1451
|
Offerdahl DK, Dorward DW, Hansen BT, Bloom ME. Cytoarchitecture of Zika virus infection in human neuroblastoma and Aedes albopictus cell lines. Virology 2017; 501:54-62. [PMID: 27863275 PMCID: PMC5201448 DOI: 10.1016/j.virol.2016.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 01/22/2023]
Abstract
The Zika virus (ZIKV) pandemic is a global concern due to its role in the development of congenital anomalies of the central nervous system. This mosquito-borne flavivirus alternates between mammalian and mosquito hosts, but information about the biogenesis of ZIKV is limited. Using a human neuroblastoma cell line (SK-N-SH) and an Aedes albopictus mosquito cell line (C6/36), we characterized ZIKV infection by immunofluorescence, transmission electron microscopy (TEM), and electron tomography (ET) to better understand infection in these disparate host cells. ZIKV replicated well in both cell lines, but infected SK-N-SH cells suffered a lytic crisis. Flaviviruses scavenge host cell membranes to serve as replication platforms and ZIKV showed the hallmarks of this process. Via TEM, we identified virus particles and 60-100nm spherular vesicles. ET revealed these vesicular replication compartments contain smaller 20-30nm spherular structures. Our studies indicate that SK-N-SH and C6/36 cells are relevant models for viral cytoarchitecture study.
Collapse
Affiliation(s)
- Danielle K Offerdahl
- Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, United States
| | - David W Dorward
- Microscopy Unit, Research Technology Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, United States
| | - Bryan T Hansen
- Microscopy Unit, Research Technology Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, United States
| | - Marshall E Bloom
- Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, United States.
| |
Collapse
|
1452
|
Matsui T, Nieto-Estévez V, Kyrychenko S, Schneider JW, Hsieh J. Retinoblastoma protein controls growth, survival and neuronal migration in human cerebral organoids. Development 2017; 144:1025-1034. [PMID: 28087635 DOI: 10.1242/dev.143636] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/16/2016] [Indexed: 01/22/2023]
Abstract
The tumor suppressor retinoblastoma protein (RB) regulates S-phase cell cycle entry via E2F transcription factors. Knockout (KO) mice have shown that RB plays roles in cell migration, differentiation and apoptosis, in developing and adult brain. In addition, the RB family is required for self-renewal and survival of human embryonic stem cells (hESCs). Since little is known about the role of RB in human brain development, we investigated its function in cerebral organoids differentiated from gene-edited hESCs lacking RB. We show that RB is abundantly expressed in neural stem and progenitor cells in organoids at 15 and 28 days of culture. RB loss promoted S-phase entry in DCX+ cells and increased apoptosis in Sox2+ neural stem and progenitor cells, and in DCX+ and Tuj1+ neurons. Associated with these cell cycle and pro-apoptotic effects, we observed increased CCNA2 and BAX gene expression, respectively. Moreover, we observed aberrant Tuj1+ neuronal migration in RB-KO organoids and upregulation of the gene encoding VLDLR, a receptor important in reelin signaling. Corroborating the results in RB-KO organoids in vitro, we observed ectopically localized Tuj1+ cells in RB-KO teratomas grown in vivo Taken together, these results identify crucial functions for RB in the cerebral organoid model of human brain development.
Collapse
Affiliation(s)
- Takeshi Matsui
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vanesa Nieto-Estévez
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sergii Kyrychenko
- Department of Internal Medicine and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jay W Schneider
- Department of Internal Medicine and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenny Hsieh
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
1453
|
Clevers H. Modeling Development and Disease with Organoids. Cell 2017; 165:1586-1597. [PMID: 27315476 DOI: 10.1016/j.cell.2016.05.082] [Citation(s) in RCA: 1987] [Impact Index Per Article: 248.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 01/10/2023]
Abstract
Recent advances in 3D culture technology allow embryonic and adult mammalian stem cells to exhibit their remarkable self-organizing properties, and the resulting organoids reflect key structural and functional properties of organs such as kidney, lung, gut, brain and retina. Organoid technology can therefore be used to model human organ development and various human pathologies 'in a dish." Additionally, patient-derived organoids hold promise to predict drug response in a personalized fashion. Organoids open up new avenues for regenerative medicine and, in combination with editing technology, for gene therapy. The many potential applications of this technology are only beginning to be explored.
Collapse
Affiliation(s)
- Hans Clevers
- Hubrecht Institute/Royal Netherlands Academy of Arts and Sciences, Princess Maxima Centre and University Medical Centre Utrecht, 3584CT Utrecht, The Netherlands.
| |
Collapse
|
1454
|
Characterization of cytopathic factors through genome-wide analysis of the Zika viral proteins in fission yeast. Proc Natl Acad Sci U S A 2017; 114:E376-E385. [PMID: 28049830 DOI: 10.1073/pnas.1619735114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The Zika virus (ZIKV) causes microcephaly and the Guillain-Barré syndrome. Little is known about how ZIKV causes these conditions or which ZIKV viral protein(s) is responsible for the associated ZIKV-induced cytopathic effects, including cell hypertrophy, growth restriction, cell-cycle dysregulation, and cell death. We used fission yeast for the rapid, global functional analysis of the ZIKV genome. All 14 proteins or small peptides were produced under an inducible promoter, and we measured the intracellular localization and the specific effects on ZIKV-associated cytopathic activities of each protein. The subcellular localization of each ZIKV protein was in overall agreement with its predicted protein structure. Five structural and two nonstructural ZIKV proteins showed various levels of cytopathic effects. The expression of these ZIKV proteins restricted cell proliferation, induced hypertrophy, or triggered cellular oxidative stress leading to cell death. The expression of premembrane protein (prM) resulted in cell-cycle G1 accumulation, whereas membrane-anchored capsid (anaC), membrane protein (M), envelope protein (E), and nonstructural protein 4A (NS4A) caused cell-cycle G2/M accumulation. A mechanistic study revealed that NS4A-induced cellular hypertrophy and growth restriction were mediated specifically through the target of rapamycin (TOR) cellular stress pathway involving Tor1 and type 2A phosphatase activator Tip41. These findings should provide a reference for future research on the prevention and treatment of ZIKV diseases.
Collapse
|
1455
|
Krauer F, Riesen M, Reveiz L, Oladapo OT, Martínez-Vega R, Porgo TV, Haefliger A, Broutet NJ, Low N, WHO Zika Causality Working Group. Zika Virus Infection as a Cause of Congenital Brain Abnormalities and Guillain-Barré Syndrome: Systematic Review. PLoS Med 2017; 14:e1002203. [PMID: 28045901 PMCID: PMC5207634 DOI: 10.1371/journal.pmed.1002203] [Citation(s) in RCA: 325] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/16/2016] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The World Health Organization (WHO) stated in March 2016 that there was scientific consensus that the mosquito-borne Zika virus was a cause of the neurological disorder Guillain-Barré syndrome (GBS) and of microcephaly and other congenital brain abnormalities based on rapid evidence assessments. Decisions about causality require systematic assessment to guide public health actions. The objectives of this study were to update and reassess the evidence for causality through a rapid and systematic review about links between Zika virus infection and (a) congenital brain abnormalities, including microcephaly, in the foetuses and offspring of pregnant women and (b) GBS in any population, and to describe the process and outcomes of an expert assessment of the evidence about causality. METHODS AND FINDINGS The study had three linked components. First, in February 2016, we developed a causality framework that defined questions about the relationship between Zika virus infection and each of the two clinical outcomes in ten dimensions: temporality, biological plausibility, strength of association, alternative explanations, cessation, dose-response relationship, animal experiments, analogy, specificity, and consistency. Second, we did a systematic review (protocol number CRD42016036693). We searched multiple online sources up to May 30, 2016 to find studies that directly addressed either outcome and any causality dimension, used methods to expedite study selection, data extraction, and quality assessment, and summarised evidence descriptively. Third, WHO convened a multidisciplinary panel of experts who assessed the review findings and reached consensus statements to update the WHO position on causality. We found 1,091 unique items up to May 30, 2016. For congenital brain abnormalities, including microcephaly, we included 72 items; for eight of ten causality dimensions (all except dose-response relationship and specificity), we found that more than half the relevant studies supported a causal association with Zika virus infection. For GBS, we included 36 items, of which more than half the relevant studies supported a causal association in seven of ten dimensions (all except dose-response relationship, specificity, and animal experimental evidence). Articles identified nonsystematically from May 30 to July 29, 2016 strengthened the review findings. The expert panel concluded that (a) the most likely explanation of available evidence from outbreaks of Zika virus infection and clusters of microcephaly is that Zika virus infection during pregnancy is a cause of congenital brain abnormalities including microcephaly, and (b) the most likely explanation of available evidence from outbreaks of Zika virus infection and GBS is that Zika virus infection is a trigger of GBS. The expert panel recognised that Zika virus alone may not be sufficient to cause either congenital brain abnormalities or GBS but agreed that the evidence was sufficient to recommend increased public health measures. Weaknesses are the limited assessment of the role of dengue virus and other possible cofactors, the small number of comparative epidemiological studies, and the difficulty in keeping the review up to date with the pace of publication of new research. CONCLUSIONS Rapid and systematic reviews with frequent updating and open dissemination are now needed both for appraisal of the evidence about Zika virus infection and for the next public health threats that will emerge. This systematic review found sufficient evidence to say that Zika virus is a cause of congenital abnormalities and is a trigger of GBS.
