal-medial (CM) SCs are more prone to astrogenesis than rostro-lateral (RL) ones, while RL-committed APs are more keen to proliferate than CM ones. Next, we assessed the control of these phenomena by 2 key transcription factor genes mastering regionalization of the early cortical primordium, Emx2 and Foxg1, via lentiviral somatic transgenesis, epistasis assays, and ad hoc rescue assays. We demonstrated that preferential CM SCs progression to astrogenesis is promoted by Emx2, mainly via Couptf1, Nfia, and Sox9 upregulation, while Foxg1 antagonizes such progression to some extent, likely via repression of Zbtb20. Finally, we showed that Foxg1 and Emx2 may be implicated-asymmetrically and antithetically-in shaping distinctive proliferative/differentiative behaviors displayed by APs in hippocampus and neocortex.
Collapse
Affiliation(s)
- Manuela Santo
- Laboratory of Cerebral Cortex Development, Department of Neuroscience, SISSA, via Bonomea 265, I-34136 Trieste, Italy
| | - Laura Rigoldi
- Laboratory of Cerebral Cortex Development, Department of Neuroscience, SISSA, via Bonomea 265, I-34136 Trieste, Italy
| | - Carmen Falcone
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, 4400 V St, CA-95817 Sacramento, USA
| | - Mariacarmine Tuccillo
- Laboratory of Cerebral Cortex Development, Department of Neuroscience, SISSA, via Bonomea 265, I-34136 Trieste, Italy
| | - Michela Calabrese
- Laboratory of Cerebral Cortex Development, Department of Neuroscience, SISSA, via Bonomea 265, I-34136 Trieste, Italy
| | - Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine & MIND Institute, UC Davis School of Medicine, 4400 V St, CA-95817 Sacramento, USA
| | - Antonello Mallamaci
- Laboratory of Cerebral Cortex Development, Department of Neuroscience, SISSA, via Bonomea 265, I-34136 Trieste, Italy
| |
Collapse
|
13
|
Olmos-Carreño CL, Figueres-Oñate M, Scicolone GE, López-Mascaraque L. Cell Fate of Retinal Progenitor Cells: In Ovo UbC-StarTrack Analysis. Int J Mol Sci 2022; 23:ijms232012388. [PMID: 36293245 PMCID: PMC9604099 DOI: 10.3390/ijms232012388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/28/2022] Open
Abstract
Clonal cell analysis outlines the ontogenic potential of single progenitor cells, allowing the elucidation of the neural heterogeneity among different cell types and their lineages. In this work, we analyze the potency of retinal stem/progenitor cells through development using the chick embryo as a model. We implemented in ovo the clonal genetic tracing strategy UbC-StarTrack for tracking retinal cell lineages derived from individual progenitors of the ciliary margin at E3.5 (HH21-22). The clonal assignment of the derived-cell progeny was performed in the neural retina at E11.5-12 (HH38) through the identification of sibling cells as cells expressing the same combination of fluorophores. Moreover, cell types were assessed based on their cellular morphology and laminar location. Ciliary margin derived-cell progenies are organized in columnar associations distributed along the peripheral retina with a limited tangential dispersion. The analysis revealed that, at the early stages of development, this region harbors multipotent and committed progenitor cells.
Collapse
Affiliation(s)
- Cindy L. Olmos-Carreño
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN), CONICET and Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, 28002 Madrid, Spain
| | - María Figueres-Oñate
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, 28002 Madrid, Spain
- Correspondence: (M.F.-O.); (L.L.-M.)
| | - Gabriel E. Scicolone
- Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN), CONICET and Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1121, Argentina
| | - Laura López-Mascaraque
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, 28002 Madrid, Spain
- Correspondence: (M.F.-O.); (L.L.-M.)
| |
Collapse
|
14
|
Joyner AL, Bayin NS. Cerebellum lineage allocation, morphogenesis and repair: impact of interplay amongst cells. Development 2022; 149:dev185587. [PMID: 36172987 PMCID: PMC9641654 DOI: 10.1242/dev.185587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The cerebellum has a simple cytoarchitecture consisting of a folded cortex with three cell layers that surrounds a nuclear structure housing the output neurons. The excitatory neurons are generated from a unique progenitor zone, the rhombic lip, whereas the inhibitory neurons and astrocytes are generated from the ventricular zone. The growth phase of the cerebellum is driven by lineage-restricted progenitor populations derived from each zone. Research during the past decade has uncovered the importance of cell-to-cell communication between the lineages through largely unknown signaling mechanisms for regulating the scaling of cell numbers and cell plasticity during mouse development and following injury in the neonatal (P0-P14) cerebellum. This Review focuses on how the interplay between cell types is key to morphogenesis, production of robust neural circuits and replenishment of cells after injury, and ends with a discussion of the implications of the greater complexity of the human cerebellar progenitor zones for development and disease.
