101
|
Le Q, Yan B, Yu X, Li Y, Song H, Zhu H, Hou W, Ma D, Wu F, Zhou Y, Ma L. Drug-seeking motivation level in male rats determines offspring susceptibility or resistance to cocaine-seeking behaviour. Nat Commun 2017; 8:15527. [PMID: 28556835 PMCID: PMC5459992 DOI: 10.1038/ncomms15527] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 04/05/2017] [Indexed: 12/17/2022] Open
Abstract
Liability to develop drug addiction is heritable, but the precise contribution of non-Mendelian factors is not well understood. Here we separate male rats into addiction-like and non-addiction-like groups, based on their incentive motivation to seek cocaine. We find that the high incentive responding of the F0 generation could be transmitted to F1 and F2 generations. Moreover, the inheritance of high incentive response to cocaine is contingent on high motivation, as it is elicited by voluntary cocaine administration, but not high intake of cocaine itself. We also find DNA methylation differences between sperm of addiction-like and non-addiction-like groups that were maintained from F0 to F1, providing an epigenetic link to transcriptomic changes of addiction-related signalling pathways in the nucleus accumbens of offspring. Our data suggest that highly motivated drug seeking experience may increase vulnerability and/or reduce resistance to drug addiction in descendants. Drug addiction is partially heritable but the non-genetic inheritance mechanisms are not well understood. The authors show that motivation of male rats in response to cocaine self-administration elicit susceptibility and/or decreased resistance to developing addiction like behaviour in offspring.
Collapse
Affiliation(s)
- Qiumin Le
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Biao Yan
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Xiangchen Yu
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yanqing Li
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Haikun Song
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Huiwen Zhu
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Weiqing Hou
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Dingailu Ma
- Laboratory of Epigenetics, Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Feizhen Wu
- Laboratory of Epigenetics, Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yuqing Zhou
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Lan Ma
- Department of Neurosurgery, and Institute of Translational Neuroscience, Huashan Hospital, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
102
|
Minovi A, Aguado A, Brunert D, Kurtenbach S, Dazert S, Hatt H, Conrad H. Isolation, culture optimization and functional characterization of stem cell neurospheres from mouse neonatal olfactory bulb and epithelium. Eur Arch Otorhinolaryngol 2017; 274:3071-3085. [PMID: 28478501 DOI: 10.1007/s00405-017-4590-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
Abstract
The olfactory epithelium contains basal cells with stem cell characteristics, which have the capacity to differentiate throughout life into olfactory receptor neurons (ORNs). Here we investigate the in vitro characteristics of stem cells taken from the olfactory bulb (OB) and the olfactory epithelium (OE) of neonatal TIS21 knock-in mice. The major aim of the study was the generation of olfactory neurospheres (ONS) derived from OB and OE of neonatal mice as a tool to further analyze the elementary processes of ORN development. Our data showed that the presence of epidermal growth factor (EGF) and fibroblast growth factor (FGF) leads to a significant increase in number of ONS derived from OB but not from OE. The differentiation of ONSs led to the formation of different neuronal cell types, in particular to bipolar-shaped cells as well as putative pyramidal-neurons, astrocytes and oligodendrocytes. Immunohistochemical staining confirmed the presence of astrocytes and neurons in both types of ONSs. In order to investigate the functionality of the neurons we performed calcium imaging and patch-clamp experiments. Calcium imaging experiments revealed that the application of high potassium concentration provokes calcium transients. No excitable properties, neither sodium currents nor action potentials, were observed for the bipolar-shaped cells derived from OB and OE neurospheres, which means that these types of cells morphologically defined as putative neuronal cells, were not physiologically active. Interestingly, patch-clamp recordings performed in the pyramidal-shaped cells of OB neurospheres showed sodium and potassium currents as well as action potentials. Our study will help to establish further models in the field of olfactology.
Collapse
Affiliation(s)
- Amir Minovi
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany.
| | - Ainhara Aguado
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany.,Department of Cell Physiology, Ruhr-University Bochum, Universitätstrasse 150, 44801, Bochum, Germany
| | - Daniela Brunert
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074, Aachen, Germany
| | - Stefan Kurtenbach
- Department of Cell Physiology, Ruhr-University Bochum, Universitätstrasse 150, 44801, Bochum, Germany
| | - Stefan Dazert
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr-University Bochum, Universitätstrasse 150, 44801, Bochum, Germany
| | - Heike Conrad
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| |
Collapse
|
103
|
SDF-1/CXCR4 Signaling Maintains Stemness Signature in Mouse Neural Stem/Progenitor Cells. Stem Cells Int 2017; 2017:2493752. [PMID: 28408934 PMCID: PMC5376953 DOI: 10.1155/2017/2493752] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 01/29/2017] [Accepted: 02/01/2017] [Indexed: 11/17/2022] Open
Abstract
SDF-1 and its primary receptor, CXCR4, are highly expressed in the embryonic central nervous system (CNS) and play a crucial role in brain architecture. Loss of SDF-1/CXCR4 signaling causes abnormal development of neural stem/progenitor cells (NSCs/NPCs) in the cerebellum, hippocampus, and cortex. However, the mechanism of SDF-1/CXCR4 axis in NSCs/NPCs regulation remains unknown. In this study, we found that elimination of SDF-1/CXCR4 transduction caused NSCs/NPCs to lose their stemness characteristics and to encounter neurogenic differentiation. Moreover, Notch and RE1 silencing transcription factor (REST) both play an essential role in NSCs/NPCs maintenance and neuronal differentiation and were dramatically downregulated following SDF-1/CXCR4 cascade inhibition. Finally, we demonstrated that the expression of achaete-scute homolog 1 (Ascl1), a proneural gene, and p27, an antiproliferative gene, were significantly increased after genetic elimination of SDF-1 alleles. Our results support that the loss of functional SDF-1/CXCR4 signaling pathway in NSCs/NPCs induces exit of cell cycle and promotes premature neural differentiation.
Collapse
|
104
|
Fong BC, Slack RS. RB: An essential player in adult neurogenesis. NEUROGENESIS 2017; 4:e1270382. [PMID: 28229086 DOI: 10.1080/23262133.2016.1270382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/30/2016] [Accepted: 12/04/2016] [Indexed: 12/16/2022]
Abstract
The fundamental mechanisms underlying adult neurogenesis remain to be fully clarified. Members of the cell cycle machinery have demonstrated key roles in regulating adult neural stem cell (NSC) quiescence and the size of the adult-born neuronal population. The retinoblastoma protein, Rb, is known to possess CNS-specific requirements that are independent from its classical role as a tumor suppressor. The recent study by Vandenbosch et al. has clarified distinct requirements for Rb during adult neurogenesis, in the restriction of proliferation, as well as long-term adult-born neuronal survival. However, Rb is no longer believed to be the main cell cycle regulator maintaining the quiescence of adult NSCs. Future studies must consider Rb as part of a larger network of regulatory effectors, including the other members of the Rb family, p107 and p130. This will help elucidate the contribution of Rb and other pocket proteins in the context of adult neurogenesis, and define its crucial role in regulating the size and fate of the neurogenic niche.
Collapse
Affiliation(s)
- Bensun C Fong
- University of Ottawa Brain and Mind Research Institute, Department of Cellular & Molecular Medicine, University of Ottawa , Ottawa, ON, Canada
| | - Ruth S Slack
- University of Ottawa Brain and Mind Research Institute, Department of Cellular & Molecular Medicine, University of Ottawa , Ottawa, ON, Canada
| |
Collapse
|
105
|
Giandomenico SL, Lancaster MA. Probing human brain evolution and development in organoids. Curr Opin Cell Biol 2017; 44:36-43. [DOI: 10.1016/j.ceb.2017.01.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 12/23/2016] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
|
106
|
Meyer HM, Teles J, Formosa-Jordan P, Refahi Y, San-Bento R, Ingram G, Jönsson H, Locke JCW, Roeder AHK. Fluctuations of the transcription factor ATML1 generate the pattern of giant cells in the Arabidopsis sepal. eLife 2017; 6:e19131. [PMID: 28145865 PMCID: PMC5333958 DOI: 10.7554/elife.19131] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 01/31/2017] [Indexed: 12/22/2022] Open
Abstract
Multicellular development produces patterns of specialized cell types. Yet, it is often unclear how individual cells within a field of identical cells initiate the patterning process. Using live imaging, quantitative image analyses and modeling, we show that during Arabidopsis thaliana sepal development, fluctuations in the concentration of the transcription factor ATML1 pattern a field of identical epidermal cells to differentiate into giant cells interspersed between smaller cells. We find that ATML1 is expressed in all epidermal cells. However, its level fluctuates in each of these cells. If ATML1 levels surpass a threshold during the G2 phase of the cell cycle, the cell will likely enter a state of endoreduplication and become giant. Otherwise, the cell divides. Our results demonstrate a fluctuation-driven patterning mechanism for how cell fate decisions can be initiated through a random yet tightly regulated process.