Collapse
Affiliation(s)
- Fabienne Krauer
- Institute of Social and Preventive Medicine, University of Bern, Switzerland
| | - Maurane Riesen
- Institute of Social and Preventive Medicine, University of Bern, Switzerland
| | - Ludovic Reveiz
- Pan American Health Organization, Washington DC, United States of America
| | - Olufemi T. Oladapo
- UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Ruth Martínez-Vega
- Escuela de Microbiologia, Universidad Industrial de Santander, Santander, Colombia
| | - Teegwendé V. Porgo
- UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Reproductive Health and Research, World Health Organization, Geneva, Switzerland
- Department of Social and Preventative Medicine, Laval University, Québec, Canada
| | - Anina Haefliger
- Institute of Social and Preventive Medicine, University of Bern, Switzerland
| | - Nathalie J. Broutet
- UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Switzerland
| | | |
Collapse
|
1456
|
Sauerzopf U, Sacco R, Novarino G, Niello M, Weidenauer A, Praschak‐Rieder N, Sitte H, Willeit M, Bolam P. Are reprogrammed cells a useful tool for studying dopamine dysfunction in psychotic disorders? A review of the current evidence. Eur J Neurosci 2017; 45:45-57. [PMID: 27690184 PMCID: PMC5811827 DOI: 10.1111/ejn.13418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/23/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022]
Abstract
Since 2006, reprogrammed cells have increasingly been used as a biomedical research technique in addition to neuro-psychiatric methods. These rapidly evolving techniques allow for the generation of neuronal sub-populations, and have sparked interest not only in monogenetic neuro-psychiatric diseases, but also in poly-genetic and poly-aetiological disorders such as schizophrenia (SCZ) and bipolar disorder (BPD). This review provides a summary of 19 publications on reprogrammed adult somatic cells derived from patients with SCZ, and five publications using this technique in patients with BPD. As both disorders are complex and heterogeneous, there is a plurality of hypotheses to be tested in vitro. In SCZ, data on alterations of dopaminergic transmission in vitro are sparse, despite the great explanatory power of the so-called DA hypothesis of SCZ. Some findings correspond to perturbations of cell energy metabolism, and observations in reprogrammed cells suggest neuro-developmental alterations. Some studies also report on the efficacy of medicinal compounds to revert alterations observed in cellular models. However, due to the paucity of replication studies, no comprehensive conclusions can be drawn from studies using reprogrammed cells at the present time. In the future, findings from cell culture methods need to be integrated with clinical, epidemiological, pharmacological and imaging data in order to generate a more comprehensive picture of SCZ and BPD.
Collapse
Affiliation(s)
- Ulrich Sauerzopf
- Department of Psychiatry and PsychotherapyMedical University of ViennaWähringer Gürtel 18‐201090ViennaAustria
| | - Roberto Sacco
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Gaia Novarino
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Marco Niello
- Institute of PharmacologyMedical University of ViennaViennaAustria
| | - Ana Weidenauer
- Department of Psychiatry and PsychotherapyMedical University of ViennaWähringer Gürtel 18‐201090ViennaAustria
| | - Nicole Praschak‐Rieder
- Department of Psychiatry and PsychotherapyMedical University of ViennaWähringer Gürtel 18‐201090ViennaAustria
| | - Harald Sitte
- Institute of PharmacologyMedical University of ViennaViennaAustria
| | - Matthäus Willeit
- Department of Psychiatry and PsychotherapyMedical University of ViennaWähringer Gürtel 18‐201090ViennaAustria
| | | |
Collapse
|
1457
|
Zhu Y, Wang L, Yin F, Yu Y, Wang Y, Shepard MJ, Zhuang Z, Qin J. Probing impaired neurogenesis in human brain organoids exposed to alcohol. Integr Biol (Camb) 2017; 9:968-978. [DOI: 10.1039/c7ib00105c] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The fetal brain is highly vulnerable to ethanol exposure, which can trigger various long-term neuronal disabilities and cognitive dysfunctions.
Collapse
Affiliation(s)
- Yujuan Zhu
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- China
- University of Chinese Academy of Sciences
| | - Li Wang
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- China
- Key Laboratory of Separation Sciences for Analytical Chemistry
| | - Fangchao Yin
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- China
- University of Chinese Academy of Sciences
| | - Yue Yu
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- China
- University of Chinese Academy of Sciences
| | - Yaqing Wang
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- China
- University of Chinese Academy of Sciences
| | - Matthew J. Shepard
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke
- National Institutes of Health
- Bethesda
- USA
- Department of Neurological Surgery
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke
- National Institutes of Health
- Bethesda
- USA
| | - Jianhua Qin
- Division of Biotechnology
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- China
- University of Chinese Academy of Sciences
| |
Collapse
|
1458
|
Hester ME, Hood AB. Generation of Cerebral Organoids Derived from Human Pluripotent Stem Cells. NEUROMETHODS 2017. [DOI: 10.1007/978-1-4939-7024-7_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
1459
|
Systematic time-dependent visualization and quantitation of the neurogenic rate in brain organoids. Biochem Biophys Res Commun 2017; 483:94-100. [DOI: 10.1016/j.bbrc.2016.12.187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/31/2016] [Indexed: 11/17/2022]
|
1460
|
Zhu Y, Wang L, Yin F, Yu Y, Wang Y, Liu H, Wang H, Sun N, Liu H, Qin J. A hollow fiber system for simple generation of human brain organoids. Integr Biol (Camb) 2017; 9:774-781. [DOI: 10.1039/c7ib00080d] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Here we present a novel approach to engineer hiPSC-derived brain organoids within hollow fibers in a simple and high throughput manner by integrating biomaterials with stem cell biology.
Collapse
|
1461
|
Li Y, Muffat J, Omer A, Bosch I, Lancaster MA, Sur M, Gehrke L, Knoblich JA, Jaenisch R. Induction of Expansion and Folding in Human Cerebral Organoids. Cell Stem Cell 2016; 20:385-396.e3. [PMID: 28041895 DOI: 10.1016/j.stem.2016.11.017] [Citation(s) in RCA: 314] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/24/2016] [Accepted: 11/29/2016] [Indexed: 01/09/2023]
Abstract
An expansion of the cerebral neocortex is thought to be the foundation for the unique intellectual abilities of humans. It has been suggested that an increase in the proliferative potential of neural progenitors (NPs) underlies the expansion of the cortex and its convoluted appearance. Here we show that increasing NP proliferation induces expansion and folding in an in vitro model of human corticogenesis. Deletion of PTEN stimulates proliferation and generates significantly larger and substantially folded cerebral organoids. This genetic modification allows sustained cell cycle re-entry, expansion of the progenitor population, and delayed neuronal differentiation, all key features of the developing human cortex. In contrast, Pten deletion in mouse organoids does not lead to folding. Finally, we utilized the expanded cerebral organoids to show that infection with Zika virus impairs cortical growth and folding. Our study provides new insights into the mechanisms regulating the structure and organization of the human cortex.
Collapse
Affiliation(s)
- Yun Li
- The Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Julien Muffat
- The Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Attya Omer
- The Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Irene Bosch
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Madeline A Lancaster
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Mriganka Sur
- The Picower Institute for Learning and Memory, Cambridge, MA 02139, USA
| | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Rudolf Jaenisch
- The Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA.
| |
Collapse
|
1462
|
Kelava I, Lancaster MA. Dishing out mini-brains: Current progress and future prospects in brain organoid research. Dev Biol 2016; 420:199-209. [PMID: 27402594 PMCID: PMC5161139 DOI: 10.1016/j.ydbio.2016.06.037] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/16/2016] [Accepted: 06/25/2016] [Indexed: 01/15/2023]
Abstract
The ability to model human brain development in vitro represents an important step in our study of developmental processes and neurological disorders. Protocols that utilize human embryonic and induced pluripotent stem cells can now generate organoids which faithfully recapitulate, on a cell-biological and gene expression level, the early period of human embryonic and fetal brain development. In combination with novel gene editing tools, such as CRISPR, these methods represent an unprecedented model system in the field of mammalian neural development. In this review, we focus on the similarities of current organoid methods to in vivo brain development, discuss their limitations and potential improvements, and explore the future venues of brain organoid research.