Collapse
Affiliation(s)
- Alexandra L. Joyner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Biochemistry Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - N. Sumru Bayin
- Wellcome Trust/Cancer Research UK Gurdon Institute, Cambridge University, Cambridge CB2 1NQ, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
| |
Collapse
|
15
|
Gómez-González GB, Becerra-González M, Martínez-Mendoza ML, Rodríguez-Arzate CA, Martínez-Torres A. Organization of the ventricular zone of the cerebellum. Front Cell Neurosci 2022; 16:955550. [PMID: 35959470 PMCID: PMC9358289 DOI: 10.3389/fncel.2022.955550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/06/2022] [Indexed: 11/14/2022] Open
Abstract
The roof of the fourth ventricle (4V) is located on the ventral part of the cerebellum, a region with abundant vascularization and cell heterogeneity that includes tanycyte-like cells that define a peculiar glial niche known as ventromedial cord. This cord is composed of a group of biciliated cells that run along the midline, contacting the ventricular lumen and the subventricular zone. Although the complex morphology of the glial cells composing the cord resembles to tanycytes, cells which are known for its proliferative capacity, scarce or non-proliferative activity has been evidenced in this area. The subventricular zone of the cerebellum includes astrocytes, oligodendrocytes, and neurons whose function has not been extensively studied. This review describes to some extent the phenotypic, morphological, and functional characteristics of the cells that integrate the roof of the 4V, primarily from rodent brains.
Collapse
|
16
|
Schober AL, Wicki-Stordeur LE, Murai KK, Swayne LA. Foundations and implications of astrocyte heterogeneity during brain development and disease. Trends Neurosci 2022; 45:692-703. [PMID: 35879116 DOI: 10.1016/j.tins.2022.06.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/25/2022] [Accepted: 06/29/2022] [Indexed: 11/25/2022]
Abstract
Astrocytes play crucial roles in regulating brain circuit formation and physiology. Recent technological advances have revealed unprecedented levels of astrocyte diversity encompassing molecular, morphological, and functional differences. This diversification is initiated during embryonic specification events and (in rodents) continues into the early postnatal period where it overlaps with peak synapse development and circuit refinement. In fact, several lines of evidence suggest astrocyte diversity both influences and is a consequence of molecular crosstalk among developing astrocytes and other cell types, notably neurons and their synapses. Neurological disease states exhibit additional layers of astrocyte heterogeneity, which could help shed light on these cells' key pathological roles. This review highlights recent advances in clarifying astrocyte heterogeneity and molecular/cellular crosstalk and identifies key outstanding questions.
Collapse
Affiliation(s)
- Alexandra L Schober
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | | | - Keith K Murai
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada; Quantitative Life Sciences Graduate Program, McGill University, Montreal, QC, Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Cellular and Physiological Sciences, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
17
|
Region-Specific Characteristics of Astrocytes and Microglia: A Possible Involvement in Aging and Diseases. Cells 2022; 11:cells11121902. [PMID: 35741031 PMCID: PMC9220858 DOI: 10.3390/cells11121902] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Although different regions of the brain are dedicated to specific functions, the intra- and inter-regional heterogeneity of astrocytes and microglia in these regions has not yet been fully understood. Recently, an advancement in various technologies, such as single-cell RNA sequencing, has allowed for the discovery of astrocytes and microglia with distinct molecular fingerprints and varying functions in the brain. In addition, the regional heterogeneity of astrocytes and microglia exhibits different functions in several situations, such as aging and neurodegenerative diseases. Therefore, investigating the region-specific astrocytes and microglia is important in understanding the overall function of the brain. In this review, we summarize up-to-date research on various intra- and inter-regional heterogeneities of astrocytes and microglia, and provide information on how they can be applied to aging and neurodegenerative diseases.
Collapse
|
18
|
Serra I, Stravs A, Osório C, Oyaga MR, Schonewille M, Tudorache C, Badura A. Tsc1 Haploinsufficiency Leads to Pax2 Dysregulation in the Developing Murine Cerebellum. Front Mol Neurosci 2022; 15:831687. [PMID: 35645731 PMCID: PMC9137405 DOI: 10.3389/fnmol.2022.831687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/04/2022] [Indexed: 12/03/2022] Open
Abstract
Tuberous sclerosis complex 1 (TSC1) is a tumor suppressor that promotes the inhibition of mechanistic target of rapamycin (mTOR) pathway, and mutations in TSC1 lead to a rare complex disorder of the same name. Despite phenotype heterogeneity, up to 50% of TSC patients present with autism spectrum disorder (ASD). Consequently, TSC models are often used to probe molecular and behavioral mechanisms of ASD development. Amongst the different brain areas proposed to play a role in the development of ASD, the cerebellum is commonly reported to be altered, and cerebellar-specific deletion of Tsc1 in mice is sufficient to induce ASD-like phenotypes. However, despite these functional changes, whether Tsc1 haploinsufficiency affects cerebellar development is still largely unknown. Given that the mTOR pathway is a master regulator of cell replication and migration, we hypothesized that dysregulation of this pathway would also disrupt the development of cell populations during critical periods of cerebellar development. Here, we used a mouse model of TSC to investigate gene and protein expression during embryonic and early postnatal periods of cerebellar development. We found that, at E18 and P7, mRNA levels of the cerebellar inhibitory interneuron marker paired box gene 2 (Pax2) were dysregulated. This dysregulation was accompanied by changes in the expression of mTOR pathway-related genes and downstream phosphorylation of S6. Differential gene correlation analysis revealed dynamic changes in correlated gene pairs across development, with an overall loss of correlation between mTOR- and cerebellar-related genes in Tsc1 mutants compared to controls. We corroborated the genetic findings by characterizing the mTOR pathway and cerebellar development on protein and cellular levels with Western blot and immunohistochemistry. We found that Pax2-expressing cells were largely unchanged at E18 and P1, while at P7, their number was increased and maturation into parvalbumin-expressing cells delayed. Our findings indicate that, in mice, Tsc1 haploinsufficiency leads to altered cerebellar development and that cerebellar interneuron precursors are particularly susceptible to mTOR pathway dysregulation.