Collapse
Affiliation(s)
- Heather M Meyer
- Weill Institute for Cell and Molecular Biology, Cornell University, United States
- The graduate field of Genetics, Genomics, and Development, Cornell University, Ithaca, United States
| | - José Teles
- Sainsbury Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Pau Formosa-Jordan
- Sainsbury Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Yassin Refahi
- Sainsbury Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Rita San-Bento
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRA, Lyon, France
| | - Gwyneth Ingram
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRA, Lyon, France
| | - Henrik Jönsson
- Sainsbury Laboratory, University of Cambridge, Cambridge, United Kingdom
- Computational Biology and Biological Physics, Lund University, Lund, Sweden
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
| | - James C W Locke
- Sainsbury Laboratory, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Microsoft Research, Cambridge, United Kingdom
| | - Adrienne H K Roeder
- Weill Institute for Cell and Molecular Biology, Cornell University, United States
- The graduate field of Genetics, Genomics, and Development, Cornell University, Ithaca, United States
- Section of Plant Biology, School of Integrative Plant Science, Cornell University, Ithaca, United States
| |
Collapse
|
107
|
Recent Zika Virus Isolates Induce Premature Differentiation of Neural Progenitors in Human Brain Organoids. Cell Stem Cell 2017; 20:397-406.e5. [PMID: 28132835 DOI: 10.1016/j.stem.2016.12.005] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/05/2016] [Accepted: 12/14/2016] [Indexed: 01/12/2023]
Abstract
The recent Zika virus (ZIKV) epidemic is associated with microcephaly in newborns. Although the connection between ZIKV and neurodevelopmental defects is widely recognized, the underlying mechanisms are poorly understood. Here we show that two recently isolated strains of ZIKV, an American strain from an infected fetal brain (FB-GWUH-2016) and a closely-related Asian strain (H/PF/2013), productively infect human iPSC-derived brain organoids. Both of these strains readily target to and replicate in proliferating ventricular zone (VZ) apical progenitors. The main phenotypic effect was premature differentiation of neural progenitors associated with centrosome perturbation, even during early stages of infection, leading to progenitor depletion, disruption of the VZ, impaired neurogenesis, and cortical thinning. The infection pattern and cellular outcome differ from those seen with the extensively passaged ZIKV strain MR766. The structural changes we see after infection with these more recently isolated viral strains closely resemble those seen in ZIKV-associated microcephaly.
Collapse
|
108
|
Dietz KC, Polanco JJ, Pol SU, Sim FJ. Targeting human oligodendrocyte progenitors for myelin repair. Exp Neurol 2016; 283:489-500. [PMID: 27001544 PMCID: PMC5666574 DOI: 10.1016/j.expneurol.2016.03.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/31/2022]
Abstract
Oligodendrocyte development has been studied for several decades, and has served as a model system for both neurodevelopmental and stem/progenitor cell biology. Until recently, the vast majority of studies have been conducted in lower species, especially those focused on rodent development and remyelination. In humans, the process of myelination requires the generation of vastly more myelinating glia, occurring over a period of years rather than weeks. Furthermore, as evidenced by the presence of chronic demyelination in a variety of human neurologic diseases, it appears likely that the mechanisms that regulate development and become dysfunctional in disease may be, in key ways, divergent across species. Improvements in isolation techniques, applied to primary human neural and oligodendrocyte progenitors from both fetal and adult brain, as well as advancements in the derivation of defined progenitors from human pluripotent stem cells, have begun to reveal the extent of both species-conserved signaling pathways and potential key differences at cellular and molecular levels. In this article, we will review the commonalities and differences in myelin development between rodents and man, describing the approaches used to study human oligodendrocyte differentiation and myelination, as well as heterogeneity within targetable progenitor pools, and discuss the advances made in determining which conserved pathways may be both modeled in rodents and translate into viable therapeutic strategies to promote myelin repair.
Collapse
Affiliation(s)
- Karen C Dietz
- Program in Neuroscience, Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, 3435 Main Street, 119 Farber Hall, Buffalo, NY 14214, United States.
| | - Jessie J Polanco
- Program in Neuroscience, Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, 3435 Main Street, 119 Farber Hall, Buffalo, NY 14214, United States.
| | - Suyog U Pol
- Program in Neuroscience, Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, 3435 Main Street, 119 Farber Hall, Buffalo, NY 14214, United States.
| | - Fraser J Sim
- Program in Neuroscience, Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, 3435 Main Street, 119 Farber Hall, Buffalo, NY 14214, United States.
| |
Collapse
|
109
|
Gai M, Bianchi FT, Vagnoni C, Vernì F, Bonaccorsi S, Pasquero S, Berto GE, Sgrò F, Chiotto AM, Annaratone L, Sapino A, Bergo A, Landsberger N, Bond J, Huttner WB, Di Cunto F. ASPM and CITK regulate spindle orientation by affecting the dynamics of astral microtubules. EMBO Rep 2016; 17:1396-1409. [PMID: 27562601 DOI: 10.15252/embr.201541823] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 07/26/2016] [Indexed: 11/09/2022] Open
Abstract
Correct orientation of cell division is considered an important factor for the achievement of normal brain size, as mutations in genes that affect this process are among the leading causes of microcephaly. Abnormal spindle orientation is associated with reduction of the neuronal progenitor symmetric divisions, premature cell cycle exit, and reduced neurogenesis. This mechanism has been involved in microcephaly resulting from mutation of ASPM, the most frequently affected gene in autosomal recessive human primary microcephaly (MCPH), but it is presently unknown how ASPM regulates spindle orientation. In this report, we show that ASPM may control spindle positioning by interacting with citron kinase (CITK), a protein whose loss is also responsible for severe microcephaly in mammals. We show that the absence of CITK leads to abnormal spindle orientation in mammals and insects. In mouse cortical development, this phenotype correlates with increased production of basal progenitors. ASPM is required to recruit CITK at the spindle, and CITK overexpression rescues ASPM phenotype. ASPM and CITK affect the organization of astral microtubules (MT), and low doses of MT-stabilizing drug revert the spindle orientation phenotype produced by their knockdown. Finally, CITK regulates both astral-MT nucleation and stability. Our results provide a functional link between two established microcephaly proteins.
Collapse
Affiliation(s)
- Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Federico T Bianchi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Cristiana Vagnoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnologies "C. Darwin", Sapienza, Università di Roma, Rome, Italy
| | - Silvia Bonaccorsi
- Department of Biology and Biotechnologies "C. Darwin", Sapienza, Università di Roma, Rome, Italy
| | - Selina Pasquero
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Gaia E Berto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Francesco Sgrò
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandra Ma Chiotto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Laura Annaratone
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Bergo
- San Raffaele Rett Research Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Nicoletta Landsberger
- San Raffaele Rett Research Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Jacqueline Bond
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | - Wieland B Huttner
- Max-Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ferdinando Di Cunto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
110
|
Pillat MM, Lameu C, Trujillo CA, Glaser T, Cappellari AR, Negraes PD, Battastini AMO, Schwindt TT, Muotri AR, Ulrich H. Bradykinin promotes neuron-generating division of neural progenitor cells through ERK activation. J Cell Sci 2016; 129:3437-48. [PMID: 27528403 DOI: 10.1242/jcs.192534] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
During brain development, cells proliferate, migrate and differentiate in highly accurate patterns. In this context, published results indicate that bradykinin functions in neural fate determination, favoring neurogenesis and migration. However, mechanisms underlying bradykinin function are yet to be explored. Our findings indicate a previously unidentified role for bradykinin action in inducing neuron-generating division in vitro and in vivo, given that bradykinin lengthened the G1-phase of the neural progenitor cells (NPC) cycle and increased TIS21 (also known as PC3 and BTG2) expression in hippocampus from newborn mice. This role, triggered by activation of the kinin-B2 receptor, was conditioned by ERK1/2 activation. Moreover, immunohistochemistry analysis of hippocampal dentate gyrus showed that the percentage of Ki67(+) cells markedly increased in bradykinin-treated mice, and ERK1/2 inhibition affected this neurogenic response. The progress of neurogenesis depended on sustained ERK phosphorylation and resulted in ERK1/2 translocation to the nucleus in NPCs and PC12 cells, changing expression of genes such as Hes1 and Ngn2 (also known as Neurog2). In agreement with the function of ERK in integrating signaling pathways, effects of bradykinin in stimulating neurogenesis were reversed following removal of protein kinase C (PKC)-mediated sustained phosphorylation.
Collapse
Affiliation(s)
- Micheli M Pillat
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Claudiana Lameu
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Cleber A Trujillo
- Departments of Pediatrics and Cellular & Molecular Medicine, University of California San Diego, San Diego, CA 92093-0695, USA
| | - Talita Glaser
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Angélica R Cappellari
- Departamento de Bioquímica, Instituto de Ciências Básicas e da Saúde, UFRGS, Porto Alegre 90035 000, Brazil
| | - Priscilla D Negraes
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil Departments of Pediatrics and Cellular & Molecular Medicine, University of California San Diego, San Diego, CA 92093-0695, USA
| | - Ana M O Battastini
- Departamento de Bioquímica, Instituto de Ciências Básicas e da Saúde, UFRGS, Porto Alegre 90035 000, Brazil
| | - Telma T Schwindt
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Alysson R Muotri
- Departments of Pediatrics and Cellular & Molecular Medicine, University of California San Diego, San Diego, CA 92093-0695, USA
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
111
|
Shimada M, Matsuzaki F, Kato A, Kobayashi J, Matsumoto T, Komatsu K. Induction of Excess Centrosomes in Neural Progenitor Cells during the Development of Radiation-Induced Microcephaly. PLoS One 2016; 11:e0158236. [PMID: 27367050 PMCID: PMC4930206 DOI: 10.1371/journal.pone.0158236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/12/2016] [Indexed: 11/19/2022] Open
Abstract
The embryonic brain is one of the tissues most vulnerable to ionizing radiation. In this study, we showed that ionizing radiation induces apoptosis in the neural progenitors of the mouse cerebral cortex, and that the surviving progenitor cells subsequently develop a considerable amount of supernumerary centrosomes. When mouse embryos at Day 13.5 were exposed to γ-rays, brains sizes were reduced markedly in a dose-dependent manner, and these size reductions persisted until birth. Immunostaining with caspase-3 antibodies showed that apoptosis occurred in 35% and 40% of neural progenitor cells at 4 h after exposure to 1 and 2 Gy, respectively, and this was accompanied by a disruption of the apical layer in which mitotic spindles were positioned in unirradiated mice. At 24 h after 1 Gy irradiation, the apoptotic cells were completely eliminated and proliferation was restored to a level similar to that of unirradiated cells, but numerous spindles were localized outside the apical layer. Similarly, abnormal cytokinesis, which included multipolar division and centrosome clustering, was observed in 19% and 24% of the surviving neural progenitor cells at 48 h after irradiation with 1 and 2 Gy, respectively. Because these cytokinesis aberrations derived from excess centrosomes result in growth delay and mitotic catastrophe-mediated cell elimination, our findings suggest that, in addition to apoptosis at an early stage of radiation exposure, radiation-induced centrosome overduplication could contribute to the depletion of neural progenitors and thereby lead to microcephaly.