Collapse
Affiliation(s)
- Iva Kelava
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, CB2 0QH Cambridge, United Kingdom
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, CB2 0QH Cambridge, United Kingdom.
| |
Collapse
|
1463
|
Ostrem B, Di Lullo E, Kriegstein A. oRGs and mitotic somal translocation - a role in development and disease. Curr Opin Neurobiol 2016; 42:61-67. [PMID: 27978479 DOI: 10.1016/j.conb.2016.11.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/18/2016] [Accepted: 11/22/2016] [Indexed: 12/30/2022]
Abstract
The evolution of the human brain has been characterized by an increase in the size of the neocortex. Underlying this expansion is a significant increase in the number of neurons produced by neural stem cells during early stages of cortical development. Here we highlight recent advances in our understating of these cell populations, consisting of ventricular radial glia and outer radial glia. We highlight how gene expression studies have identified molecular signatures for radial glial cell populations and outline what has been learned about the mechanisms underlying the characteristic mode of division observed in outer radial glia cells, mitotic somal translocation. Understanding the significance of this behavior may help us explain human cortical expansion and further elucidate neurodevelopmental diseases.
Collapse
Affiliation(s)
- Bridget Ostrem
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Elizabeth Di Lullo
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
| | - Arnold Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
1464
|
Transfer of convalescent serum to pregnant mice prevents Zika virus infection and microcephaly in offspring. Cell Res 2016; 27:158-160. [PMID: 27922617 DOI: 10.1038/cr.2016.144] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
1465
|
Abstract
The re-emergence of Zika virus (ZIKV) and its suspected link with various disorders in newborns and adults led the World Health Organization to declare a global health emergency. In response, the stem cell field quickly established platforms for modeling ZIKV exposure using human pluripotent stem cell-derived neural progenitors and brain organoids, fetal tissues, and animal models. These efforts provided significant insight into cellular targets, pathogenesis, and underlying biological mechanisms of ZIKV infection as well as platforms for drug testing. Here we review the remarkable progress in stem cell-based ZIKV research and discuss current challenges and future opportunities.
Collapse
Affiliation(s)
- Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
1466
|
Li H, Saucedo-Cuevas L, Shresta S, Gleeson JG. The Neurobiology of Zika Virus. Neuron 2016; 92:949-958. [DOI: 10.1016/j.neuron.2016.11.031] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/21/2016] [Accepted: 11/17/2016] [Indexed: 01/17/2023]
|
1467
|
Vagaska B, Ferretti P. Toward modeling the human nervous system in a dish: recent progress and outstanding challenges. Regen Med 2016; 12:15-23. [PMID: 27900887 DOI: 10.2217/rme-2016-0106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Studying the cellular and molecular bases governing development, and normal and abnormal functions of the human CNS is hampered by its complexity and the very limited possibility of experimentally manipulating it in vivo. Development of 3D, tissue-like culture systems offers much promise for boosting our understanding of human neural development, birth defects, neurodegenerative diseases and neural injury, and for providing platforms that will more accurately predict efficacy of putative therapeutic compounds and assess responses to potentially neurotoxic agents. Although novel technological developments and a more interdisciplinary approach to modeling the human CNS are accelerating the pace of discovery, increasing the complexity of in vitro systems increases the ordeals to be overcome to establish highly reproducible models amenable to quantitative analysis.
Collapse
Affiliation(s)
- Barbora Vagaska
- Stem Cell & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Patrizia Ferretti
- Stem Cell & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| |
Collapse
|
1468
|
Delvecchio R, Higa LM, Pezzuto P, Valadão AL, Garcez PP, Monteiro FL, Loiola EC, Dias AA, Silva FJM, Aliota MT, Caine EA, Osorio JE, Bellio M, O'Connor DH, Rehen S, de Aguiar RS, Savarino A, Campanati L, Tanuri A. Chloroquine, an Endocytosis Blocking Agent, Inhibits Zika Virus Infection in Different Cell Models. Viruses 2016; 8:v8120322. [PMID: 27916837 PMCID: PMC5192383 DOI: 10.3390/v8120322] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 11/16/2016] [Accepted: 11/18/2016] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV) infection in utero might lead to microcephaly and other congenital defects. Since no specific therapy is available thus far, there is an urgent need for the discovery of agents capable of inhibiting its viral replication and deleterious effects. Chloroquine is widely used as an antimalarial drug, anti-inflammatory agent, and it also shows antiviral activity against several viruses. Here we show that chloroquine exhibits antiviral activity against ZIKV in Vero cells, human brain microvascular endothelial cells, human neural stem cells, and mouse neurospheres. We demonstrate that chloroquine reduces the number of ZIKV-infected cells in vitro, and inhibits virus production and cell death promoted by ZIKV infection without cytotoxic effects. In addition, chloroquine treatment partially reveres morphological changes induced by ZIKV infection in mouse neurospheres.
Collapse
Affiliation(s)
- Rodrigo Delvecchio
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Luiza M Higa
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Paula Pezzuto
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Ana Luiza Valadão
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Patrícia P Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro 22281-100, Brazil.
| | - Fábio L Monteiro
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Erick C Loiola
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro 22281-100, Brazil.
| | - André A Dias
- Department of Immunology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Fábio J M Silva
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Matthew T Aliota
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Elizabeth A Caine
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Jorge E Osorio
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Maria Bellio
- Department of Immunology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Stevens Rehen
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro 22281-100, Brazil.
| | - Renato Santana de Aguiar
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Andrea Savarino
- Istituto Superiore di Sanità, Deptartment of Infectious Diseases, 299 Viale Regina Elena, 00161 Rome, Italy.
| | - Loraine Campanati
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| | - Amilcar Tanuri
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
1469
|
Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci U S A 2016; 113:14408-14413. [PMID: 27911847 DOI: 10.1073/pnas.1618029113] [Citation(s) in RCA: 356] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The rapid spread of Zika virus (ZIKV) and its association with abnormal brain development constitute a global health emergency. Congenital ZIKV infection produces a range of mild to severe pathologies, including microcephaly. To understand the pathophysiology of ZIKV infection, we used models of the developing brain that faithfully recapitulate the tissue architecture in early to midgestation. We identify the brain cell populations that are most susceptible to ZIKV infection in primary human tissue, provide evidence for a mechanism of viral entry, and show that a commonly used antibiotic protects cultured brain cells by reducing viral proliferation. In the brain, ZIKV preferentially infected neural stem cells, astrocytes, oligodendrocyte precursor cells, and microglia, whereas neurons were less susceptible to infection. These findings suggest mechanisms for microcephaly and other pathologic features of infants with congenital ZIKV infection that are not explained by neural stem cell infection alone, such as calcifications in the cortical plate. Furthermore, we find that blocking the glia-enriched putative viral entry receptor AXL reduced ZIKV infection of astrocytes in vitro, and genetic knockdown of AXL in a glial cell line nearly abolished infection. Finally, we evaluate 2,177 compounds, focusing on drugs safe in pregnancy. We show that the macrolide antibiotic azithromycin reduced viral proliferation and virus-induced cytopathic effects in glial cell lines and human astrocytes. Our characterization of infection in the developing human brain clarifies the pathogenesis of congenital ZIKV infection and provides the basis for investigating possible therapeutic strategies to safely alleviate or prevent the most severe consequences of the epidemic.
Collapse
|
1470
|
Hajra A, Bandyopadhyay D, Heise LR, Bhadra R, Ball S, Hajra SK. Zika and pregnancy: A comprehensive review. Am J Reprod Immunol 2016; 77. [DOI: 10.1111/aji.12607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 10/27/2016] [Indexed: 12/31/2022] Open
Affiliation(s)
- Adrija Hajra
- Department of Internal Medicine; IPGMER; Kolkata India
| | | | - Lyndsey R. Heise
- Department of Internal Medicine; University of Nebraska Medical Center; Omaha NE USA
| | - Rajarshi Bhadra
- Department of Internal Medicine; St Vincent Charity Medical Center, A teaching affiliate to; Case Western Reserve University; Cleveland OH USA
| | - Somedeb Ball
- Department of Cardiology; Apollo Gleneagles Hospital; Kolkata India
| | | |
Collapse
|
1471
|
The Future is The Past: Methylation QTLs in Schizophrenia. Genes (Basel) 2016; 7:genes7120104. [PMID: 27886132 PMCID: PMC5192480 DOI: 10.3390/genes7120104] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/13/2016] [Accepted: 11/16/2016] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies (GWAS) have remarkably advanced insight into the genetic basis of schizophrenia (SCZ). Still, most of the functional variance in disease risk remains unexplained. Hence, there is a growing need to map genetic variability-to-genes-to-functions for understanding the pathophysiology of SCZ and the development of better treatments. Genetic variation can regulate various cellular functions including DNA methylation, an epigenetic mark with important roles in transcription and the mediation of environmental influences. Methylation quantitative trait loci (meQTLs) are derived by mapping levels of DNA methylation in genetically different, genotyped individuals and define loci at which DNA methylation is influenced by genetic variation. Recent evidence points to an abundance of meQTLs in brain tissues whose functional contributions to development and mental diseases are still poorly understood. Interestingly, fetal meQTLs reside in regulatory domains affecting methylome reconfiguration during early brain development and are enriched in loci identified by GWAS for SCZ. Moreover, fetal meQTLs are preserved in the adult brain and could trace early epigenomic deregulation during vulnerable periods. Overall, these findings highlight the role of fetal meQTLs in the genetic risk for and in the possible neurodevelopmental origin of SCZ.