Collapse
Affiliation(s)
- Ines Serra
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Ana Stravs
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Catarina Osório
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Maria Roa Oyaga
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- *Correspondence: Aleksandra Badura,
| |
Collapse
|
19
|
Boda E, Boscia F, Lohr C. Editorial: The Role of Astroglia and Oligodendroglia in CNS Development, Plasticity, and Disease – Novel Tools and Investigative Approaches. Front Cell Neurosci 2022; 16:901820. [PMID: 35614969 PMCID: PMC9126039 DOI: 10.3389/fncel.2022.901820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
- *Correspondence: Enrica Boda
| | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
20
|
Boda E, Lorenzati M, Parolisi R, Harding B, Pallavicini G, Bonfanti L, Moccia A, Bielas S, Di Cunto F, Buffo A. Molecular and functional heterogeneity in dorsal and ventral oligodendrocyte progenitor cells of the mouse forebrain in response to DNA damage. Nat Commun 2022; 13:2331. [PMID: 35484145 PMCID: PMC9051058 DOI: 10.1038/s41467-022-30010-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/07/2022] [Indexed: 11/09/2022] Open
Abstract
In the developing mouse forebrain, temporally distinct waves of oligodendrocyte progenitor cells (OPCs) arise from different germinal zones and eventually populate either dorsal or ventral regions, where they present as transcriptionally and functionally equivalent cells. Despite that, developmental heterogeneity influences adult OPC responses upon demyelination. Here we show that accumulation of DNA damage due to ablation of citron-kinase or cisplatin treatment cell-autonomously disrupts OPC fate, resulting in cell death and senescence in the dorsal and ventral subsets, respectively. Such alternative fates are associated with distinct developmental origins of OPCs, and with a different activation of NRF2-mediated anti-oxidant responses. These data indicate that, upon injury, dorsal and ventral OPC subsets show functional and molecular diversity that can make them differentially vulnerable to pathological conditions associated with DNA damage.
Collapse
Affiliation(s)
- Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy.
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy.
| | - Martina Lorenzati
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Roberta Parolisi
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Brian Harding
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gianmarco Pallavicini
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Luca Bonfanti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Amanda Moccia
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Bielas
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ferdinando Di Cunto
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Regione Gonzole 10, IT-10043, Orbassano (Turin), Italy
| |
Collapse
|
21
|
Gagliano G, Monteverdi A, Casali S, Laforenza U, Gandini Wheeler-Kingshott CAM, D’Angelo E, Mapelli L. Non-Linear Frequency Dependence of Neurovascular Coupling in the Cerebellar Cortex Implies Vasodilation-Vasoconstriction Competition. Cells 2022; 11:1047. [PMID: 35326498 PMCID: PMC8947624 DOI: 10.3390/cells11061047] [Citation(s) in RCA: 3] [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: 02/08/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 01/28/2023] Open
Abstract
Neurovascular coupling (NVC) is the process associating local cerebral blood flow (CBF) to neuronal activity (NA). Although NVC provides the basis for the blood oxygen level dependent (BOLD) effect used in functional MRI (fMRI), the relationship between NVC and NA is still unclear. Since recent studies reported cerebellar non-linearities in BOLD signals during motor tasks execution, we investigated the NVC/NA relationship using a range of input frequencies in acute mouse cerebellar slices of vermis and hemisphere. The capillary diameter increased in response to mossy fiber activation in the 6-300 Hz range, with a marked inflection around 50 Hz (vermis) and 100 Hz (hemisphere). The corresponding NA was recorded using high-density multi-electrode arrays and correlated to capillary dynamics through a computational model dissecting the main components of granular layer activity. Here, NVC is known to involve a balance between the NMDAR-NO pathway driving vasodilation and the mGluRs-20HETE pathway driving vasoconstriction. Simulations showed that the NMDAR-mediated component of NA was sufficient to explain the time course of the capillary dilation but not its non-linear frequency dependence, suggesting that the mGluRs-20HETE pathway plays a role at intermediate frequencies. These parallel control pathways imply a vasodilation-vasoconstriction competition hypothesis that could adapt local hemodynamics at the microscale bearing implications for fMRI signals interpretation.