Collapse
Affiliation(s)
- Mikio Shimada
- Department of Genome Repair Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, Center for Developmental Biology, RIKEN, Kobe, Japan
| | - Akihiro Kato
- Department of Genome Repair Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Junya Kobayashi
- Department of Genome Repair Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Tomohiro Matsumoto
- Department of Radiation System Biology, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Kenshi Komatsu
- Department of Genome Repair Dynamics, Radiation Biology Center, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
112
|
Ghosh S, Hui SP. Regeneration of Zebrafish CNS: Adult Neurogenesis. Neural Plast 2016; 2016:5815439. [PMID: 27382491 PMCID: PMC4921647 DOI: 10.1155/2016/5815439] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
Regeneration in the animal kingdom is one of the most fascinating problems that have allowed scientists to address many issues of fundamental importance in basic biology. However, we came to know that the regenerative capability may vary across different species. Among vertebrates, fish and amphibians are capable of regenerating a variety of complex organs through epimorphosis. Zebrafish is an excellent animal model, which can repair several organs like damaged retina, severed spinal cord, injured brain and heart, and amputated fins. The focus of the present paper is on spinal cord regeneration in adult zebrafish. We intend to discuss our current understanding of the cellular and molecular mechanism(s) that allows formation of proliferating progenitors and controls neurogenesis, which involve changes in epigenetic and transcription programs. Unlike mammals, zebrafish retains radial glia, a nonneuronal cell type in their adult central nervous system. Injury induced proliferation involves radial glia which proliferate, transcribe embryonic genes, and can give rise to new neurons. Recent technological development of exquisite molecular tools in zebrafish, such as cell ablation, lineage analysis, and novel and substantial microarray, together with advancement in stem cell biology, allowed us to investigate how progenitor cells contribute to the generation of appropriate structures and various underlying mechanisms like reprogramming.
Collapse
Affiliation(s)
- Sukla Ghosh
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Subhra Prakash Hui
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| |
Collapse
|
113
|
Katsukawa M, Nakajima Y, Fukumoto A, Doi D, Takahashi J. Fail-Safe Therapy by Gamma-Ray Irradiation Against Tumor Formation by Human-Induced Pluripotent Stem Cell-Derived Neural Progenitors. Stem Cells Dev 2016; 25:815-25. [DOI: 10.1089/scd.2015.0394] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mitsuko Katsukawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Signal Transductions, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yusuke Nakajima
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Akiko Fukumoto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Daisuke Doi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Neurosurgery, Clinical Neuroscience, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
114
|
Kim DY, Lee J, Kang D, Lee DH, Kim YJ, Hwang SG, Kim DI, Lee CW, Lee KH. Multipotent neurogenic fate of mesenchymal stem cell is determined by Cdk4-mediated hypophosphorylation of Smad-STAT3. Cell Cycle 2016; 15:1787-95. [PMID: 27192561 DOI: 10.1080/15384101.2016.1188230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Cyclin-dependent kinase (Cdk) in complex with a corresponding cyclin plays a pivotal role in neurogenic differentiation. In particular, Cdk4 activity acts as a signaling switch to direct human mesenchymal stem cells (MSCs) to neural transdifferentiation. However, the molecular evidence of how Cdk4 activity converts MSCs to neurogenic lineage remains unknown. Here, we found that Cdk4 inhibition in human MSCs enriches the populations of neural stem and progenitor pools rather than differentiated glial and neuronal cell pools. Interestingly, Cdk4 inhibition directly inactivates Smads and subsequently STAT3 signaling by hypophosphorylation, and both Cdk4 and Smads levels are linked during the processes of neural transdifferentiation and differentiation. In summary, our results provide novel molecular evidence in which Cdk4 inhibition leads to directing human MSCs to a multipotent neurogenic fate by inactivating Smads-STAT3 signaling.
Collapse
Affiliation(s)
- Dong-Young Kim
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea
| | - Janet Lee
- b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Dongrim Kang
- b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Do-Hyeong Lee
- b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Yoon-Ja Kim
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea
| | - Sang-Gu Hwang
- c Division of Radiation Cancer Biology , Korea Institute of Radiological & Medical Sciences , Seoul , Republic of Korea
| | - Dong-Ik Kim
- d Department of Vascular Surgery , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea.,e Samsung Seoul Hospital , Seoul , Republic of Korea
| | - Chang-Woo Lee
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea.,b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Kyung-Hoon Lee
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea.,f Department of Anatomy , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| |
Collapse
|
115
|
Sun T, Li W, Ling S. miR-30c and semaphorin 3A determine adult neurogenesis by regulating proliferation and differentiation of stem cells in the subventricular zones of mouse. Cell Prolif 2016; 49:270-80. [PMID: 27198082 DOI: 10.1111/cpr.12261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/31/2016] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Mechanisms that regulate proliferation of adult neural stem cells are largely unknown. Here, we have investigated the role of microR-30c (miR-30c) and its target, semaphoring 3A (sema3A), in regulating adult neurogenesis and mechanisms underlying this process. MATERIALS AND METHODS In situ hybridization, immunofluorescence and quantitative real-time PCR were used to assess complementary expression patterns of miR-30c and sema3A in mice. Effects of miR-30c in the subventricular zone (SVZ) were examined by stereotaxic injection of up- and down-regulating lentiviruses. 5'-bromo-2'-deoxyuridine labelling was performed to investigate effects of miR-30c and sema3A on adult neurogenesis. Real-time cell assays, morphological analysis and cell cycle measurements were used to reveal the mechanisms by which miR-30c and sema3A regulate adult neurogenesis. RESULTS Expression of miR-30c negatively correlated with that of sema3A in neurons, and levels of miR-30c and sema3A correlated positively with numbers of newborn cells in the SVZ and rostral migration stream. miR-30c and sema3A affected adult neurogenesis by regulating proliferation and differentiation, as well as cycles of stem cells in the SVZ. CONCLUSIONS miR-30c and sema3A regulate adult neurogenesis by controlling proliferation and differentiation of stem cells in the SVZ. This finding reveals a novel regulatory mechanism of adult neurogenesis.
Collapse
Affiliation(s)
- Tingting Sun
- Institute of Neuroscience and Anatomy, Zhejiang University, School of Medicine, Hangzhou, 310058, China
| | - Weiyun Li
- Institute of Neuroscience and Anatomy, Zhejiang University, School of Medicine, Hangzhou, 310058, China
| | - Shucai Ling
- Institute of Neuroscience and Anatomy, Zhejiang University, School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
116
|
Herrgen L, Akerman CJ. Mapping neurogenesis onset in the optic tectum of Xenopus laevis. Dev Neurobiol 2016; 76:1328-1341. [PMID: 27012549 DOI: 10.1002/dneu.22393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/22/2016] [Accepted: 03/22/2016] [Indexed: 11/06/2022]
Abstract
Neural progenitor cells have a central role in the development and evolution of the vertebrate brain. During early brain development, neural progenitors first expand their numbers through repeated proliferative divisions and then begin to exhibit neurogenic divisions. The transparent and experimentally accessible optic tectum of Xenopus laevis is an excellent model system for the study of the cell biology of neurogenesis, but the precise spatial and temporal relationship between proliferative and neurogenic progenitors has not been explored in this system. Here we construct a spatial map of proliferative and neurogenic divisions through lineage tracing of individual progenitors and their progeny. We find a clear spatial separation of proliferative and neurogenic progenitors along the anterior-posterior axis of the optic tectum, with proliferative progenitors located more posteriorly and neurogenic progenitors located more anteriorly. Since individual progenitors are repositioned toward more anterior locations as they mature, this spatial separation likely reflects an increasing restriction in the proliferative potential of individual progenitors. We then examined whether the transition from proliferative to neurogenic behavior correlates with cellular properties that have previously been implicated in regulating neurogenesis onset. Our data reveal that the transition from proliferation to neurogenesis is associated with a small change in cleavage plane orientation and a more pronounced change in cell cycle kinetics in a manner reminiscent of observations from mammalian systems. Our findings highlight the potential to use the optic tectum of Xenopus laevis as an accessible system for the study of the cell biology of neurogenesis. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1328-1341, 2016.
Collapse
Affiliation(s)
- Leah Herrgen
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom.,Centre for Neuroregeneration, University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Colin J Akerman
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| |
Collapse
|
117
|
Gabriel E, Wason A, Ramani A, Gooi LM, Keller P, Pozniakovsky A, Poser I, Noack F, Telugu NS, Calegari F, Šarić T, Hescheler J, Hyman AA, Gottardo M, Callaini G, Alkuraya FS, Gopalakrishnan J. CPAP promotes timely cilium disassembly to maintain neural progenitor pool. EMBO J 2016; 35:803-19. [PMID: 26929011 PMCID: PMC4972140 DOI: 10.15252/embj.201593679] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/05/2016] [Indexed: 12/28/2022] Open
Abstract
A mutation in the centrosomal‐P4.1‐associated protein (CPAP) causes Seckel syndrome with microcephaly, which is suggested to arise from a decline in neural progenitor cells (NPCs) during development. However, mechanisms of NPCs maintenance remain unclear. Here, we report an unexpected role for the cilium in NPCs maintenance and identify CPAP as a negative regulator of ciliary length independent of its role in centrosome biogenesis. At the onset of cilium disassembly, CPAP provides a scaffold for the cilium disassembly complex (CDC), which includes Nde1, Aurora A, and OFD1, recruited to the ciliary base for timely cilium disassembly. In contrast, mutated CPAP fails to localize at the ciliary base associated with inefficient CDC recruitment, long cilia, retarded cilium disassembly, and delayed cell cycle re‐entry leading to premature differentiation of patient iPS‐derived NPCs. Aberrant CDC function also promotes premature differentiation of NPCs in Seckel iPS‐derived organoids. Thus, our results suggest a role for cilia in microcephaly and its involvement during neurogenesis and brain size control.