Collapse
|
1472
|
Mini-Brains Make Big Leaps for Studying Human Neural Development and Disease. Epilepsy Curr 2016; 16:402-404. [PMID: 27857624 DOI: 10.5698/1535-7511-16.6.402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
1473
|
Manangeeswaran M, Ireland DDC, Verthelyi D. Zika (PRVABC59) Infection Is Associated with T cell Infiltration and Neurodegeneration in CNS of Immunocompetent Neonatal C57Bl/6 Mice. PLoS Pathog 2016; 12:e1006004. [PMID: 27855206 PMCID: PMC5113993 DOI: 10.1371/journal.ppat.1006004] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/16/2016] [Indexed: 02/04/2023] Open
Abstract
The recent spread of Zika virus (ZIKV) and its association with increased rates of Guillain Barre and other neurological disorders as well as congenital defects that include microcephaly has created an urgent need to develop animal models to examine the pathogenesis of the disease and explore the efficacy of potential therapeutics and vaccines. Recently developed infection models for ZIKV utilize mice defective in interferon responses. In this study we establish and characterize a new model of peripheral ZIKV infection using immunocompetent neonatal C57BL/6 mice and compare its clinical progression, virus distribution, immune response, and neuropathology with that of C57BL/6-IFNAR KO mice. We show that while ZIKV infected IFNAR KO mice develop bilateral hind limb paralysis and die 5–6 days post-infection (dpi), immunocompetent B6 WT mice develop signs of neurological disease including unsteady gait, kinetic tremors, severe ataxia and seizures by 13 dpi that subside gradually over 2 weeks. Immunohistochemistry show viral antigen predominantly in cerebellum at the peak of the disease in both models. However, whereas IFNAR KO mice showed infiltration by neutrophils and macrophages and higher expression of IL-1, IL-6 and Cox2, B6 WT mice show a cellular infiltration in the CNS composed predominantly of T cells, particularly CD8+ T cells, and increased mRNA expression levels of IFNg, GzmB and Prf1 at peak of disease. Lastly, the CNS of B6 WT mice shows evidence of neurodegeneration predominantly in the cerebellum that are less prominent in mice lacking the IFN response possibly due to the difference in cellular infiltrates and rapid progression of the disease in that model. The development of the B6 WT model of ZIKV infection will provide insight into the immunopathology of the virus and facilitate assessments of possible therapeutics and vaccines. The recent spread of Zika virus (ZIKV) and its association with increased rates of neurological disorders and congenital defects created an urgent need for animal models to examine the pathogenesis of the disease and explore the efficacy of potential therapeutics and vaccines. We describe the first symptomatic PRVABC59(ZIKV) animal model in immunocompetent B6 WT mice showing that a subcutaneous challenge in 1 day old mice leads to non-lethal neurological disease that is characterized by unsteady gait, kinetic tremors, severe ataxia and seizures that subsides after 2 weeks. ZIKV infects neurons in cerebellum of mice and elicits the infiltration of lymphocytes into the brain. The immune response protects mice from death but may also contribute to neurodegeneration as mice with defective interferon responses have increased virus loads in brain and peripheral organs, succumbing to the disease in 5–6 days, but have fewer signs of neurodegeneration. This mouse model bypasses transplacental transmission and consequent placental insufficiency and will facilitate detailed investigations into the pathogenesis of the disease as well as mechanistic studies for possible therapeutics and vaccines. Lastly, its non-lethal outcome allows for studies assessing the long term effects of the infection, and exploring conditions that could lead to disease reactivation.
Collapse
Affiliation(s)
- Mohanraj Manangeeswaran
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Derek D. C. Ireland
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Daniela Verthelyi
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
1474
|
Rajah MM, Pardy RD, Condotta SA, Richer MJ, Sagan SM. Zika Virus: Emergence, Phylogenetics, Challenges, and Opportunities. ACS Infect Dis 2016; 2:763-772. [PMID: 27704772 DOI: 10.1021/acsinfecdis.6b00161] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Zika virus (ZIKV) is an emerging arthropod-borne pathogen that has recently gained notoriety due to its rapid and ongoing geographic expansion and its novel association with neurological complications. Reports of ZIKV-associated Guillain-Barré syndrome as well as fetal microcephaly place emphasis on the need to develop preventative measures and therapeutics to combat ZIKV infection. Thus, it is imperative that models to study ZIKV replication and pathogenesis and the immune response are developed in conjunction with integrated vector control strategies to mount an efficient response to the pandemic. This paper summarizes the current state of knowledge on ZIKV, including the clinical features, phylogenetic analyses, pathogenesis, and the immune response to infection. Potential challenges in developing diagnostic tools, treatment, and prevention strategies are also discussed.
Collapse
Affiliation(s)
- Maaran M. Rajah
- Department of Microbiology and
Immunology and ‡Microbiome and Disease Tolerance Centre (MDTC), McGill University, Montréal, Québec, Canada H3A 2B4
| | - Ryan D. Pardy
- Department of Microbiology and
Immunology and ‡Microbiome and Disease Tolerance Centre (MDTC), McGill University, Montréal, Québec, Canada H3A 2B4
| | - Stephanie A. Condotta
- Department of Microbiology and
Immunology and ‡Microbiome and Disease Tolerance Centre (MDTC), McGill University, Montréal, Québec, Canada H3A 2B4
| | - Martin J. Richer
- Department of Microbiology and
Immunology and ‡Microbiome and Disease Tolerance Centre (MDTC), McGill University, Montréal, Québec, Canada H3A 2B4
| | - Selena M. Sagan
- Department of Microbiology and
Immunology and ‡Microbiome and Disease Tolerance Centre (MDTC), McGill University, Montréal, Québec, Canada H3A 2B4
| |
Collapse
|
1475
|
Christian KM. Modeling Brain Development Using Human Cells for the Study and Treatment of Zika Virus Infections. Curr Behav Neurosci Rep 2016. [DOI: 10.1007/s40473-016-0101-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1476
|
ZIKA virus elicits P53 activation and genotoxic stress in human neural progenitors similar to mutations involved in severe forms of genetic microcephaly. Cell Death Dis 2016; 7:e2440. [PMID: 27787521 PMCID: PMC5133962 DOI: 10.1038/cddis.2016.266] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 12/30/2022]
Abstract
Epidemiological evidence from the current outbreak of Zika virus (ZIKV) and recent studies in animal models indicate a strong causal link between ZIKV and microcephaly. ZIKV infection induces cell-cycle arrest and apoptosis in proliferating neural progenitors. However, the mechanisms leading to these phenotypes are still largely obscure. In this report, we explored the possible similarities between transcriptional responses induced by ZIKV in human neural progenitors and those elicited by three different genetic mutations leading to severe forms of microcephaly in mice. We found that the strongest similarity between all these conditions is the activation of common P53 downstream genes. In agreement with these observations, we report that ZIKV infection increases total P53 levels and nuclear accumulation, as well as P53 Ser15 phosphorylation, correlated with genotoxic stress and apoptosis induction. Interestingly, increased P53 activation and apoptosis are induced not only in cells expressing high levels of viral antigens but also in cells showing low or undetectable levels of the same proteins. These results indicate that P53 activation is an early and specific event in ZIKV-infected cells, which could result from cell-autonomous and/or non-cell-autonomous mechanisms. Moreover, we highlight a small group of P53 effector proteins that could act as critical mediators, not only in ZIKV-induced microcephaly but also in many genetic microcephaly syndromes.
Collapse
|
1477
|
Yao Z, Mich JK, Ku S, Menon V, Krostag AR, Martinez RA, Furchtgott L, Mulholland H, Bort S, Fuqua MA, Gregor BW, Hodge RD, Jayabalu A, May RC, Melton S, Nelson AM, Ngo NK, Shapovalova NV, Shehata SI, Smith MW, Tait LJ, Thompson CL, Thomsen ER, Ye C, Glass IA, Kaykas A, Yao S, Phillips JW, Grimley JS, Levi BP, Wang Y, Ramanathan S. A Single-Cell Roadmap of Lineage Bifurcation in Human ESC Models of Embryonic Brain Development. Cell Stem Cell 2016; 20:120-134. [PMID: 28094016 DOI: 10.1016/j.stem.2016.09.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 07/18/2016] [Accepted: 09/29/2016] [Indexed: 01/29/2023]
Abstract
During human brain development, multiple signaling pathways generate diverse cell types with varied regional identities. Here, we integrate single-cell RNA sequencing and clonal analyses to reveal lineage trees and molecular signals underlying early forebrain and mid/hindbrain cell differentiation from human embryonic stem cells (hESCs). Clustering single-cell transcriptomic data identified 41 distinct populations of progenitor, neuronal, and non-neural cells across our differentiation time course. Comparisons with primary mouse and human gene expression data demonstrated rostral and caudal progenitor and neuronal identities from early brain development. Bayesian analyses inferred a unified cell-type lineage tree that bifurcates between cortical and mid/hindbrain cell types. Two methods of clonal analyses confirmed these findings and further revealed the importance of Wnt/β-catenin signaling in controlling this lineage decision. Together, these findings provide a rich transcriptome-based lineage map for studying human brain development and modeling developmental disorders.