Collapse
Affiliation(s)
- Giuseppe Gagliano
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
| | - Anita Monteverdi
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Stefano Casali
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
| | - Umberto Laforenza
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Claudia A. M. Gandini Wheeler-Kingshott
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
- IRCCS Mondino Foundation, 27100 Pavia, Italy
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London WC1N3 BG, UK
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Lisa Mapelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (G.G.); (A.M.); (S.C.); (C.A.M.G.W.-K.)
| |
Collapse
|
22
|
Multicolor strategies for investigating clonal expansion and tissue plasticity. Cell Mol Life Sci 2022; 79:141. [PMID: 35187598 PMCID: PMC8858928 DOI: 10.1007/s00018-021-04077-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 09/27/2021] [Accepted: 10/14/2021] [Indexed: 12/20/2022]
Abstract
Understanding the generation of complexity in living organisms requires the use of lineage tracing tools at a multicellular scale. In this review, we describe the different multicolor strategies focusing on mouse models expressing several fluorescent reporter proteins, generated by classical (MADM, Brainbow and its multiple derivatives) or acute (StarTrack, CLoNe, MAGIC Markers, iOn, viral vectors) transgenesis. After detailing the multi-reporter genetic strategies that serve as a basis for the establishment of these multicolor mouse models, we briefly mention other animal and cellular models (zebrafish, chicken, drosophila, iPSC) that also rely on these constructs. Then, we highlight practical applications of multicolor mouse models to better understand organogenesis at single progenitor scale (clonal analyses) in the brain and briefly in several other tissues (intestine, skin, vascular, hematopoietic and immune systems). In addition, we detail the critical contribution of multicolor fate mapping strategies in apprehending the fine cellular choreography underlying tissue morphogenesis in several models with a particular focus on brain cytoarchitecture in health and diseases. Finally, we present the latest technological advances in multichannel and in-depth imaging, and automated analyses that enable to better exploit the large amount of data generated from multicolored tissues.
Collapse
|
23
|
Bayin NS, Mizrak D, Stephen DN, Lao Z, Sims PA, Joyner AL. Injury-induced ASCL1 expression orchestrates a transitory cell state required for repair of the neonatal cerebellum. SCIENCE ADVANCES 2021; 7:eabj1598. [PMID: 34878841 PMCID: PMC8654303 DOI: 10.1126/sciadv.abj1598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 10/20/2021] [Indexed: 05/23/2023]
Abstract
To understand repair processes, it is critical to identify the molecular foundations underlying progenitor diversity and plasticity. Upon injury to the neonatal cerebellum, a normally gliogenic nestin-expressing progenitor (NEP) in the Bergmann glia layer (BgL) undergoes adaptive reprograming to restore granule cell production. However, the cellular states and genes regulating the NEP fate switch are unknown. Using single-cell RNA sequencing and fate mapping, we defined molecular subtypes of NEPs and their lineages under homeostasis and repair. NEPs contain two major subtypes: Hopx+ astrogliogenic and Ascl1+ neurogenic NEPs that are further subdivided based on their location, lineage, and differentiation status. Upon injury, an Ascl1+ transitory cellular state arises from Hopx+ BgL-NEPs. Furthermore, mutational analysis revealed that induction of Ascl1 is required for adaptive reprogramming by orchestrating a glial-to-neural switch in vivo following injury. Thus, we provide molecular and cellular insights into context-dependent progenitor plasticity and repair mechanisms in the brain.
Collapse
Affiliation(s)
- N. Sumru Bayin
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Dogukan Mizrak
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Daniel N. Stephen
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Zhimin Lao
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Peter A. Sims
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biochemistry and Biophysics, Columbia University, New York, NY, USA
| | - Alexandra L. Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| |
Collapse
|
24
|
Ojalvo-Sanz AC, López-Mascaraque L. Gliogenic Potential of Single Pallial Radial Glial Cells in Lower Cortical Layers. Cells 2021; 10:3237. [PMID: 34831460 PMCID: PMC8621618 DOI: 10.3390/cells10113237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 01/16/2023] Open
Abstract
During embryonic development, progenitor cells are progressively restricted in their potential to generate different neural cells. A specific progenitor cell type, the radial glial cells, divides symmetrically and then asymmetrically to produce neurons, astrocytes, oligodendrocytes, and NG2-glia in the cerebral cortex. However, the potential of individual progenitors to form glial lineages remains poorly understood. To further investigate the cell progeny of single pallial GFAP-expressing progenitors, we used the in vivo genetic lineage-tracing method, the UbC-(GFAP-PB)-StarTrack. After targeting those progenitors in embryonic mice brains, we tracked their adult glial progeny in lower cortical layers. Clonal analyses revealed the presence of clones containing sibling cells of either a glial cell type (uniform clones) or two different glial cell types (mixed clones). Further, the clonal size and rostro-caudal cell dispersion of sibling cells differed depending on the cell type. We concluded that pallial E14 neural progenitors are a heterogeneous cell population with respect to which glial cell type they produce, as well as the clonal size of their cell progeny.
Collapse
Affiliation(s)
| | - Laura López-Mascaraque
- Cellular, Molecular and Developmental Neurobiology Department, Instituto Cajal-CSIC, 8002 Madrid, Spain;
| |
Collapse
|
25
|
Adebiyi OE, Omobowale TO, Abatan MO. Neurocognitive domains and neuropathological changes in experimental infection with Trypanosoma brucei brucei in Wistar rats. Heliyon 2021; 7:e08260. [PMID: 34765779 PMCID: PMC8571699 DOI: 10.1016/j.heliyon.2021.e08260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/29/2021] [Accepted: 10/22/2021] [Indexed: 01/05/2023] Open
Abstract
Trypanosoma brucei brucei causes animal trypanosomiasis in several vertebrates and human African trypanosomiasis. Previous studies have only explored the incidence, clinical symptoms, haematology and biochemical changes associated with the disease. The behavioral manipulation hypothesis posits that parasites alter the behavior of host to increase the reproductive abilities of such parasites. Hence, the present study was carried out to investigate changes in behavior and cognition following experimental infection of T. brucei brucei in rat model. This study involved two groups of animals (uninfected control and T. brucei infected) with 8 rats per group. After confirmation of parasitaemia in the infected rats both groups were assessed to investigate if infection led to behavioral alterations and neuropathological changes using the open field, social interaction and forelimb suspension tests. Immunohistochemistry was performed on brain tissues using glial fibrillary acidic protein and anticalbindin-D28k, antibodies. We demonstrated that T. brucei infection triggered a significant decrease in exploratory activity, anxiety-like behavior, altered recognition of social novelty and reduced hanging latency in the hanging wire test. Immunohistochemistry revealed significant astrocytosis, loss of dendritic spines and reduction of Purkinje cell layer of the cerebellum. These results demonstrate that T. brucei infection induce signs of anxiety, impaired motor co-ordination with degeneration and loss of Purkinje cells.