Collapse
Affiliation(s)
- Elke Gabriel
- Center for Molecular Medicine and Institute for Biochemistry I of the University of Cologne, Cologne, Germany
| | - Arpit Wason
- Center for Molecular Medicine and Institute for Biochemistry I of the University of Cologne, Cologne, Germany
| | - Anand Ramani
- Center for Molecular Medicine and Institute for Biochemistry I of the University of Cologne, Cologne, Germany
| | - Li Ming Gooi
- Center for Molecular Medicine and Institute for Biochemistry I of the University of Cologne, Cologne, Germany
| | - Patrick Keller
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrei Pozniakovsky
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Florian Noack
- DFG-Research Center and Cluster of Excellence for Regenerative Therapies, TU-Dresden, Dresden, Germany
| | - Narasimha Swamy Telugu
- Center for Physiology and Pathophysiology, Institute for Neurophysiology Medical Faculty University of Cologne, Cologne, Germany
| | - Federico Calegari
- DFG-Research Center and Cluster of Excellence for Regenerative Therapies, TU-Dresden, Dresden, Germany
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology Medical Faculty University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology Medical Faculty University of Cologne, Cologne, Germany
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marco Gottardo
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Fowzan Sami Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center Alfasial University, Riyadh, Saudi Arabia Department of Anatomy and Cell Biology, College of Medicine Alfasial University, Riyadh, Saudi Arabia
| | - Jay Gopalakrishnan
- Center for Molecular Medicine and Institute for Biochemistry I of the University of Cologne, Cologne, Germany
| |
Collapse
|
118
|
Julian LM, Carpenedo RL, Rothberg JLM, Stanford WL. Formula G1: Cell cycle in the driver's seat of stem cell fate determination. Bioessays 2016; 38:325-32. [DOI: 10.1002/bies.201500187] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lisa M. Julian
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
| | - Richard L. Carpenedo
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
| | - Janet L. Manias Rothberg
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa ON Canada
| | - William L. Stanford
- Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa ON Canada
- Faculty of Graduate and Postdoctoral Studies; Ottawa; ON Canada
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa ON Canada
- Department of Biochemistry; Microbiology and Immunology; University of Ottawa; Ottawa ON Canada
- Ottawa Institute of Systems Biology; Ottawa; Ontario Canada
| |
Collapse
|
119
|
Cell proliferation and cell death are disturbed during prenatal and postnatal brain development after uranium exposure. Neurotoxicology 2016; 52:34-45. [DOI: 10.1016/j.neuro.2015.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/13/2015] [Accepted: 10/13/2015] [Indexed: 11/24/2022]
|
120
|
Muthukrishnan SD, Yang X, Friesel R, Oxburgh L. Concurrent BMP7 and FGF9 signalling governs AP-1 function to promote self-renewal of nephron progenitor cells. Nat Commun 2015; 6:10027. [PMID: 26634297 PMCID: PMC4686668 DOI: 10.1038/ncomms10027] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/28/2015] [Indexed: 01/10/2023] Open
Abstract
Self-renewal of nephron progenitor cells (NPCs) is governed by BMP, FGF and WNT signalling. Mechanisms underlying cross-talk between these pathways at the molecular level are largely unknown. Here we delineate the pathway through which the proliferative BMP7 signal is transduced in NPCs in the mouse. BMP7 activates the MAPKs TAK1 and JNK to phosphorylate the transcription factor JUN, which in turn governs transcription of AP-1-element containing G1-phase cell cycle regulators such as Myc and Ccnd1 to promote NPC proliferation. Conditional inactivation of Tak1 or Jun in cap mesenchyme causes identical phenotypes characterized by premature depletion of NPCs. While JUN is regulated by BMP7, we find that its partner FOS is regulated by FGF9. We demonstrate that BMP7 and FGF9 coordinately regulate AP-1 transcription to promote G1-S cell cycle progression and NPC proliferation. Our findings identify a molecular mechanism explaining the important cooperation between two major NPC self-renewal pathways. The growth factors BMP and FGF both stimulate the self-renewal of nephron progenitor cells (NPCs), but how these signals overlap is unclear. Here in the mouse, Muthukrishnan et al. find BMP7 and FGF9 coordinately regulate AP-1 transcriptional activity, promoting G1-S cell cycle progression and NPC proliferation.
Collapse
Affiliation(s)
- Sree Deepthi Muthukrishnan
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine 04469, USA
| | - Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| | - Robert Friesel
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| |
Collapse
|
121
|
Thuret R, Auger H, Papalopulu N. Analysis of neural progenitors from embryogenesis to juvenile adult in Xenopus laevis reveals biphasic neurogenesis and continuous lengthening of the cell cycle. Biol Open 2015; 4:1772-81. [PMID: 26621828 PMCID: PMC4736028 DOI: 10.1242/bio.013391] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Xenopus laevis is a prominent model system for studying neural development, but our understanding of the long-term temporal dynamics of neurogenesis remains incomplete. Here, we present the first continuous description of neurogenesis in X. laevis, covering the entire period of development from the specification of neural ectoderm during gastrulation to juvenile frog. We have used molecular markers to identify progenitors and neurons, short-term bromodeoxyuridine (BrdU) incorporation to map the generation of newborn neurons and dual pulse S-phase labelling to characterise changes in their cell cycle length. Our study revealed the persistence of Sox3-positive progenitor cells from the earliest stages of neural development through to the juvenile adult. Two periods of intense neuronal generation were observed, confirming the existence of primary and secondary waves of neurogenesis, punctuated by a period of quiescence before metamorphosis and culminating in another period of quiescence in the young adult. Analysis of multiple parameters indicates that neural progenitors alternate between global phases of differentiation and amplification and that, regardless of their behaviour, their cell cycle lengthens monotonically during development, at least at the population level.
Collapse
Affiliation(s)
- Raphaël Thuret
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Hélène Auger
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Nancy Papalopulu
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
122
|
Sgrò F, Bianchi FT, Falcone M, Pallavicini G, Gai M, Chiotto AMA, Berto GE, Turco E, Chang YJ, Huttner WB, Di Cunto F. Tissue-specific control of midbody microtubule stability by Citron kinase through modulation of TUBB3 phosphorylation. Cell Death Differ 2015; 23:801-13. [PMID: 26586574 DOI: 10.1038/cdd.2015.142] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 09/13/2015] [Accepted: 09/29/2015] [Indexed: 01/02/2023] Open
Abstract
Cytokinesis, the physical separation of daughter cells at the end of cell cycle, is commonly considered a highly stereotyped phenomenon. However, in some specialized cells this process may involve specific molecular events that are still largely unknown. In mammals, loss of Citron-kinase (CIT-K) leads to massive cytokinesis failure and apoptosis only in neuronal progenitors and in male germ cells, resulting in severe microcephaly and testicular hypoplasia, but the reasons for this specificity are unknown. In this report we show that CIT-K modulates the stability of midbody microtubules and that the expression of tubulin β-III (TUBB3) is crucial for this phenotype. We observed that TUBB3 is expressed in proliferating CNS progenitors, with a pattern correlating with the susceptibility to CIT-K loss. More importantly, depletion of TUBB3 in CIT-K-dependent cells makes them resistant to CIT-K loss, whereas TUBB3 overexpression increases their sensitivity to CIT-K knockdown. The loss of CIT-K leads to a strong decrease in the phosphorylation of S444 on TUBB3, a post-translational modification associated with microtubule stabilization. CIT-K may promote this event by interacting with TUBB3 and by recruiting at the midbody casein kinase-2α (CK2α) that has previously been reported to phosphorylate the S444 residue. Indeed, CK2α is lost from the midbody in CIT-K-depleted cells. Moreover, expression of the nonphosphorylatable TUBB3 mutant S444A induces cytokinesis failure, whereas expression of the phospho-mimetic mutant S444D rescues the cytokinesis failure induced by both CIT-K and CK2α loss. Altogether, our findings reveal that expression of relatively low levels of TUBB3 in mitotic cells can be detrimental for their cytokinesis and underscore the importance of CIT-K in counteracting this event.
Collapse
Affiliation(s)
- F Sgrò
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - F T Bianchi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - M Falcone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - G Pallavicini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - M Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - A M A Chiotto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - G E Berto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - E Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Y J Chang
- Max-Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - W B Huttner
- Max-Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - F Di Cunto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy.,Neuroscience Institute of Turin, Turin, Italy
| |
Collapse
|
123
|
ASPM regulates symmetric stem cell division by tuning Cyclin E ubiquitination. Nat Commun 2015; 6:8763. [PMID: 26581405 DOI: 10.1038/ncomms9763] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/28/2015] [Indexed: 11/08/2022] Open
Abstract
We generate a mouse model for the human microcephaly syndrome by mutating the ASPM locus, and demonstrate a premature exhaustion of the neuronal progenitor pool due to dysfunctional self-renewal processes. Earlier studies have linked ASPM mutant progenitor excessive cell cycle exit to a mitotic orientation defect. Here, we demonstrate a mitotic orientation-independent effect of ASPM on cell cycle duration. We pinpoint the cell fate-determining factor to the length of time spent in early G1 before traversing the restriction point. Characterization of the molecular mechanism reveals an interaction between ASPM and the Cdk2/Cyclin E complex, regulating the Cyclin activity by modulating its ubiquitination, phosphorylation and localization into the nucleus, before the cell is fated to transverse the restriction point. Thus, we reveal a novel function of ASPM in mediating the tightly coordinated Ubiquitin- Cyclin E- Retinoblastoma- E2F bistable-signalling pathway controlling restriction point progression and stem cell maintenance.