Collapse
Affiliation(s)
- Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - John K Mich
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Sherman Ku
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Vilas Menon
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Leon Furchtgott
- Molecular and Cellular Biology Department, Harvard University, Cambridge, MA 02138, USA
| | | | - Susan Bort
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Ben W Gregor
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Anu Jayabalu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ryan C May
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Samuel Melton
- School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA
| | | | - N Kiet Ngo
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Leah J Tait
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Chaoyang Ye
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ian A Glass
- Division of Genetic Medicine, University of Washington, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Ajamete Kaykas
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Shuyuan Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA.
| | - Yanling Wang
- Allen Institute for Brain Science, Seattle, WA 98109, USA.
| | - Sharad Ramanathan
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Molecular and Cellular Biology Department, Harvard University, Cambridge, MA 02138, USA; School of Engineering and Applied Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
1478
|
The promises and challenges of human brain organoids as models of neuropsychiatric disease. Nat Med 2016; 22:1220-1228. [DOI: 10.1038/nm.4214] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/22/2016] [Indexed: 12/12/2022]
|
1479
|
Maestre AM, Fernandez-Sesma A. ZIKV Strains' Different Phenotypes in Human Neural Cells Could be a Hint for the Emergence of the New Clinical Neurological Outcomes. EBioMedicine 2016; 13:35-36. [PMID: 27765639 PMCID: PMC5264277 DOI: 10.1016/j.ebiom.2016.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Ana M Maestre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, United States; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, United States.
| |
Collapse
|
1480
|
Muffat J, Li Y, Jaenisch R. CNS disease models with human pluripotent stem cells in the CRISPR age. Curr Opin Cell Biol 2016; 43:96-103. [PMID: 27768957 DOI: 10.1016/j.ceb.2016.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/19/2016] [Accepted: 10/02/2016] [Indexed: 12/31/2022]
Abstract
In vitro differentiation of human pluripotent stem cells provides a systematic platform to investigate the physiological development and function of the human nervous system, as well as the etiology and consequence when these processes go awry. Recent development in three-dimensional (3D) organotypic culture systems allows modeling of the complex structure formation of the human CNS, and the intricate interactions between various resident neuronal and glial cell types. Combined with an ever-expanding genome editing and regulation toolkit such as CRISPR/Cas9, it is now a possibility to study human neurological disease in the relevant molecular, cellular and anatomical context. In this article, we review recent progress in 3D neural culture and the implications for disease modeling.
Collapse
Affiliation(s)
- Julien Muffat
- Whitehead Institute for Biomedical Research, 9Cambridge Center, Cambridge, MA 02142, United States
| | - Yun Li
- Whitehead Institute for Biomedical Research, 9Cambridge Center, Cambridge, MA 02142, United States
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, 9Cambridge Center, Cambridge, MA 02142, United States.
| |
Collapse
|
1481
|
Temme SJ, Maher BJ, Christian KM. Using Induced Pluripotent Stem Cells to Investigate Complex Genetic Psychiatric Disorders. Curr Behav Neurosci Rep 2016; 3:275-284. [PMID: 28191386 DOI: 10.1007/s40473-016-0100-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Induced pluripotent stem cells (iPSCs) can be generated from human patient tissue samples, differentiated into any somatic cell type, and studied under controlled culture conditions. We review how iPSCs are used to investigate genetic factors and biological mechanisms underlying psychiatric disorders, and considerations for synthesizing data across studies. RECENT FINDINGS Results from patient specific-iPSC studies often reveal cellular phenotypes consistent with postmortem and brain imaging studies. Unpredicted findings illustrate the power of iPSCs as a discovery tool, but may also be attributable to limitations in modeling dynamic neural networks or difficulty in identifying the most affected neural subtype or developmental stage. SUMMARY Technological advances in differentiation protocols and organoid generation will enhance our ability to model the salient pathology underlying psychiatric disorders using iPSCs. The field will also benefit from context-driven interpretations of iPSC studies that recognize all potential sources of variability, including differences in patient symptomatology, genetic risk factors and affected cellular subtype.
Collapse
Affiliation(s)
- Stephanie J Temme
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brady J Maher
- Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kimberly M Christian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
1482
|
Zhang F, Hammack C, Ogden SC, Cheng Y, Lee EM, Wen Z, Qian X, Nguyen HN, Li Y, Yao B, Xu M, Xu T, Chen L, Wang Z, Feng H, Huang WK, Yoon KJ, Shan C, Huang L, Qin Z, Christian KM, Shi PY, Xu M, Xia M, Zheng W, Wu H, Song H, Tang H, Ming GL, Jin P. Molecular signatures associated with ZIKV exposure in human cortical neural progenitors. Nucleic Acids Res 2016; 44:8610-8620. [PMID: 27580721 PMCID: PMC5063002 DOI: 10.1093/nar/gkw765] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/21/2016] [Accepted: 08/22/2016] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV) infection causes microcephaly and has been linked to other brain abnormalities. How ZIKV impairs brain development and function is unclear. Here we systematically profiled transcriptomes of human neural progenitor cells exposed to Asian ZIKVC, African ZIKVM, and dengue virus (DENV). In contrast to the robust global transcriptome changes induced by DENV, ZIKV has a more selective and larger impact on expression of genes involved in DNA replication and repair. While overall expression profiles are similar, ZIKVC, but not ZIKVM, induces upregulation of viral response genes and TP53. P53 inhibitors can block the apoptosis induced by both ZIKVC and ZIKVM in hNPCs, with higher potency against ZIKVC-induced apoptosis. Our analyses reveal virus- and strain-specific molecular signatures associated with ZIKV infection. These datasets will help to investigate ZIKV-host interactions and identify neurovirulence determinants of ZIKV.
Collapse
Affiliation(s)
- Feiran Zhang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christy Hammack
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Sarah C Ogden
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Yichen Cheng
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Emily M Lee
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Zhexing Wen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Departments of Psychiatry and Behavioral Science, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xuyu Qian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ha Nam Nguyen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yujing Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Tianlei Xu
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Li Chen
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Zhiqin Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hao Feng
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Wei-Kai Huang
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ki-Jun Yoon
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chao Shan
- Department of Biochemistry & Molecular Biology, Department of Pharmacology & Toxicology, Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Luoxiu Huang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhaohui Qin
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Kimberly M Christian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, Department of Pharmacology & Toxicology, Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mingjiang Xu
- Sylvester Comprehensive Cancer Center, Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892, USA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
1483
|
Shao Q, Herrlinger S, Yang SL, Lai F, Moore JM, Brindley MA, Chen JF. Zika virus infection disrupts neurovascular development and results in postnatal microcephaly with brain damage. Development 2016; 143:4127-4136. [PMID: 27729407 DOI: 10.1242/dev.143768] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022]
Abstract
Zika virus (ZIKV) infection of pregnant women can result in fetal brain abnormalities. It has been established that ZIKV disrupts neural progenitor cells (NPCs) and leads to embryonic microcephaly. However, the fate of other cell types in the developing brain and their contributions to ZIKV-associated brain abnormalities remain largely unknown. Using intracerebral inoculation of embryonic mouse brains, we found that ZIKV infection leads to postnatal growth restriction including microcephaly. In addition to cell cycle arrest and apoptosis of NPCs, ZIKV infection causes massive neuronal death and axonal rarefaction, which phenocopy fetal brain abnormalities in humans. Importantly, ZIKV infection leads to abnormal vascular density and diameter in the developing brain, resulting in a leaky blood-brain barrier (BBB). Massive neuronal death and BBB leakage indicate brain damage, which is further supported by extensive microglial activation and astrogliosis in virally infected brains. Global gene analyses reveal dysregulation of genes associated with immune responses in virus-infected brains. Thus, our data suggest that ZIKV triggers a strong immune response and disrupts neurovascular development, resulting in postnatal microcephaly with extensive brain damage.
Collapse
Affiliation(s)
- Qiang Shao
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Stephanie Herrlinger
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Si-Lu Yang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Fan Lai
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Department of Human Genetics, Miami, FL 33136, USA
| | - Julie M Moore
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Melinda A Brindley
- Department of Infectious Diseases, Department of Population Health and Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Jian-Fu Chen
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
1484
|
Development and characterization of a human embryonic stem cell-derived 3D neural tissue model for neurotoxicity testing. Toxicol In Vitro 2016; 38:124-135. [PMID: 27729293 DOI: 10.1016/j.tiv.2016.10.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 02/08/2023]
Abstract
Alternative models for more rapid compound safety testing are of increasing demand. With emerging techniques using human pluripotent stem cells, the possibility of generating human in vitro models has gained interest, as factors related to species differences could be potentially eliminated. When studying potential neurotoxic effects of a compound it is of crucial importance to have both neurons and glial cells. We have successfully developed a protocol for generating in vitro 3D human neural tissues, using neural progenitor cells derived from human embryonic stem cells. These 3D neural tissues can be maintained for two months and undergo progressive differentiation. We showed a gradual decreased expression of early neural lineage markers, paralleled by an increase in markers specific for mature neurons, astrocytes and oligodendrocytes. At the end of the two-month culture period the neural tissues not only displayed synapses and immature myelin sheaths around axons, but electrophysiological measurements also showed spontaneous activity. Neurotoxicity testing - comparing non-neurotoxic to known neurotoxic model compounds - showed an expected increase in the marker of astroglial reactivity after exposure to known neurotoxicants methylmercury and trimethyltin. Although further characterization and refinement of the model is required, these results indicate its potential usefulness for in vitro neurotoxicity testing.