Collapse
Affiliation(s)
| | | | - Mathew Oluwole Abatan
- Department of Veterinary Pharmacology and Toxicology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
26
|
Sarropoulos I, Sepp M, Frömel R, Leiss K, Trost N, Leushkin E, Okonechnikov K, Joshi P, Giere P, Kutscher LM, Cardoso-Moreira M, Pfister SM, Kaessmann H. Developmental and evolutionary dynamics of cis-regulatory elements in mouse cerebellar cells. Science 2021; 373:eabg4696. [PMID: 34446581 PMCID: PMC7611596 DOI: 10.1126/science.abg4696] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022]
Abstract
Organ development is orchestrated by cell- and time-specific gene regulatory networks. In this study, we investigated the regulatory basis of mouse cerebellum development from early neurogenesis to adulthood. By acquiring snATAC-seq (single-nucleus assay for transposase accessible chromatin using sequencing) profiles for ~90,000 cells spanning 11 stages, we mapped cerebellar cell types and identified candidate cis-regulatory elements (CREs). We detected extensive spatiotemporal heterogeneity among progenitor cells and a gradual divergence in the regulatory programs of cerebellar neurons during differentiation. Comparisons to vertebrate genomes and snATAC-seq profiles for ∼20,000 cerebellar cells from the marsupial opossum revealed a shared decrease in CRE conservation during development and differentiation as well as differences in constraint between cell types. Our work delineates the developmental and evolutionary dynamics of gene regulation in cerebellar cells and provides insights into mammalian organ development.
Collapse
Affiliation(s)
- Ioannis Sarropoulos
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany.
| | - Mari Sepp
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany.
| | - Robert Frömel
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Kevin Leiss
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Nils Trost
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Evgeny Leushkin
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Konstantin Okonechnikov
- Hopp Children's Cancer Center (KiTZ) Heidelberg, Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), and German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Piyush Joshi
- Hopp Children's Cancer Center (KiTZ) Heidelberg, Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), and German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Peter Giere
- Museum für Naturkunde, Leibniz Institute for Evolution and Biodiversity Science, Berlin, Germany
| | - Lena M Kutscher
- Hopp Children's Cancer Center (KiTZ) Heidelberg, Developmental Origins of Pediatric Cancer Group, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Margarida Cardoso-Moreira
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
- Evolutionary Developmental Biology Laboratory, Francis Crick Institute, London NW1 1AT, UK
| | - Stefan M Pfister
- Hopp Children's Cancer Center (KiTZ) Heidelberg, Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), and German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany.
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Henrik Kaessmann
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany.
| |
Collapse
|
27
|
Kantzer CG, Parmigiani E, Cerrato V, Tomiuk S, Knauel M, Jungblut M, Buffo A, Bosio A. ACSA-2 and GLAST classify subpopulations of multipotent and glial-restricted cerebellar precursors. J Neurosci Res 2021; 99:2228-2249. [PMID: 34060113 PMCID: PMC8453861 DOI: 10.1002/jnr.24842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
The formation of the cerebellum is highly coordinated to obtain its characteristic morphology and all cerebellar cell types. During mouse postnatal development, cerebellar progenitors with astroglial‐like characteristics generate mainly astrocytes and oligodendrocytes. However, a subset of astroglial‐like progenitors found in the prospective white matter (PWM) produces astroglia and interneurons. Characterizing these cerebellar astroglia‐like progenitors and distinguishing their developmental fates is still elusive. Here, we reveal that astrocyte cell surface antigen‐2 (ACSA‐2), lately identified as ATPase, Na+/K+ transporting, beta 2 polypeptide, is expressed by glial precursors throughout postnatal cerebellar development. In contrast to common astrocyte markers, ACSA‐2 appears on PWM cells but is absent on Bergmann glia (BG) precursors. In the adult cerebellum, ACSA‐2 is broadly expressed extending to velate astrocytes in the granular layer, white matter astrocytes, and to a lesser extent to BG. Cell transplantation and transcriptomic analysis revealed that marker staining discriminates two postnatal progenitor pools. One subset is defined by the co‐expression of ACSA‐2 and GLAST and the expression of markers typical of parenchymal astrocytes. These are PWM precursors that are exclusively gliogenic. They produce predominantly white matter and granular layer astrocytes. Another subset is constituted by GLAST positive/ACSA‐2 negative precursors that express neurogenic and BG‐like progenitor genes. This population displays multipotency and gives rise to interneurons besides all glial types, including BG. In conclusion, this work reports about ACSA‐2, a marker that in combination with GLAST enables for the discrimination and isolation of multipotent and glia‐committed progenitors, which generate different types of cerebellar astrocytes.