Collapse
|
124
|
Aneuploidy causes premature differentiation of neural and intestinal stem cells. Nat Commun 2015; 6:8894. [PMID: 26573328 PMCID: PMC4660207 DOI: 10.1038/ncomms9894] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 10/14/2015] [Indexed: 12/31/2022] Open
Abstract
Aneuploidy is associated with a variety of diseases such as cancer and microcephaly. Although many studies have addressed the consequences of a non-euploid genome in cells, little is known about their overall consequences in tissue and organism development. Here we use two different mutant conditions to address the consequences of aneuploidy during tissue development and homeostasis in Drosophila. We show that aneuploidy causes brain size reduction due to a decrease in the number of proliferative neural stem cells (NSCs), but not through apoptosis. Instead, aneuploid NSCs present an extended G1 phase, which leads to cell cycle exit and premature differentiation. Moreover, we show that this response to aneuploidy is also present in adult intestinal stem cells but not in the wing disc. Our work highlights a neural and intestine stem cell-specific response to aneuploidy, which prevents their proliferation and expansion. It is unclear why certain tissues are more susceptible to the consequences of aneuploidy. Here, in Drosophila, Gogendeau et al. identify aneuploidy as the cause of lengthened G1 and premature differentiation in both neural and adult intestinal stem cells, which prevents cells with abnormal genomes from cycling.
Collapse
|
125
|
Rodrigo Albors A, Tazaki A, Rost F, Nowoshilow S, Chara O, Tanaka EM. Planar cell polarity-mediated induction of neural stem cell expansion during axolotl spinal cord regeneration. eLife 2015; 4:e10230. [PMID: 26568310 PMCID: PMC4755742 DOI: 10.7554/elife.10230] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/12/2015] [Indexed: 02/07/2023] Open
Abstract
Axolotls are uniquely able to mobilize neural stem cells to regenerate all missing regions of the spinal cord. How a neural stem cell under homeostasis converts after injury to a highly regenerative cell remains unknown. Here, we show that during regeneration, axolotl neural stem cells repress neurogenic genes and reactivate a transcriptional program similar to embryonic neuroepithelial cells. This dedifferentiation includes the acquisition of rapid cell cycles, the switch from neurogenic to proliferative divisions, and the re-expression of planar cell polarity (PCP) pathway components. We show that PCP induction is essential to reorient mitotic spindles along the anterior-posterior axis of elongation, and orthogonal to the cell apical-basal axis. Disruption of this property results in premature neurogenesis and halts regeneration. Our findings reveal a key role for PCP in coordinating the morphogenesis of spinal cord outgrowth with the switch from a homeostatic to a regenerative stem cell that restores missing tissue. DOI:http://dx.doi.org/10.7554/eLife.10230.001 Stem cells found in adult tissues are vitally important for tissue repair and maintenance. These cells divide in two main ways: equally to create two new stem cells, or unequally to create a stem cell and a cell that can develop into one of the cell types in the tissue. A key challenge for biologists is to understand how these tissue-resident stem cells are activated and organized to regenerate injured or missing tissue. Throughout the life of the axolotl salamander, neural stem cells in the spinal cord occasionally divide to add new nerve cells to the healthy spinal cord. However, the axolotl can also regenerate part of its spinal cord, for example if its tail is lost. Under these conditions, the neural stem cells can convert into a highly regenerative stem cell that can produce all the different cell types needed to regrow the spinal cord. As a stem cell becomes a new cell type, it activates different sets of genes. Therefore, Rodrigo Albors, Tazaki et al. measured gene activity in the neural stem cells involved in axolotl spinal cord regeneration to uncover how these cells develop into a more regenerative form. This revealed that when an axolotl tail is amputated, resident stem cells turn off the genes that are specifically active in neuron-generating cells. In addition, they activate a similar set of genes to that seen in the embryonic cells that form the developing nervous system. These genes speed up cell division and activate an important signaling pathway. This pathway – the Wnt/PCP pathway – fulfils various developmental roles, one being to orient cell divisions, particularly in elongating tissues. In axolotls, this pathway causes the stem cells to divide equally to increase the number of available stem cells, and orients the direction of these divisions to ensure that the regenerating spinal cord elongates correctly. If this pathway is disrupted, the cells return to dividing unequally, generating nerve cells prematurely and halting the growth of the spinal cord. Such insights could help develop methods of repairing damaged nervous tissue in other animals that cannot regenerate to the extent that axolotls can. DOI:http://dx.doi.org/10.7554/eLife.10230.002
Collapse
Affiliation(s)
- Aida Rodrigo Albors
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Akira Tazaki
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Fabian Rost
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | - Sergej Nowoshilow
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Osvaldo Chara
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany.,Institute of Physics of Liquids and Biological Systems, National Scientific and Technical Research Council, University of La Plata, La Plata, Argentina
| | - Elly M Tanaka
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
126
|
Effects of addictive drugs on adult neural stem/progenitor cells. Cell Mol Life Sci 2015; 73:327-48. [PMID: 26468052 DOI: 10.1007/s00018-015-2067-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/04/2015] [Accepted: 10/08/2015] [Indexed: 12/18/2022]
Abstract
Neural stem/progenitor cells (NSPCs) undergo a series of developmental processes before giving rise to newborn neurons, astrocytes and oligodendrocytes in adult neurogenesis. During the past decade, the role of NSPCs has been highlighted by studies on adult neurogenesis modulated by addictive drugs. It has been proven that these drugs regulate the proliferation, differentiation and survival of adult NSPCs in different manners, which results in the varying consequences of adult neurogenesis. The effects of addictive drugs on NSPCs are exerted via a variety of different mechanisms and pathways, which interact with one another and contribute to the complexity of NSPC regulation. Here, we review the effects of different addictive drugs on NSPCs, and the related experimental methods and paradigms. We also discuss the current understanding of major signaling molecules, especially the putative common mechanisms, underlying such effects. Finally, we review the future directions of research in this area.
Collapse
|
127
|
Farioli-Vecchioli S, Tirone F. Control of the Cell Cycle in Adult Neurogenesis and its Relation with Physical Exercise. Brain Plast 2015; 1:41-54. [PMID: 29765834 PMCID: PMC5928538 DOI: 10.3233/bpl-150013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the adult brain the neurogenesis is mainly restricted to two neurogenic regions: newly generated neurons arise at the subventricular zone (SVZ) of the lateral ventricle and at the subgranular zone of the hippocampal subregion named the dentate gyrus. The hippocampus is involved in learning and memory paradigms and the generation of new hippocampal neurons has been hypothesized to be a pivotal form of plasticity involved in the process. Moreover the dysregulation of hippocampal adult neurogenesis has been recognized and could anticipate several varieties of brain disease such as Alzheimer disease, epilepsy and depression. Over the last few decades numerous intrinsic, epigenetic and environmental factors have been revealed to deeply influence the process of adult neurogenesis, although the underlying mechanisms remain largely unknown. Growing evidence indicates that physical exercise represents one of the main extrinsic factor able to profoundly increase hippocampal adult neurogenesis, by altering neurochemistry and function of newly generated neurons. The present review surveys how neurogenesis can be modulated by cell cycle kinetics and highlights the putative role of the cell cycle length as a key component of the beneficial effect of running for hippocampal adult neurogenesis, both in physiological conditions and in the presence of defective neurogenesis.
Collapse
Affiliation(s)
- Stefano Farioli-Vecchioli
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Rome, Italy
| | - Felice Tirone
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione S.Lucia, Rome, Italy
| |
Collapse
|
128
|
Beckervordersandforth R, Zhang CL, Lie DC. Transcription-Factor-Dependent Control of Adult Hippocampal Neurogenesis. Cold Spring Harb Perspect Biol 2015; 7:a018879. [PMID: 26430216 DOI: 10.1101/cshperspect.a018879] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adult-generated dentate granule neurons have emerged as major contributors to hippocampal plasticity. New neurons are generated from neural stem cells through a complex sequence of proliferation, differentiation, and maturation steps. Development of the new neuron is dependent on the precise temporal activity of transcription factors, which coordinate the expression of stage-specific genetic programs. Here, we review current knowledge in transcription factor-mediated regulation of mammalian neural stem cells and neurogenesis and will discuss potential mechanisms of how transcription factor networks, on one hand, allow for precise execution of the developmental sequence and, on the other hand, allow for adaptation of the rate and timing of adult neurogenesis in response to complex stimuli. Understanding transcription factor-mediated control of neuronal development will provide new insights into the mechanisms underlying neurogenesis-dependent plasticity in health and disease.
Collapse
Affiliation(s)
- Ruth Beckervordersandforth
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Dieter Chichung Lie
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
129
|
Homem CCF, Repic M, Knoblich JA. Proliferation control in neural stem and progenitor cells. Nat Rev Neurosci 2015; 16:647-59. [PMID: 26420377 DOI: 10.1038/nrn4021] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural circuit function can be drastically affected by variations in the number of cells that are produced during development or by a reduction in adult cell number owing to disease. For this reason, unique cell cycle and cell growth control mechanisms operate in the developing and adult brain. In Drosophila melanogaster and in mammalian neural stem and progenitor cells, these mechanisms are intricately coordinated with the developmental age and the nutritional, metabolic and hormonal state of the animal. Defects in neural stem cell proliferation that result in the generation of incorrect cell numbers or defects in neural stem cell differentiation can cause microcephaly or megalencephaly.