Collapse
|
1485
|
Huang WC, Abraham R, Shim BS, Choe H, Page DT. Zika virus infection during the period of maximal brain growth causes microcephaly and corticospinal neuron apoptosis in wild type mice. Sci Rep 2016; 6:34793. [PMID: 27713505 PMCID: PMC5054421 DOI: 10.1038/srep34793] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/16/2016] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) infection in pregnant women has been established as a cause of microcephaly in newborns. Here we test the hypothesis that neurodevelopmental stages when the brain is undergoing rapid growth are particularly vulnerable to the effects of ZIKV infection. We injected ZIKV intracranially into wild type C57BL/6 mice at two different time points: early postnatal development, when the brain is growing at its maximal rate, and at weaning, when the brain has largely reached adult size. Both time points showed widespread immunoreactivity for ZIKV and cleaved caspase 3 (CC3, a marker of apoptosis) throughout the brain. However, in early postnatal ZIKV injected mice, some brain areas and cell types display particularly large increases in apoptosis that we did not observe in older animals. Corticospinal pyramidal neurons, a cell type implicated in human microcephaly associated with ZIKV infection, are an example of one such cell type. Proliferating cells in the ventricular zone stem cell compartment are also depleted. These findings are consistent with the hypothesis that periods of rapid brain growth are especially susceptible to neurodevelopmental effects of ZIKV infection, and establish a valuable model to investigate mechanisms underlying neurodevelopmental effects of ZIKV infection and explore candidate therapeutics.
Collapse
Affiliation(s)
- Wen-Chin Huang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA.,The Doctoral Program in Chemical and Biological Sciences at The Scripps Research Institute, Jupiter, Florida, USA
| | - Rachy Abraham
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, Florida, USA
| | - Byoung-Shik Shim
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, Florida, USA
| | - Hyeryun Choe
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, Florida, USA
| | - Damon T Page
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA.,The Doctoral Program in Chemical and Biological Sciences at The Scripps Research Institute, Jupiter, Florida, USA
| |
Collapse
|
1486
|
Zare Mehrjardi M, Keshavarz E, Poretti A, Hazin AN. Neuroimaging findings of Zika virus infection: a review article. Jpn J Radiol 2016; 34:765-770. [DOI: 10.1007/s11604-016-0588-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/22/2016] [Indexed: 10/20/2022]
|
1487
|
Abstract
CONTEXT -As the number of Zika virus (ZIKV) infections continues to grow, so, too, does the spectrum of recognized clinical disease, in both adult and congenital infections. Defining the tissue pathology associated with the various disease manifestations provides insight into pathogenesis and diagnosis, and potentially future prevention and treatment, of ZIKV infections. OBJECTIVE -To summarize the syndromes and pathology associated with ZIKV infection, the implications of pathologic findings in the pathogenesis of ZIKV disease, and the use of pathology specimens for diagnosis of ZIKV infection. DATA SOURCES -The major sources of information for this review were published articles obtained from PubMed and pathologic findings from cases submitted to the Infectious Diseases Pathology Branch at the Centers for Disease Control and Prevention. CONCLUSIONS -Pathologic findings associated with ZIKV infection are characteristic but not specific. In congenital Zika syndrome, tissue pathology is due to direct viral infection of neural structures, whereas in Guillain-Barré syndrome, pathology is likely due to a postviral, aberrant host-directed immune response. Both fetal and placental pathology specimens are useful for ZIKV diagnosis by molecular and immunohistochemical assays; however, the implications of ZIKV detection in placentas from second- and third-trimester normal live births are unclear, as the potential postnatal effects of late gestational exposure remain to be seen.
Collapse
Affiliation(s)
| | | | - Sherif R Zaki
- From the Infectious Diseases Pathology Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
1488
|
Xu M, Lee EM, Wen Z, Cheng Y, Huang WK, Qian X, Tcw J, Kouznetsova J, Ogden SC, Hammack C, Jacob F, Nguyen HN, Itkin M, Hanna C, Shinn P, Allen C, Michael SG, Simeonov A, Huang W, Christian KM, Goate A, Brennand KJ, Huang R, Xia M, Ming GL, Zheng W, Song H, Tang H. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat Med 2016; 22:1101-1107. [PMID: 27571349 PMCID: PMC5386783 DOI: 10.1038/nm.4184] [Citation(s) in RCA: 529] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022]
Abstract
In response to the current global health emergency posed by the Zika virus (ZIKV) outbreak and its link to microcephaly and other neurological conditions, we performed a drug repurposing screen of ∼6,000 compounds that included approved drugs, clinical trial drug candidates and pharmacologically active compounds; we identified compounds that either inhibit ZIKV infection or suppress infection-induced caspase-3 activity in different neural cells. A pan-caspase inhibitor, emricasan, inhibited ZIKV-induced increases in caspase-3 activity and protected human cortical neural progenitors in both monolayer and three-dimensional organoid cultures. Ten structurally unrelated inhibitors of cyclin-dependent kinases inhibited ZIKV replication. Niclosamide, a category B anthelmintic drug approved by the US Food and Drug Administration, also inhibited ZIKV replication. Finally, combination treatments using one compound from each category (neuroprotective and antiviral) further increased protection of human neural progenitors and astrocytes from ZIKV-induced cell death. Our results demonstrate the efficacy of this screening strategy and identify lead compounds for anti-ZIKV drug development.
Collapse
Affiliation(s)
- Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Emily M Lee
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yichen Cheng
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Wei-Kai Huang
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xuyu Qian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julia Tcw
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jennifer Kouznetsova
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarah C Ogden
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Christy Hammack
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Fadi Jacob
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ha Nam Nguyen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Misha Itkin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Catherine Hanna
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Paul Shinn
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Chase Allen
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Samuel G Michael
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenwei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Kimberly M Christian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristen J Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
1489
|
Simonin Y, Loustalot F, Desmetz C, Foulongne V, Constant O, Fournier-Wirth C, Leon F, Molès JP, Goubaud A, Lemaitre JM, Maquart M, Leparc-Goffart I, Briant L, Nagot N, Van de Perre P, Salinas S. Zika Virus Strains Potentially Display Different Infectious Profiles in Human Neural Cells. EBioMedicine 2016; 12:161-169. [PMID: 27688094 PMCID: PMC5078617 DOI: 10.1016/j.ebiom.2016.09.020] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 01/28/2023] Open
Abstract
The recent Zika virus (ZIKV) epidemic has highlighted the poor knowledge on its physiopathology. Recent studies showed that ZIKV of the Asian lineage, responsible for this international outbreak, causes neuropathology in vitro and in vivo. However, two African lineages exist and the virus is currently found circulating in Africa. The original African strain was also suggested to be neurovirulent but its laboratory usage has been criticized due to its multiple passages. In this study, we compared the French Polynesian (Asian) ZIKV strain to an African strain isolated in Central African Republic and show a difference in infectivity and cellular response between both strains in human neural stem cells and astrocytes. Consistently, this African strain led to a higher infection rate and viral production, as well as stronger cell death and anti-viral response. Our results highlight the need to better characterize the physiopathology and predict neurological impairment associated with African ZIKV.
Collapse
Affiliation(s)
- Yannick Simonin
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Université de Montpellier, Montpellier, France.
| | - Fabien Loustalot
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France
| | | | - Vincent Foulongne
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Orianne Constant
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France
| | - Chantal Fournier-Wirth
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Etablissement Français du Sang, Montpellier, France
| | - Fanny Leon
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Etablissement Français du Sang, Montpellier, France
| | - Jean-Pierre Molès
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France
| | - Aurélien Goubaud
- Institut de Médecine Régénératrice et Biothérapies, INSERM, U1183, Université de Montpellier, CHU Montpellier, Montpellier, France; Plateforme CHU SAFE-IPS, Infrastructure Nationale INGESTEM, Montpellier, France
| | - Jean-Marc Lemaitre
- Institut de Médecine Régénératrice et Biothérapies, INSERM, U1183, Université de Montpellier, CHU Montpellier, Montpellier, France; Plateforme CHU SAFE-IPS, Infrastructure Nationale INGESTEM, Montpellier, France
| | - Marianne Maquart
- Centre National de Référence des Arbovirus, Institut de Recherche Biomédicale des Armées, Marseille, France
| | - Isabelle Leparc-Goffart
- Centre National de Référence des Arbovirus, Institut de Recherche Biomédicale des Armées, Marseille, France
| | - Laurence Briant
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, FRE3689, CNRS-Université de Montpellier, Montpellier, France
| | - Nicolas Nagot
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Philippe Van de Perre
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Sara Salinas
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France.