Collapse
Affiliation(s)
- Christina Geraldine Kantzer
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany.,Department of Cell and Molecular Biology, Karolinska Institute, Solna, Sweden
| | - Elena Parmigiani
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy.,Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Stefan Tomiuk
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Michail Knauel
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | | | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, Turin, Italy.,Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Andreas Bosio
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| |
Collapse
|
28
|
Developmental Maturation of the Cerebellar White Matter-an Instructive Environment for Cerebellar Inhibitory Interneurons. THE CEREBELLUM 2020; 19:286-308. [PMID: 32002802 PMCID: PMC7082410 DOI: 10.1007/s12311-020-01111-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In the developing cerebellum, the nascent white matter (WM) serves as an instructive niche for cerebellar cortical inhibitory interneurons. As their Pax2 expressing precursors transit the emerging WM, their laminar fate is programmed. The source(s) and nature of the signals involved remain unknown. Here, we used immunocytochemistry to follow the cellular maturation of the murine cerebellar WM during this critical period. During the first few days of postnatal development, when most Pax2 expressing cells are formed and many of them reach the cerebellar gray matter, only microglial cells can be identified in the territories through which Pax2 cells migrate. From p4 onward, cells expressing the oligodendrocytic or astrocyte markers, CNP-1, MBP or GFAP, started to appear in the nascent WM. Expression of macroglial markers increased with cerebellar differentiation, yet deep nuclei remained GFAP-negative at all ages. The progressive spread of maturing glia did not correlate with the exit of Pax2 cells from the WM, as indicated by the extensive mingling of these cells up to p15. Whereas sonic hedgehog-associated p75NTR expression could be verified in granule cell precursors, postmitotic Pax2 cells are p75NTR negative at all ages analyzed. Thus, if Pax2 cells, like their precursors, are sensitive to sonic hedgehog, this does not affect their expression of p75NTR. Our findings document that subsequently generated sets of Pax2 expressing precursors of inhibitory cerebellar interneurons are confronted with a dynamically changing complement of cerebellar glia. The eventual identification of fate-defining pathways should profit from the covariation with glial maturation predicted by the present findings.
Collapse
|
29
|
Abstract
Astrocytes, initially described as merely support cells, are now known as a heterogeneous population of cells actively involved in a variety of biological functions such as: neuronal migration and differentiation; regulation of cerebral blood flow; metabolic control of extracellular potassium concentration; and modulation of synapse formation and elimination; among others. Cerebellar glial cells have been shown to play a significant role in proliferation, differentiation, migration, and synaptogenesis. However, less evidence is available about the role of neuron-astrocyte interactions during cerebellar development and their impact on diseases of the cerebellum. In this review, we will focus on the mechanisms underlying cellular interactions, specifically neuron-astrocyte interactions, during cerebellar development, function, and disease. We will discuss how cerebellar glia, astrocytes, and Bergmann glia play a fundamental role in several steps of cerebellar development, such as granule cell migration, axonal growth, neuronal differentiation, and synapse formation, and in diseases associated with the cerebellum. We will focus on how astrocytes and thyroid hormones impact cerebellar development. Furthermore, we will provide evidence of how growth factors secreted by glial cells, such as epidermal growth factor and transforming growth factors, control cerebellar organogenesis. Finally, we will argue that glia are a key mediator of cerebellar development and that identification of molecules and pathways involved in neuron-glia interactions may contribute to a better understanding of cerebellar development and associated disorders.
Collapse
|
30
|
Sánchez-González R, Figueres-Oñate M, Ojalvo-Sanz AC, López-Mascaraque L. Cell Progeny in the Olfactory Bulb After Targeting Specific Progenitors with Different UbC-StarTrack Approaches. Genes (Basel) 2020; 11:genes11030305. [PMID: 32183100 PMCID: PMC7140809 DOI: 10.3390/genes11030305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
The large phenotypic variation in the olfactory bulb may be related to heterogeneity in the progenitor cells. Accordingly, the progeny of subventricular zone (SVZ) progenitor cells that are destined for the olfactory bulb is of particular interest, specifically as there are many facets of these progenitors and their molecular profiles remain unknown. Using modified StarTrack genetic tracing strategies, specific SVZ progenitor cells were targeted in E12 mice embryos, and the cell fate of these neural progenitors was determined in the adult olfactory bulb. This study defined the distribution and the phenotypic diversity of olfactory bulb interneurons from specific SVZ-progenitor cells, focusing on their spatial pallial origin, heterogeneity, and genetic profile.