Collapse
Affiliation(s)
- Catarina C F Homem
- Institute of Molecular Biotechnology, Dr. Bohr Gasse 3, 1030, Vienna, Austria.,Chronic Diseases Research Center, NOVA Medical School, Rua Camara Pestana, 6, 1150-082 Lisbon, Portugal
| | - Marko Repic
- Institute of Molecular Biotechnology, Dr. Bohr Gasse 3, 1030, Vienna, Austria
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology, Dr. Bohr Gasse 3, 1030, Vienna, Austria
| |
Collapse
|
130
|
Abstract
The neocortex is the part of the brain responsible for execution of higher-order brain functions, including cognition, sensory perception, and sophisticated motor control. During evolution, the neocortex has developed an unparalleled neuronal diversity, which still remains partly unclassified and unmapped at the functional level. Here, we broadly review the structural blueprint of the neocortex and discuss the current classification of its neuronal diversity. We then cover the principles and mechanisms that build neuronal diversity during cortical development and consider the impact of neuronal class-specific identity in shaping cortical connectivity and function.
Collapse
Affiliation(s)
- Simona Lodato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| |
Collapse
|
131
|
Abstract
The neocortex is the part of the brain responsible for execution of higher-order brain functions, including cognition, sensory perception, and sophisticated motor control. During evolution, the neocortex has developed an unparalleled neuronal diversity, which still remains partly unclassified and unmapped at the functional level. Here, we broadly review the structural blueprint of the neocortex and discuss the current classification of its neuronal diversity. We then cover the principles and mechanisms that build neuronal diversity during cortical development and consider the impact of neuronal class-specific identity in shaping cortical connectivity and function.
Collapse
Affiliation(s)
- Simona Lodato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| |
Collapse
|
132
|
Micheli L, Ceccarelli M, Farioli-Vecchioli S, Tirone F. Control of the Normal and Pathological Development of Neural Stem and Progenitor Cells by the PC3/Tis21/Btg2 and Btg1 Genes. J Cell Physiol 2015; 230:2881-90. [DOI: 10.1002/jcp.25038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/05/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Laura Micheli
- Institute of Cell Biology and Neurobiology; National Research Council; Fondazione S.Lucia Rome Italy
| | - Manuela Ceccarelli
- Institute of Cell Biology and Neurobiology; National Research Council; Fondazione S.Lucia Rome Italy
| | - Stefano Farioli-Vecchioli
- Institute of Cell Biology and Neurobiology; National Research Council; Fondazione S.Lucia Rome Italy
| | - Felice Tirone
- Institute of Cell Biology and Neurobiology; National Research Council; Fondazione S.Lucia Rome Italy
| |
Collapse
|
133
|
Míguez DG. A Branching Process to Characterize the Dynamics of Stem Cell Differentiation. Sci Rep 2015; 5:13265. [PMID: 26286123 PMCID: PMC4541069 DOI: 10.1038/srep13265] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/23/2015] [Indexed: 01/15/2023] Open
Abstract
The understanding of the regulatory processes that orchestrate stem cell maintenance is a cornerstone in developmental biology. Here, we present a mathematical model based on a branching process formalism that predicts average rates of proliferative and differentiative divisions in a given stem cell population. In the context of vertebrate neurogenesis, the model predicts complex non-monotonic variations in the rates of pp, pd and dd modes of division as well as in cell cycle length, in agreement with experimental results. Moreover, the model shows that the differentiation probability follows a binomial distribution, allowing us to develop equations to predict the rates of each mode of division. A phenomenological simulation of the developing spinal cord informed with the average cell cycle length and division rates predicted by the mathematical model reproduces the correct dynamics of proliferation and differentiation in terms of average numbers of progenitors and differentiated cells. Overall, the present mathematical framework represents a powerful tool to unveil the changes in the rate and mode of division of a given stem cell pool by simply quantifying numbers of cells at different times.
Collapse
Affiliation(s)
- David G Míguez
- Depto. de Física de la Materia Condensada, Instituto Nicolás Cabrera and IFIMAC, Universidad Autónoma de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
134
|
Abstract
A close relationship between proliferation and cell fate specification has been well documented in many developmental systems. In addition to the gradual cell fate changes accompanying normal development and tissue homeostasis, it is now commonly appreciated that cell fate could also undergo drastic changes, as illustrated by the induction of pluripotency from many differentiated somatic cell types during the process of Yamanaka reprogramming. Strikingly, the drastic cell fate change induced by Yamanaka factors (Oct4, Sox2, Klf4, and c-Myc) is preceded by extensive cell cycle acceleration. Prompted by our recent discovery that progression toward pluripotency from rare somatic cells could bypass the stochastic phase of reprogramming and that a key feature of these somatic cells is an ultrafast cell cycle (~8 h/cycle), we assess whether cell cycle dynamics could provide a general framework for controlling cell fate. Several potential mechanisms on how cell cycle dynamics may impact cell fate determination by regulating chromatin, key transcription factor concentration, or their interactions are discussed. Specific challenges and implications for studying and manipulating cell fate are considered.
Collapse
|
135
|
Morris-Rosendahl DJ, Kaindl AM. What next-generation sequencing (NGS) technology has enabled us to learn about primary autosomal recessive microcephaly (MCPH). Mol Cell Probes 2015; 29:271-81. [PMID: 26050940 DOI: 10.1016/j.mcp.2015.05.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 11/27/2022]
Abstract
The impact that next-generation sequencing technology (NGS) is having on many aspects of molecular and cell biology, is becoming increasingly apparent. One of the most noticeable outcomes of the new technology in human genetics, has been the accelerated rate of identification of disease-causing genes. Especially for rare, heterogeneous disorders, such as autosomal recessive primary microcephaly (MCPH), the handful of genes previously known to harbour disease-causing mutations, has grown at an unprecedented rate within a few years. Knowledge of new genes mutated in MCPH over the last four years has contributed to our understanding of the disorder at both the clinical and cellular levels. The functions of proteins such as WDR62, CASC5, PHC1, CDK6, CENP-E, CENP-F, CEP63, ZNF335, PLK4 and TUBGPC, have been added to the complex network of critical cellular processes known to be involved in brain growth and size. In addition to the importance of mitotic spindle assembly and structure, centrosome and centriole function and DNA repair and damage response, new mechanisms involving kinetochore-associated proteins and chromatin remodelling complexes have been elucidated. Two of the major contributions to our clinical knowledge are the realisation that primary microcephaly caused by mutations in genes at the MCPH loci is seldom an isolated clinical feature and is often accompanied either by additional cortical malformations or primordial dwarfism. Gene-phenotype correlations are being revisited, with a new dimension of locus heterogeneity and phenotypic variability being revealed.
Collapse
Affiliation(s)
- Deborah J Morris-Rosendahl
- Clinical Genetics and Genomics, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Angela M Kaindl
- Department of Pediatric Neurology, Charité University Medicine, Berlin, Germany; Institute of Cell Biology and Neurobiology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
136
|
Turrero García M, Chang Y, Arai Y, Huttner WB. S-phase duration is the main target of cell cycle regulation in neural progenitors of developing ferret neocortex. J Comp Neurol 2015; 524:456-70. [PMID: 25963823 PMCID: PMC5008145 DOI: 10.1002/cne.23801] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/30/2015] [Accepted: 05/06/2015] [Indexed: 01/22/2023]
Abstract
The evolutionary expansion of the neocortex primarily reflects increases in abundance and proliferative capacity of cortical progenitors and in the length of the neurogenic period during development. Cell cycle parameters of neocortical progenitors are an important determinant of cortical development. The ferret (Mustela putorius furo), a gyrencephalic mammal, has gained increasing importance as a model for studying corticogenesis. Here, we have studied the abundance, proliferation, and cell cycle parameters of different neural progenitor types, defined by their differential expression of the transcription factors Pax6 and Tbr2, in the various germinal zones of developing ferret neocortex. We focused our analyses on postnatal day 1, a late stage of cortical neurogenesis when upper‐layer neurons are produced. Based on cumulative 5‐ethynyl‐2′‐deoxyuridine (EdU) labeling as well as Ki67 and proliferating cell nuclear antigen (PCNA) immunofluorescence, we determined the duration of the various cell cycle phases of the different neocortical progenitor subpopulations. Ferret neocortical progenitors were found to exhibit longer cell cycles than those of rodents and little variation in the duration of G1 among distinct progenitor types, also in contrast to rodents. Remarkably, the main difference in cell cycle parameters among the various progenitor types was the duration of S‐phase, which became shorter as progenitors progressively changed transcription factor expression from patterns characteristic of self‐renewal to those of neuron production. Hence, S‐phase duration emerges as major target of cell cycle regulation in cortical progenitors of this gyrencephalic mammal. J. Comp. Neurol. 524:456–470, 2016. © 2015 The Authors The Journal of Comparative Neurology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Miguel Turrero García
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| | - YoonJeung Chang
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| | - Yoko Arai
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| |
Collapse
|
137
|
Julian LM, Blais A. Transcriptional control of stem cell fate by E2Fs and pocket proteins. Front Genet 2015; 6:161. [PMID: 25972892 PMCID: PMC4412126 DOI: 10.3389/fgene.2015.00161] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 04/08/2015] [Indexed: 01/04/2023] Open
Abstract
E2F transcription factors and their regulatory partners, the pocket proteins (PPs), have emerged as essential regulators of stem cell fate control in a number of lineages. In mammals, this role extends from both pluripotent stem cells to those encompassing all embryonic germ layers, as well as extra-embryonic lineages. E2F/PP-mediated regulation of stem cell decisions is highly evolutionarily conserved, and is likely a pivotal biological mechanism underlying stem cell homeostasis. This has immense implications for organismal development, tissue maintenance, and regeneration. In this article, we discuss the roles of E2F factors and PPs in stem cell populations, focusing on mammalian systems. We discuss emerging findings that position the E2F and PP families as widespread and dynamic epigenetic regulators of cell fate decisions. Additionally, we focus on the ever expanding landscape of E2F/PP target genes, and explore the possibility that E2Fs are not simply regulators of general ‘multi-purpose’ cell fate genes but can execute tissue- and cell type-specific gene regulatory programs.