| |
Collapse
|
1490
|
White MK, Wollebo HS, David Beckham J, Tyler KL, Khalili K. Zika virus: An emergent neuropathological agent. Ann Neurol 2016; 80:479-89. [PMID: 27464346 PMCID: PMC5086418 DOI: 10.1002/ana.24748] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 01/07/2023]
Abstract
The emergence of Zika virus in the Americas has followed a pattern that is familiar from earlier epidemics of other viruses, where a new disease is introduced into a human population and then spreads rapidly with important public health consequences. In the case of Zika virus, an accumulating body of recent evidence implicates the virus in the etiology of serious pathologies of the human nervous system, that is, the occurrence of microcephaly in neonates and Guillain-Barré syndrome in adults. Zika virus is an arbovirus (arthropod-borne virus) and a member of the family Flaviviridae, genus Flavivirus. Zika virions are enveloped and icosahedral, and contain a nonsegmented, single-stranded, positive-sense RNA genome, which encodes 3 structural and 7 nonstructural proteins that are expressed as a single polyprotein that undergoes cleavage. Zika genomic RNA replicates in the cytoplasm of infected host cells. Zika virus was first detected in 1947 in the blood of a febrile monkey in Uganda's Zika Forest and in crushed suspensions of the Aedes mosquito, which is one of the vectors for Zika virus. The virus remained obscure, with a few human cases confined to Africa and Asia. There are two lineages of the Zika virus, African and Asian, with the Asian strain causing outbreaks in Micronesia in 2007 and French Polynesia in 2013-2014. From here, the virus spread to Brazil with the first report of autochthonous Zika transmission in the Americas in March 2015. The rapid advance of the virus in the Americas and its likely association with microcephaly and Guillain-Barré syndrome make Zika an urgent public health concern. Ann Neurol 2016;80:479-489.
Collapse
Affiliation(s)
- Martyn K White
- Department of Neuroscience, Center for Neurovirology, Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Hassen S Wollebo
- Department of Neuroscience, Center for Neurovirology, Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - J David Beckham
- Division of Infectious Diseases, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO
- Department of Neurology, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Kenneth L Tyler
- Division of Infectious Diseases, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO
- Department of Neurology, Anschutz Medical Campus, University of Colorado, Aurora, CO
- Department of Microbiology and Immunology, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA.
| |
Collapse
|
1491
|
Han YG. Sonic hedgehog signaling: A conserved mechanism for the expansion of outer radial glia and intermediate progenitor cells and for the growth and folding of the neocortex. NEUROGENESIS 2016; 3:e1242957. [PMID: 28255571 PMCID: PMC5325666 DOI: 10.1080/23262133.2016.1242957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/12/2016] [Accepted: 09/25/2016] [Indexed: 11/23/2022]
Abstract
The expansion of outer radial glia (oRGs, also called basal RGs) and intermediate progenitor cells (IPCs) has played a key role in the evolutionary expansion and folding of the neocortex, resulting in superior sensorimotor and cognitive abilities. In particular, oRGs, which are critical for both the increased production and lateral dispersion of neurons, are rare in lisencephalic species but vastly expanded in gyrencephalic species. However, the mechanisms that expand oRGs and IPCs are not well understood. We recently identified Sonic hedgehog (Shh) signaling as the first known signaling pathway necessary and sufficient to expand both oRGs and IPCs. Elevated Shh signaling in the embryonic neocortex leads to neocortical expansion and folding with normal cytoarchitecture in otherwise smooth mouse neocortex, whereas the loss of Shh signaling decreases oRGs, IPCs, and neocortical size. We also showed that SHH signaling activity in fetal neocortex is stronger in humans than in mice and that blocking SHH signaling decreases oRGs in human cerebral organoids. Shh signaling may be a conserved mechanism that promotes oRG and IPC expansion, driving neocortical growth and folding in humans and other species. Understanding the mechanisms underlying species-specific differences in Shh signaling activity and how Shh signaling expands oRGs and IPCs will provide insights into the mechanisms of neocortical development and evolution.
Collapse
Affiliation(s)
- Young-Goo Han
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children's Research Hospital , Memphis, TN, USA
| |
Collapse
|
1492
|
Mora-Bermúdez F, Badsha F, Kanton S, Camp JG, Vernot B, Köhler K, Voigt B, Okita K, Maricic T, He Z, Lachmann R, Pääbo S, Treutlein B, Huttner WB. Differences and similarities between human and chimpanzee neural progenitors during cerebral cortex development. eLife 2016; 5:e18683. [PMID: 27669147 PMCID: PMC5110243 DOI: 10.7554/elife.18683] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/22/2016] [Indexed: 12/25/2022] Open
Abstract
Human neocortex expansion likely contributed to the remarkable cognitive abilities of humans. This expansion is thought to primarily reflect differences in proliferation versus differentiation of neural progenitors during cortical development. Here, we have searched for such differences by analysing cerebral organoids from human and chimpanzees using immunohistofluorescence, live imaging, and single-cell transcriptomics. We find that the cytoarchitecture, cell type composition, and neurogenic gene expression programs of humans and chimpanzees are remarkably similar. Notably, however, live imaging of apical progenitor mitosis uncovered a lengthening of prometaphase-metaphase in humans compared to chimpanzees that is specific to proliferating progenitors and not observed in non-neural cells. Consistent with this, the small set of genes more highly expressed in human apical progenitors points to increased proliferative capacity, and the proportion of neurogenic basal progenitors is lower in humans. These subtle differences in cortical progenitors between humans and chimpanzees may have consequences for human neocortex evolution.
Collapse
Affiliation(s)
| | - Farhath Badsha
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sabina Kanton
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - J Gray Camp
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Benjamin Vernot
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Kathrin Köhler
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Birger Voigt
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keisuke Okita
- Department of Reprogramming Science, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Tomislav Maricic
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Zhisong He
- CAS-MPG Partner Institute for Computational Biology, Shanghai, China
| | - Robert Lachmann
- Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Technische Universität Dresden, Dresden, Germany
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Barbara Treutlein
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
1493
|
Di Guardo G, Braga PBB, Peron JPS. Zika virus-associated brain damage: animal models and open issues. Emerg Microbes Infect 2016; 5:e106. [PMID: 27651092 PMCID: PMC5113054 DOI: 10.1038/emi.2016.103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/15/2016] [Accepted: 07/30/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Giovanni Di Guardo
- Faculty of Veterinary Medicine, University of Teramo, Località Piano d'Accio, Teramo 64100, Italy
| | | | - Jean Pierre Schatzmann Peron
- Department of Immunology, Neuroimmune Interactions Laboratory, University of São Paulo, Saint Paul 05508-000, Brazil
| |
Collapse
|
1494
|
Alvarado MG, Schwartz DA. Zika Virus Infection in Pregnancy, Microcephaly, and Maternal and Fetal Health: What We Think, What We Know, and What We Think We Know. Arch Pathol Lab Med 2016; 141:26-32. [PMID: 27636525 DOI: 10.5858/arpa.2016-0382-ra] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT -The global epidemic of Zika virus (ZIKV) infection has emerged as an important public health problem affecting pregnant women and their infants. OBJECTIVES -To review the causal association between ZIKV infection during pregnancy and intrauterine fetal infection, microcephaly, brain damage, congenital malformation syndrome, and experimental laboratory models of fetal infection. Many questions remain regarding the risk factors, pathophysiology, epidemiology, and timing of maternal-fetal transmission and disease. These include mechanisms of fetal brain damage and microcephaly; the role of covariables, such as viral burden, duration of viremia, and host genetics, on vertical transmission; and the clinical and pathologic spectrum of congenital Zika syndrome. Additional questions include defining the potential long-term physical and neurobehavioral outcomes for infected infants, whether maternal or fetal host genetics influence the clinical outcome, and whether ZIKV infection can cause maternal morbidity. Finally, are experimental laboratory and animal models of ZIKV infection helpful in addressing maternal-fetal viral transmission and the development of congenital microcephaly? This communication provides current information and attempts to address some of these important questions. DATA SOURCES -Comprehensive review of published scientific literature. CONCLUSIONS -Recent advances in epidemiology, clinical medicine, pathology, and experimental studies have provided a great amount of new information regarding vertical ZIKV transmission and the mechanisms of congenital microcephaly, brain damage, and congenital Zika syndrome in a relatively short time. However, much work still needs to be performed to more completely understand the maternal and fetal aspects of this new and emerging viral disease.