Collapse
|
31
|
Cerebellar Astrocytes: Much More Than Passive Bystanders In Ataxia Pathophysiology. J Clin Med 2020; 9:jcm9030757. [PMID: 32168822 PMCID: PMC7141261 DOI: 10.3390/jcm9030757] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/22/2022] Open
Abstract
Ataxia is a neurodegenerative syndrome, which can emerge as a major element of a disease or represent a symptom of more complex multisystemic disorders. It comprises several forms with a highly variegated etiology, mainly united by motor, balance, and speech impairments and, at the tissue level, by cerebellar atrophy and Purkinje cells degeneration. For this reason, the contribution of astrocytes to this disease has been largely overlooked in the past. Nevertheless, in the last few decades, growing evidences are pointing to cerebellar astrocytes as crucial players not only in the progression but also in the onset of distinct forms of ataxia. Although the current knowledge on this topic is very fragmentary and ataxia type-specific, the present review will attempt to provide a comprehensive view of astrocytes’ involvement across the distinct forms of this pathology. Here, it will be highlighted how, through consecutive stage-specific mechanisms, astrocytes can lead to non-cell autonomous neurodegeneration and, consequently, to the behavioral impairments typical of this disease. In light of that, treating astrocytes to heal neurons will be discussed as a potential complementary therapeutic approach for ataxic patients, a crucial point provided the absence of conclusive treatments for this disease.
Collapse
|
32
|
Willett RT, Bayin NS, Lee AS, Krishnamurthy A, Wojcinski A, Lao Z, Stephen D, Rosello-Diez A, Dauber-Decker KL, Orvis GD, Wu Z, Tessier-Lavigne M, Joyner AL. Cerebellar nuclei excitatory neurons regulate developmental scaling of presynaptic Purkinje cell number and organ growth. eLife 2019; 8:e50617. [PMID: 31742552 PMCID: PMC6890462 DOI: 10.7554/elife.50617] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/18/2019] [Indexed: 01/17/2023] Open
Abstract
For neural systems to function effectively, the numbers of each cell type must be proportioned properly during development. We found that conditional knockout of the mouse homeobox genes En1 and En2 in the excitatory cerebellar nuclei neurons (eCN) leads to reduced postnatal growth of the cerebellar cortex. A subset of medial and intermediate eCN are lost in the mutants, with an associated cell non-autonomous loss of their presynaptic partner Purkinje cells by birth leading to proportional scaling down of neuron production in the postnatal cerebellar cortex. Genetic killing of embryonic eCN throughout the cerebellum also leads to loss of Purkinje cells and reduced postnatal growth but throughout the cerebellar cortex. Thus, the eCN play a key role in scaling the size of the cerebellum by influencing the survival of their Purkinje cell partners, which in turn regulate production of granule cells and interneurons via the amount of sonic hedgehog secreted.
Collapse
Affiliation(s)
- Ryan T Willett
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
| | - N Sumru Bayin
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
| | - Andrew S Lee
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
- Neuroscience ProgramWeill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Anjana Krishnamurthy
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
- Neuroscience ProgramWeill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | | | - Zhimin Lao
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
| | - Daniel Stephen
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
| | | | | | - Grant D Orvis
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
| | - Zhuhao Wu
- The Laboratory of Brain Development and RepairThe Rockefeller UniversityNew YorkUnited States
| | - Marc Tessier-Lavigne
- The Laboratory of Brain Development and RepairThe Rockefeller UniversityNew YorkUnited States
| | - Alexandra L Joyner
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
- Neuroscience ProgramWeill Cornell Graduate School of Medical SciencesNew YorkUnited States
- Biochemistry, Cell and Molecular Biology ProgramWeill Cornell Graduate School of Medical SciencesNew YorkUnited States
| |
Collapse
|
33
|
Cortical astrocytes develop in a plastic manner at both clonal and cellular levels. Nat Commun 2019; 10:4884. [PMID: 31653848 PMCID: PMC6814723 DOI: 10.1038/s41467-019-12791-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
Astrocytes play essential roles in the neural tissue where they form a continuous network, while displaying important local heterogeneity. Here, we performed multiclonal lineage tracing using combinatorial genetic markers together with a new large volume color imaging approach to study astrocyte development in the mouse cortex. We show that cortical astrocyte clones intermix with their neighbors and display extensive variability in terms of spatial organization, number and subtypes of cells generated. Clones develop through 3D spatial dispersion, while at the individual level astrocytes acquire progressively their complex morphology. Furthermore, we find that the astroglial network is supplied both before and after birth by ventricular progenitors that scatter in the neocortex and can give rise to protoplasmic as well as pial astrocyte subtypes. Altogether, these data suggest a model in which astrocyte precursors colonize the neocortex perinatally in a non-ordered manner, with local environment likely determining astrocyte clonal expansion and final morphotype. Previous studies on astrocyte development have led to controversial results due to a lack of pertinent tools. Here, authors analyze large numbers of astrocyte clones generated by nearby cortical progenitors using the MAGIC Markers strategy and ChroMS 3D imaging, and show that clonally-related astrocytes organize in a non-stereotyped manner and that cortical astrocyte subtypes are not intrinsically specified.