Collapse
Affiliation(s)
- Lisa M Julian
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON Canada
| | - Alexandre Blais
- Ottawa Institute of Systems Biology, Ottawa, ON Canada ; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON Canada
| |
Collapse
|
138
|
Tissue-specific targeting of cell fate regulatory genes by E2f factors. Cell Death Differ 2015; 23:565-75. [PMID: 25909886 DOI: 10.1038/cdd.2015.36] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 02/03/2015] [Accepted: 03/04/2015] [Indexed: 12/30/2022] Open
Abstract
Cell cycle proteins are important regulators of diverse cell fate decisions, and in this capacity have pivotal roles in neurogenesis and brain development. The mechanisms by which cell cycle regulation is integrated with cell fate control in the brain and other tissues are poorly understood, and an outstanding question is whether the cell cycle machinery regulates fate decisions directly or instead as a secondary consequence of proliferative control. Identification of the genes targeted by E2 promoter binding factor (E2f) transcription factors, effectors of the pRb/E2f cell cycle pathway, will provide essential insights into these mechanisms. We identified the promoter regions bound by three neurogenic E2f factors in neural precursor cells in a genome-wide manner. Through bioinformatic analyses and integration of published genomic data sets we uncovered hundreds of transcriptionally active E2f-bound promoters corresponding to genes that control cell fate processes, including key transcriptional regulators and members of the Notch, fibroblast growth factor, Wnt and Tgf-β signaling pathways. We also demonstrate a striking enrichment of the CCCTC binding factor transcription factor (Ctcf) at E2f3-bound nervous system-related genes, suggesting a potential regulatory co-factor for E2f3 in controlling differentiation. Finally, we provide the first demonstration of extensive tissue specificity among E2f target genes in mammalian cells, whereby E2f3 promoter binding is well conserved between neural and muscle precursors at genes associated with cell cycle processes, but is tissue-specific at differentiation-associated genes. Our findings implicate the cell cycle pathway as a widespread regulator of cell fate genes, and suggest that E2f3 proteins control cell type-specific differentiation programs by regulating unique sets of target genes. This work significantly enhances our understanding of how the cell cycle machinery impacts cell fate and differentiation, and will importantly drive further discovery regarding the mechanisms of cell fate control and transcriptional regulation in the brain, as well as in other tissues.
Collapse
|
139
|
Dehay C, Kennedy H, Kosik KS. The outer subventricular zone and primate-specific cortical complexification. Neuron 2015; 85:683-94. [PMID: 25695268 DOI: 10.1016/j.neuron.2014.12.060] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Evolutionary expansion and complexification of the primate cerebral cortex are largely linked to the emergence of the outer subventricular zone (OSVZ), a uniquely structured germinal zone that generates the expanded primate supragranular layers. The primate OSVZ departs from rodent germinal zones in that it includes a higher diversity of precursor types, inter-related in bidirectional non-hierarchical lineages. In addition, primate-specific regulatory mechanisms are operating in primate cortical precursors via the occurrence of novel miRNAs. Here, we propose that the origin and evolutionary importance of the OSVZ is related to genetic changes in multiple regulatory loops and that cell-cycle regulation is a favored target for evolutionary adaptation of the cortex.
Collapse
Affiliation(s)
- Colette Dehay
- Stem Cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lepine, 69500 Bron, France; Université de Lyon, Université Lyon I, 69003, Lyon, France.
| | - Henry Kennedy
- Stem Cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lepine, 69500 Bron, France; Université de Lyon, Université Lyon I, 69003, Lyon, France.
| | - Kenneth S Kosik
- Neuroscience Research Institute and Dept Cellular Molecular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
140
|
Slowly dividing neural progenitors are an embryonic origin of adult neural stem cells. Nat Neurosci 2015; 18:657-65. [PMID: 25821910 DOI: 10.1038/nn.3989] [Citation(s) in RCA: 230] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/05/2015] [Indexed: 12/13/2022]
Abstract
The mechanism by which adult neural stem cells (NSCs) are established during development is unclear. In this study, analysis of cell cycle progression by examining retention of a histone 2B (H2B)-GFP fusion protein revealed that, in a subset of mouse embryonic neural progenitor cells (NPCs), the cell cycle slows between embryonic day (E) 13.5 and E15.5 while other embryonic NPCs continue to divide rapidly. By allowing H2B-GFP expressed at E9.5 to become diluted in dividing cells until the young adult stage, we determined that a majority of NSCs in the young adult subependymal zone (SEZ) originated from these slowly dividing embryonic NPCs. The cyclin-dependent kinase inhibitor p57 is highly expressed in this embryonic subpopulation, and the deletion of p57 impairs the emergence of adult NSCs. Our results suggest that a substantial fraction of adult SEZ NSCs is derived from a slowly dividing subpopulation of embryonic NPCs and identify p57 as a key factor in generating this embryonic origin of adult SEZ NSCs.
Collapse
|
141
|
O'Leary CJ, Bradford D, Chen M, White A, Blackmore DG, Cooper HM. The Netrin/RGM Receptor, Neogenin, Controls Adult Neurogenesis by Promoting Neuroblast Migration and Cell Cycle Exit. Stem Cells 2015; 33:503-14. [DOI: 10.1002/stem.1861] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 07/31/2014] [Accepted: 09/06/2014] [Indexed: 01/16/2023]
Affiliation(s)
- Conor J. O'Leary
- The University of Queensland, Queensland Brain Institute; Brisbane Queensland Australia
| | - DanaKai Bradford
- The University of Queensland, Queensland Brain Institute; Brisbane Queensland Australia
| | - Min Chen
- The University of Queensland, Queensland Brain Institute; Brisbane Queensland Australia
| | - Amanda White
- The University of Queensland, Queensland Brain Institute; Brisbane Queensland Australia
| | - Daniel G. Blackmore
- The University of Queensland, Queensland Brain Institute; Brisbane Queensland Australia
| | - Helen M. Cooper
- The University of Queensland, Queensland Brain Institute; Brisbane Queensland Australia
| |
Collapse
|
142
|
Najas S, Arranz J, Lochhead PA, Ashford AL, Oxley D, Delabar JM, Cook SJ, Barallobre MJ, Arbonés ML. DYRK1A-mediated Cyclin D1 Degradation in Neural Stem Cells Contributes to the Neurogenic Cortical Defects in Down Syndrome. EBioMedicine 2015; 2:120-34. [PMID: 26137553 PMCID: PMC4484814 DOI: 10.1016/j.ebiom.2015.01.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 01/02/2023] Open
Abstract
Alterations in cerebral cortex connectivity lead to intellectual disability and in Down syndrome, this is associated with a deficit in cortical neurons that arises during prenatal development. However, the pathogenic mechanisms that cause this deficit have not yet been defined. Here we show that the human DYRK1A kinase on chromosome 21 tightly regulates the nuclear levels of Cyclin D1 in embryonic cortical stem (radial glia) cells, and that a modest increase in DYRK1A protein in transgenic embryos lengthens the G1 phase in these progenitors. These alterations promote asymmetric proliferative divisions at the expense of neurogenic divisions, producing a deficit in cortical projection neurons that persists in postnatal stages. Moreover, radial glial progenitors in the Ts65Dn mouse model of Down syndrome have less Cyclin D1, and Dyrk1a is the triplicated gene that causes both early cortical neurogenic defects and decreased nuclear Cyclin D1 levels in this model. These data provide insights into the mechanisms that couple cell cycle regulation and neuron production in cortical neural stem cells, emphasizing that the deleterious effect of DYRK1A triplication in the formation of the cerebral cortex begins at the onset of neurogenesis, which is relevant to the search for early therapeutic interventions in Down syndrome.
Collapse
Affiliation(s)
- Sònia Najas
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Juan Arranz
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Pamela A. Lochhead
- Signalling Programme, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Anne L. Ashford
- Signalling Programme, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - David Oxley
- Proteomics Group, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Jean M. Delabar
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, 75013 Paris, France
| | - Simon J. Cook
- Signalling Programme, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - María José Barallobre
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Maria L. Arbonés
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| |
Collapse
|
143
|
Watanabe N, Kageyama R, Ohtsuka T. Hbp1 regulates the timing of neuronal differentiation during cortical development by controlling cell cycle progression. Development 2015; 142:2278-90. [DOI: 10.1242/dev.120477] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/26/2015] [Indexed: 11/20/2022]
Abstract
In the developing mammalian brain, neural stem cells (NSCs) initially expand the progenitor pool by symmetric divisions. NSCs then shift from symmetric to asymmetric division and commence neurogenesis. Although the precise mechanisms regulating the developmental timing of this transition have not been fully elucidated, gradual elongation in the length of the cell cycle and coinciding accumulation of determinants that promote neuronal differentiation may function as a biological clock that regulates the onset of asymmetric division and neurogenesis. We conducted gene expression profiling of embryonic NSCs in the cortical regions and found that expression of high mobility group box transcription factor 1 (Hbp1) was upregulated during neurogenic stages. Induced conditional knockout mice of Hbp1 generated by crossing with Nestin-CreERT2 mice exhibited a remarkable dilatation of the telencephalic vesicles with a tangentially expanded ventricular zone and a thinner cortical plate containing reduced numbers of neurons. In these Hbp1-deficient mouse embryos, neural stem/progenitor cells continued to divide with a shorter cell cycle length. And downstream target genes of the Wnt signaling, such as cyclin D1 and c-jun, were upregulated in the germinal zone of the cortical regions. These results indicate that Hbp1 plays a critical role in regulating the timing of cortical neurogenesis by elongating the cell cycle and is essential for normal cortical development.