Collapse
Affiliation(s)
| | - David A Schwartz
- From the Department of Anthropology, Georgia State University, Atlanta (Dr Alvarado); and the Department of Pathology, Medical College of Georgia, Augusta University, Augusta (Dr Schwartz)
| |
Collapse
|
1495
|
Self-Organizing 3D Human Neural Tissue Derived from Induced Pluripotent Stem Cells Recapitulate Alzheimer's Disease Phenotypes. PLoS One 2016; 11:e0161969. [PMID: 27622770 PMCID: PMC5021368 DOI: 10.1371/journal.pone.0161969] [Citation(s) in RCA: 379] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022] Open
Abstract
The dismal success rate of clinical trials for Alzheimer's disease (AD) motivates us to develop model systems of AD pathology that have higher predictive validity. The advent of induced pluripotent stem cells (iPSCs) allows us to model pathology and study disease mechanisms directly in human neural cells from healthy individual as well as AD patients. However, two-dimensional culture systems do not recapitulate the complexity of neural tissue, and phenotypes such as extracellular protein aggregation are difficult to observe. We report brain organoids that use pluripotent stem cells derived from AD patients and recapitulate AD-like pathologies such as amyloid aggregation, hyperphosphorylated tau protein, and endosome abnormalities. These pathologies are observed in an age-dependent manner in organoids derived from multiple familial AD (fAD) patients harboring amyloid precursor protein (APP) duplication or presenilin1 (PSEN1) mutation, compared to controls. The incidence of AD pathology was consistent amongst several fAD lines, which carried different mutations. Although these are complex assemblies of neural tissue, they are also highly amenable to experimental manipulation. We find that treatment of patient-derived organoids with β- and γ-secretase inhibitors significantly reduces amyloid and tau pathology. Moreover, these results show the potential of this model system to greatly increase the translatability of pre-clinical drug discovery in AD.
Collapse
|
1496
|
Abbink P, Larocca RA, De La Barrera RA, Bricault CA, Moseley ET, Boyd M, Kirilova M, Li Z, Ng'ang'a D, Nanayakkara O, Nityanandam R, Mercado NB, Borducchi EN, Agarwal A, Brinkman AL, Cabral C, Chandrashekar A, Giglio PB, Jetton D, Jimenez J, Lee BC, Mojta S, Molloy K, Shetty M, Neubauer GH, Stephenson KE, Peron JPS, Zanotto PMDA, Misamore J, Finneyfrock B, Lewis MG, Alter G, Modjarrad K, Jarman RG, Eckels KH, Michael NL, Thomas SJ, Barouch DH. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys. Science 2016; 353:1129-32. [PMID: 27492477 PMCID: PMC5237380 DOI: 10.1126/science.aah6157] [Citation(s) in RCA: 403] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 07/28/2016] [Indexed: 01/10/2023]
Abstract
Zika virus (ZIKV) is responsible for a major ongoing epidemic in the Americas and has been causally associated with fetal microcephaly. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Here we demonstrate that three different vaccine platforms protect against ZIKV challenge in rhesus monkeys. A purified inactivated virus vaccine induced ZIKV-specific neutralizing antibodies and completely protected monkeys against ZIKV strains from both Brazil and Puerto Rico. Purified immunoglobulin from vaccinated monkeys also conferred passive protection in adoptive transfer studies. A plasmid DNA vaccine and a single-shot recombinant rhesus adenovirus serotype 52 vector vaccine, both expressing ZIKV premembrane and envelope, also elicited neutralizing antibodies and completely protected monkeys against ZIKV challenge. These data support the rapid clinical development of ZIKV vaccines for humans.
Collapse
Affiliation(s)
- Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - Christine A Bricault
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Edward T Moseley
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michael Boyd
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Marinela Kirilova
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zhenfeng Li
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Ng'ang'a
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ovini Nanayakkara
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ramya Nityanandam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Noe B Mercado
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Arshi Agarwal
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Amanda L Brinkman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Crystal Cabral
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Patricia B Giglio
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Jetton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jessica Jimenez
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Benjamin C Lee
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shanell Mojta
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Katherine Molloy
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mayuri Shetty
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - George H Neubauer
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Kayvon Modjarrad
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA. Henry M. Jackson Foundation, Bethesda, MD 20817, USA
| | - Richard G Jarman
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Kenneth H Eckels
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Nelson L Michael
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Stephen J Thomas
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
1497
|
Onorati M, Li Z, Liu F, Sousa AMM, Nakagawa N, Li M, Dell'Anno MT, Gulden FO, Pochareddy S, Tebbenkamp ATN, Han W, Pletikos M, Gao T, Zhu Y, Bichsel C, Varela L, Szigeti-Buck K, Lisgo S, Zhang Y, Testen A, Gao XB, Mlakar J, Popovic M, Flamand M, Strittmatter SM, Kaczmarek LK, Anton ES, Horvath TL, Lindenbach BD, Sestan N. Zika Virus Disrupts Phospho-TBK1 Localization and Mitosis in Human Neuroepithelial Stem Cells and Radial Glia. Cell Rep 2016; 16:2576-2592. [PMID: 27568284 PMCID: PMC5135012 DOI: 10.1016/j.celrep.2016.08.038] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/27/2016] [Accepted: 08/12/2016] [Indexed: 02/08/2023] Open
Abstract
The mechanisms underlying Zika virus (ZIKV)-related microcephaly and other neurodevelopment defects remain poorly understood. Here, we describe the derivation and characterization, including single-cell RNA-seq, of neocortical and spinal cord neuroepithelial stem (NES) cells to model early human neurodevelopment and ZIKV-related neuropathogenesis. By analyzing human NES cells, organotypic fetal brain slices, and a ZIKV-infected micrencephalic brain, we show that ZIKV infects both neocortical and spinal NES cells as well as their fetal homolog, radial glial cells (RGCs), causing disrupted mitoses, supernumerary centrosomes, structural disorganization, and cell death. ZIKV infection of NES cells and RGCs causes centrosomal depletion and mitochondrial sequestration of phospho-TBK1 during mitosis. We also found that nucleoside analogs inhibit ZIKV replication in NES cells, protecting them from ZIKV-induced pTBK1 relocalization and cell death. We established a model system of human neural stem cells to reveal cellular and molecular mechanisms underlying neurodevelopmental defects associated with ZIKV infection and its potential treatment.
Collapse
Affiliation(s)
- Marco Onorati
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Zhen Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Fuchen Liu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - André M M Sousa
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Naoki Nakagawa
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Mingfeng Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Maria Teresa Dell'Anno
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Forrest O Gulden
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sirisha Pochareddy
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrew T N Tebbenkamp
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Wenqi Han
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mihovil Pletikos
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tianliuyun Gao
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ying Zhu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Candace Bichsel
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Luis Varela
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Klara Szigeti-Buck
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Steven Lisgo
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE13BZ, UK
| | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anze Testen
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Xiao-Bing Gao
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jernej Mlakar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Mara Popovic
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Marie Flamand
- Department of Virology, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Tamas L Horvath
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; Yale Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Brett D Lindenbach
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Genetics and Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
1498
|
Tang BL. Zika virus as a causative agent for primary microencephaly: the evidence so far. Arch Microbiol 2016; 198:595-601. [PMID: 27412681 DOI: 10.1007/s00203-016-1268-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 06/30/2016] [Accepted: 07/08/2016] [Indexed: 02/06/2023]
Abstract
Zika virus (ZIKV) infection has been associated with congenital microcephaly and peripheral neuropathy. The ongoing epidemic has triggered swift responses in the scientific community, and a number of recent reports have now confirmed a causal relationship between ZIKV infection and birth defect. In particular, ZIKV has been shown to be capable of compromising and crossing the placental barrier and infect the developing fetal brain, resulting in the demise and functional impairment of neuroprogenitor cells critical for fetal cortex development. Here, the evidence for ZIKV as a teratogenic agent that causes microcephaly is reviewed, and its association with other disorders is discussed.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD7, 8 Medical Drive, Singapore, 117597, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore.
| |
Collapse
|
1499
|
|
1500
|
Krencik R, van Asperen JV, Ullian EM. Human astrocytes are distinct contributors to the complexity of synaptic function. Brain Res Bull 2016; 129:66-73. [PMID: 27570101 DOI: 10.1016/j.brainresbull.2016.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/07/2016] [Accepted: 08/22/2016] [Indexed: 01/03/2023]
Abstract
Cellular components of synaptic circuits have been adjusted for increased human brain size, neural cell density, energy consumption and developmental duration. How does the human brain make these accommodations? There is evidence that astrocytes are one of the most divergent neural cell types in primate brain evolution and it is now becoming clear that they have critical roles in controlling synaptic development, function and plasticity. Yet, we still do not know how the precise developmental appearance of these cells and subsequent astrocyte-derived signals modulate diverse neuronal circuit subtypes. Here, we discuss what is currently known about the influence of glial factors on synaptic maturation and focus on unique features of human astrocytes including their potential roles in regenerative and translational medicine. Human astrocyte distinctiveness may be a major contributor to high level neuronal processing of the human brain and act in novel ways during various neuropathies ranging from autism spectrum disorders, viral infection, injury and neurodegenerative conditions.
Collapse
Affiliation(s)
- Robert Krencik
- Departments of Ophthalmology and Physiology, Neuroscience Program, University of California San Francisco, United States.
| | - Jessy V van Asperen
- Departments of Ophthalmology and Physiology, Neuroscience Program, University of California San Francisco, United States
| | - Erik M Ullian
- Departments of Ophthalmology and Physiology, Neuroscience Program, University of California San Francisco, United States
| |
Collapse
|