Collapse
|
34
|
Figueres-Oñate M, Sánchez-Villalón M, Sánchez-González R, López-Mascaraque L. Lineage Tracing and Cell Potential of Postnatal Single Progenitor Cells In Vivo. Stem Cell Reports 2019; 13:700-712. [PMID: 31543472 PMCID: PMC6829765 DOI: 10.1016/j.stemcr.2019.08.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 01/01/2023] Open
Abstract
Understanding the contribution of adult neural progenitor cells (NPCs) and their lineage potential is a great challenge in neuroscience. To reveal progenitor diversity and cell-lineage relationships of postnatal NPCs in the subventricular zone (SVZ), we performed in vivo lineage-tracing genetic analysis using the UbC-StarTrack. We determined the progeny of single SVZ-NPCs, the number of cells per clone, the dispersion of sibling cells, and the cell types within clones. Long-term analysis revealed that both the cell-dispersion pattern and number of cells comprising clones varied depending on the glial/neuronal nature of sibling cells. Sibling-olfactory interneurons were primarily located within the same layer, while sibling-glial cells populated SVZ-adjacent areas. Sibling astrocytes and interneurons did not form big clones, whereas oligodendroglial-lineage clones comprised the largest clones originated in adult brains. These results demonstrate the existence of SVZ postnatal bipotential progenitors that give rise to clones widely dispersed across the olfactory bulb and SVZ-adjacent areas. Bipotent postnatal progenitors produce clones of olfactory neurons and glial cells Different clonal cell patterns in astroglial, oligodendroglial, and neuronal lineages Sibling neuroblasts migrating to the olfactory bulb widespread along the RMS axis Sibling astrocytes and interneurons form discrete cell clones
Collapse
|
35
|
Yang Z, Joyner AL. YAP1 is involved in replenishment of granule cell precursors following injury to the neonatal cerebellum. Dev Biol 2019; 455:458-472. [PMID: 31376393 DOI: 10.1016/j.ydbio.2019.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 01/08/2023]
Abstract
The cerebellum undergoes major rapid growth during the third trimester and early neonatal stage in humans, making it vulnerable to injuries in pre-term babies. Experiments in mice have revealed a remarkable ability of the neonatal cerebellum to recover from injuries around birth. In particular, recovery following irradiation-induced ablation of granule cell precursors (GCPs) involves adaptive reprogramming of Nestin-expressing glial progenitors (NEPs). Sonic hedgehog signaling is required for the initial step in NEP reprogramming; however, the full spectrum of developmental signaling pathways that promote NEP-driven regeneration is not known. Since the growth regulatory Hippo pathway has been implicated in the repair of several tissue types, we tested whether Hippo signaling is involved in regeneration of the cerebellum. Using mouse models, we found that the Hippo pathway transcriptional co-activator YAP1 (Yes-associated protein 1) but not TAZ (transcriptional coactivator with PDZ binding motif, or WWTR1) is required in NEPs for full recovery of cerebellar growth following irradiation one day after birth. Although Yap1 plays only a minor role during normal development in differentiation of NEPs or GCPs, the size of the cerebellum, and in particular the internal granule cell layer produced by GCPs, is significantly reduced in Yap1 mutants after irradiation, and the organization of Purkinje cells and Bergmann glial fibers is disrupted. The initial proliferative response of Yap1 mutant NEPs to irradiation is normal and the cells migrate to the GCP niche, but subsequently there is increased cell death of GCPs and altered migration of granule cells, possibly due to defects in Bergmann glia. Moreover, loss of Taz along with Yap1 in NEPs does not abrogate regeneration or alter development of the cerebellum. Our study provides new insights into the molecular signaling underlying postnatal cerebellar development and regeneration.
Collapse
Affiliation(s)
- Zhaohui Yang
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, 10065, United States; Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, United States
| | - Alexandra L Joyner
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, 10065, United States; Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, United States.
| |
Collapse
|
36
|
Salvi M, Cerrato V, Buffo A, Molinari F. Automated segmentation of brain cells for clonal analyses in fluorescence microscopy images. J Neurosci Methods 2019; 325:108348. [PMID: 31283938 DOI: 10.1016/j.jneumeth.2019.108348] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 11/26/2022]
Abstract
The understanding of how cell diversity within and across distinct brain regions is ontogenetically achieved is a pivotal topic in neuroscience. Clonal analyses based on multicolor cell labeling represent a powerful tool to tackle this issue and disclose lineage relationships, but produce enormous sets of fluorescence images, leading to time consuming analyses that may be biased by the operator's subjectivity. Thus, time-efficient automated software are needed to analyze images easily, accurately and without subjective bias. In this paper, we present a fully automated method, named FAST ('Fluorescent cell Analysis Segmentation Tool'), for the segmentation of neural cells labeled by multicolor combinations of fluorophores and for their classification into clones. The proposed method was tested on 77 high-magnification fluorescence images of adult mouse cerebellar tissues acquired using a confocal microscope. Automatic results were compared with manual annotations and two open-source software designed for cell detection in microscopic imaging. The algorithm showed very good performance in the cellular detection and in the assignment of the clonal identity. To the best of our knowledge, FAST is the first fully automated technique for the analysis of cellular clones based on combinatorial expression of fluorescent proteins. The proposed approach allows to perform clonal analyses easily, accurately and objectively, overcoming those biases and errors that may result from manual annotations. Moreover, it can be broadly applied to the quantification and colocalization within cells of fluorescent markers, therefore representing a versatile and powerful tool for automated quantitative analyses in fluorescence microscopy.
Collapse
Affiliation(s)
- Massimo Salvi
- Biolab, Department of Electronics and Telecommunications, Politecnico di Torino, Turin, Italy.
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin and Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Turin, Italy.
| | - Annalisa Buffo
- Department of Neuroscience Rita Levi-Montalcini, University of Turin and Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Turin, Italy.
| | - Filippo Molinari
- Biolab, Department of Electronics and Telecommunications, Politecnico di Torino, Turin, Italy.
| |
Collapse
|