Collapse
Affiliation(s)
- Naoki Watanabe
- Institute for Virus Research, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto 606-8507, Japan
- Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Ryoichiro Kageyama
- Institute for Virus Research, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto 606-8507, Japan
- Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Toshiyuki Ohtsuka
- Institute for Virus Research, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
144
|
Can the ‘neuron theory’ be complemented by a universal mechanism for generic neuronal differentiation. Cell Tissue Res 2014; 359:343-84. [DOI: 10.1007/s00441-014-2049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
|
145
|
Lewitus E, Kelava I, Kalinka AT, Tomancak P, Huttner WB. An adaptive threshold in mammalian neocortical evolution. PLoS Biol 2014; 12:e1002000. [PMID: 25405475 PMCID: PMC4236020 DOI: 10.1371/journal.pbio.1002000] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 10/09/2014] [Indexed: 01/19/2023] Open
Abstract
A study of the evolutionary history of cortical folding in mammals, its relationship to physiological and life-history traits and the underlying cortical progenitor behavior during embryogenesis, explains the diversity of folding we see across modern mammals. The diversity of neocortical folding among mammals can be explained by two distinct neurogenic programs, which give rise to mammals with a highly folded neocortex and mammals with slightly folded or unfolded neocortex, each occupying a distinct ecological niche. Expansion of the neocortex is a hallmark of human evolution. However, determining which adaptive mechanisms facilitated its expansion remains an open question. Here we show, using the gyrencephaly index (GI) and other physiological and life-history data for 102 mammalian species, that gyrencephaly is an ancestral mammalian trait. We find that variation in GI does not evolve linearly across species, but that mammals constitute two principal groups above and below a GI threshold value of 1.5, approximately equal to 109 neurons, which may be characterized by distinct constellations of physiological and life-history traits. By integrating data on neurogenic period, neuroepithelial founder pool size, cell-cycle length, progenitor-type abundances, and cortical neuron number into discrete mathematical models, we identify symmetric proliferative divisions of basal progenitors in the subventricular zone of the developing neocortex as evolutionarily necessary for generating a 14-fold increase in daily prenatal neuron production, traversal of the GI threshold, and thus establishment of two principal groups. We conclude that, despite considerable neuroanatomical differences, changes in the length of the neurogenic period alone, rather than any novel neurogenic progenitor lineage, are sufficient to explain differences in neuron number and neocortical size between species within the same principal group. What are the key differences in the development and evolution of the cerebral cortex that underlie the differences in its size and degree of folding across mammals? Here, we present phylogenetic evidence that the Jurassic era mammalian ancestor may have been a relatively large-brained species with a folded neocortex. We then show that variation in the degree of cortical folding (gyrencephaly index [GI]) does not evolve linearly across species, as previously assumed, but that mammals fall into two principal groups associated with distinct ecological niches: low-GI mammals (such as mice and tarsiers) and high-GI mammals (such as dolphins and humans), which are found to generate on average 14-fold more brain weight per day of gestation. This greater daily brain weight production in mammals with a highly folded neocortex requires a specific class of progenitor cell-type to adopt a special mode of cell division, which is absent in mammals with slightly folded or unfolded neocortices. Differences among mammals within the same GI group (high or low) are not due to different programming, but rather the result of differences in the length of the neurogenic period. So, the impressively large and folded human neocortex, which is three times the size of the chimpanzee neocortex, can be explained by a modest evolutionary extension of the neurogenic period with respect to its closest primate ancestors.
Collapse
Affiliation(s)
- Eric Lewitus
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (EL); (WBH)
| | - Iva Kelava
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Alex T. Kalinka
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Pavel Tomancak
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (EL); (WBH)
| |
Collapse
|
146
|
Richards GS, Rentzsch F. Transgenic analysis of a SoxB gene reveals neural progenitor cells in the cnidarian Nematostella vectensis. Development 2014; 141:4681-9. [PMID: 25395455 DOI: 10.1242/dev.112029] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Bilaterian neurogenesis is characterized by the generation of diverse neural cell types from dedicated neural stem/progenitor cells (NPCs). However, the evolutionary origin of NPCs is unclear, as neurogenesis in representatives of the bilaterian sister group, the Cnidaria, occurs via interstitial stem cells that also possess broader, non-neural, developmental potential. We address this question by analysing neurogenesis in an anthozoan cnidarian, Nematostella vectensis. Using a transgenic reporter line, we show that NvSoxB(2) - an orthologue of bilaterian SoxB genes that have conserved roles in neurogenesis - is expressed in a cell population that gives rise to sensory neurons, ganglion neurons and nematocytes: the three primary neural cell types of cnidarians. EdU labelling together with in situ hybridization, and within the NvSoxB(2)::mOrange transgenic line, demonstrates that cells express NvSoxB(2) before mitosis and identifies asymmetric behaviours of sibling cells within NvSoxB(2)(+) lineages. Morpholino-mediated gene knockdown of NvSoxB(2) blocks the formation of all three neural cell types, thereby identifying NvSoxB(2) as an essential positive regulator of nervous system development. Our results demonstrate that diverse neural cell types derive from an NvSoxB(2)-expressing population of mitotic cells in Nematostella and that SoxB genes are ancient components of a neurogenic program. To our knowledge this is the first description of a lineage-restricted, multipotent cell population outside the Bilateria and we propose that neurogenesis via dedicated, SoxB-expressing NPCs predates the split between cnidarians and bilaterians.
Collapse
Affiliation(s)
- Gemma Sian Richards
- Sars Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, Bergen N-5008, Norway
| | - Fabian Rentzsch
- Sars Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, Bergen N-5008, Norway
| |
Collapse
|
147
|
Yamada T, Minoda R, Miwa T, Ise M, Takeda H, Yumoto E. Neurogenesis of the spiral ganglion cells in the cochlea requires the transcriptional cofactor TIS21. Neurosci Lett 2014; 584:265-9. [PMID: 25451728 DOI: 10.1016/j.neulet.2014.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 12/25/2022]
Abstract
The molecular mechanisms controlling the proliferation and differentiation of spiral ganglion cells (SGCs) in the inner ear are still largely unknown. TIS21 is a transcriptional cofactor that shows antiproliferative, antiapoptotic, and prodifferentiative effects on neural progenitor cells. To investigate the function of TIS21 during SGC development, we analyzed SGC neurogenesis from embryonic day 13.5 (E13.5) to postnatal day 4 (P4) in Tis21-GFP knock-in mice, in which the protein-encoding exon of the Tis21 gene was replaced by EGFP. Through E13.5 to P4, we found fewer SGCs in homozygous Tis21-GFP knock-in mice than in wild-type mice. Our results suggest that TIS21 is required for development of SGCs. Deleting Tis21 may affect progenitor cells or neuroblasts at the beginning of cochlear-vestibular ganglion formation and would consequently lead to a decrease in the number of SGCs.
Collapse
Affiliation(s)
- Takao Yamada
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Graduate School of Medicine, 1-1-1 Honjo Chuouku, Kumamoto city, Kumamoto 860-8556, Japan
| | - Ryosei Minoda
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Graduate School of Medicine, 1-1-1 Honjo Chuouku, Kumamoto city, Kumamoto 860-8556, Japan.
| | - Toru Miwa
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Graduate School of Medicine, 1-1-1 Honjo Chuouku, Kumamoto city, Kumamoto 860-8556, Japan
| | - Momoko Ise
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Graduate School of Medicine, 1-1-1 Honjo Chuouku, Kumamoto city, Kumamoto 860-8556, Japan
| | - Hiroki Takeda
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Graduate School of Medicine, 1-1-1 Honjo Chuouku, Kumamoto city, Kumamoto 860-8556, Japan
| | - Eiji Yumoto
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Graduate School of Medicine, 1-1-1 Honjo Chuouku, Kumamoto city, Kumamoto 860-8556, Japan
| |
Collapse
|
148
|
Cyclin D1 acts as a barrier to pluripotent reprogramming by promoting neural progenitor fate commitment. FEBS Lett 2014; 588:4008-17. [DOI: 10.1016/j.febslet.2014.08.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/11/2014] [Accepted: 08/27/2014] [Indexed: 11/21/2022]
|
149
|
Gonsalvez DG, Li-Yuen-Fong M, Cane KN, Stamp LA, Young HM, Anderson CR. Different neural crest populations exhibit diverse proliferative behaviors. Dev Neurobiol 2014; 75:287-301. [DOI: 10.1002/dneu.22229] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/16/2014] [Accepted: 09/02/2014] [Indexed: 01/02/2023]
Affiliation(s)
- David G. Gonsalvez
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Mathew Li-Yuen-Fong
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Kylie N. Cane
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Lincon A. Stamp
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Heather M. Young
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Colin R. Anderson
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| |
Collapse
|
150
|
Chavali PL, Pütz M, Gergely F. Small organelle, big responsibility: the role of centrosomes in development and disease. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130468. [PMID: 25047622 PMCID: PMC4113112 DOI: 10.1098/rstb.2013.0468] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The centrosome, a key microtubule organizing centre, is composed of centrioles, embedded in a protein-rich matrix. Centrosomes control the internal spatial organization of somatic cells, and as such contribute to cell division, cell polarity and migration. Upon exiting the cell cycle, most cell types in the human body convert their centrioles into basal bodies, which drive the assembly of primary cilia, involved in sensing and signal transduction at the cell surface. Centrosomal genes are targeted by mutations in numerous human developmental disorders, ranging from diseases exclusively affecting brain development, through global growth failure syndromes to diverse pathologies associated with ciliary malfunction. Despite our much-improved understanding of centrosome function in cellular processes, we know remarkably little of its role in the organismal context, especially in mammals. In this review, we examine how centrosome dysfunction impacts on complex physiological processes and speculate on the challenges we face when applying knowledge generated from in vitro and in vivo model systems to human development.
Collapse
Affiliation(s)
- Pavithra L Chavali
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Monika Pütz
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|