1
|
Ryu JR, Ko K, Sun W. Polarization of organoids by bioengineered symmetry breaking. IBRO Neurosci Rep 2024; 17:22-31. [PMID: 38881849 PMCID: PMC11176950 DOI: 10.1016/j.ibneur.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/08/2024] [Indexed: 06/18/2024] Open
Abstract
Symmetry breaking leading to axis formation and spatial patterning is crucial for achieving more accurate recapitulation of human development in organoids. While these processes can occur spontaneously by self-organizing capabilities of pluripotent stem cells, they can often result in variation in structure and composition of cell types within organoids. To address this limitation, bioengineering techniques that utilize geometric, topological and stiffness factors are increasingly employed to enhance control and consistency. Here, we review how spontaneous manners and engineering tools such as micropattern, microfluidics, biomaterials, etc. can facilitate the process of symmetry breaking leading to germ layer patterning and the formation of anteroposterior and dorsoventral axes in blastoids, gastruloids, neuruloids and neural organoids. Furthermore, brain assembloids, which are composed of multiple brain regions through fusion processes are discussed. The overview of organoid polarization in terms of patterning tools can offer valuable insights for enhancing the physiological relevance of organoid system.
Collapse
Affiliation(s)
- Jae Ryun Ryu
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kahee Ko
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
2
|
Hamazaki N, Yang W, Kubo CA, Qiu C, Martin BK, Garge RK, Regalado SG, Nichols EK, Pendyala S, Bradley N, Fowler DM, Lee C, Daza RM, Srivatsan S, Shendure J. Retinoic acid induces human gastruloids with posterior embryo-like structures. Nat Cell Biol 2024; 26:1790-1803. [PMID: 39164488 PMCID: PMC11469962 DOI: 10.1038/s41556-024-01487-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Gastruloids are a powerful in vitro model of early human development. However, although elongated and composed of all three germ layers, human gastruloids do not morphologically resemble post-implantation human embryos. Here we show that an early pulse of retinoic acid (RA), together with later Matrigel, robustly induces human gastruloids with posterior embryo-like morphological structures, including a neural tube flanked by segmented somites and diverse cell types, including neural crest, neural progenitors, renal progenitors and myocytes. Through in silico staging based on single-cell RNA sequencing, we find that human RA-gastruloids progress further than other human or mouse embryo models, aligning to E9.5 mouse and CS11 cynomolgus monkey embryos. We leverage chemical and genetic perturbations of RA-gastruloids to confirm that WNT and BMP signalling regulate somite formation and neural tube length in the human context, while transcription factors TBX6 and PAX3 underpin presomitic mesoderm and neural crest, respectively. Looking forward, RA-gastruloids are a robust, scalable model for decoding early human embryogenesis.
Collapse
Affiliation(s)
- Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
| | - Wei Yang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Connor A Kubo
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Riddhiman K Garge
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Samuel G Regalado
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Eva K Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sriram Pendyala
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Nicholas Bradley
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Choli Lee
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| |
Collapse
|
3
|
Gu L, Cai H, Chen L, Gu M, Tchieu J, Guo F. Functional Neural Networks in Human Brain Organoids. BME FRONTIERS 2024; 5:0065. [PMID: 39314749 PMCID: PMC11418062 DOI: 10.34133/bmef.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/12/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Human brain organoids are 3-dimensional brain-like tissues derived from human pluripotent stem cells and hold promising potential for modeling neurological, psychiatric, and developmental disorders. While the molecular and cellular aspects of human brain organoids have been intensively studied, their functional properties such as organoid neural networks (ONNs) are largely understudied. Here, we summarize recent research advances in understanding, characterization, and application of functional ONNs in human brain organoids. We first discuss the formation of ONNs and follow up with characterization strategies including microelectrode array (MEA) technology and calcium imaging. Moreover, we highlight recent studies utilizing ONNs to investigate neurological diseases such as Rett syndrome and Alzheimer's disease. Finally, we provide our perspectives on the future challenges and opportunities for using ONNs in basic research and translational applications.
Collapse
Affiliation(s)
- Longjun Gu
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Lei Chen
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Jason Tchieu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| |
Collapse
|
4
|
Bertaccini GA, Casanellas I, Evans EL, Nourse JL, Dickinson GD, Liu G, Seal S, Ly AT, Holt JR, Wijerathne TD, Yan S, Hui EE, Lacroix JJ, Panicker MM, Upadhyayula S, Parker I, Pathak MM. Visualizing PIEZO1 Localization and Activity in hiPSC-Derived Single Cells and Organoids with HaloTag Technology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573117. [PMID: 38187535 PMCID: PMC10769387 DOI: 10.1101/2023.12.22.573117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
PIEZO1 is critical to numerous physiological processes, transducing diverse mechanical stimuli into electrical and chemical signals. Recent studies underscore the importance of visualizing endogenous PIEZO1 activity and localization to understand its functional roles. To enable physiologically and clinically relevant studies on human PIEZO1, we genetically engineered human induced pluripotent stem cells (hiPSCs) to express a HaloTag fused to endogenous PIEZO1. Combined with advanced imaging, our chemogenetic platform allows precise visualization of PIEZO1 localization dynamics in various cell types. Furthermore, the PIEZO1-HaloTag hiPSC technology facilitates the non-invasive monitoring of channel activity across diverse cell types using Ca2+-sensitive HaloTag ligands, achieving temporal resolution approaching that of patch clamp electrophysiology. Finally, we used lightsheet imaging of hiPSC-derived neural organoids to achieve molecular scale imaging of PIEZO1 in three-dimensional tissue organoids. Our advances offer a novel platform for studying PIEZO1 mechanotransduction in human cells and tissues, with potential for elucidating disease mechanisms and targeted therapeutic development.
Collapse
Affiliation(s)
- Gabriella A Bertaccini
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Ignasi Casanellas
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Elizabeth L Evans
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jamison L Nourse
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - George D Dickinson
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Gaoxiang Liu
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Sayan Seal
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Alan T Ly
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Jesse R Holt
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Tharaka D Wijerathne
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Shijun Yan
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Elliot E Hui
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - Jerome J Lacroix
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Mitradas M Panicker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Srigokul Upadhyayula
- Advanced Bioimaging Center, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ian Parker
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Medha M Pathak
- Department of Physiology and Biophysics, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| |
Collapse
|
5
|
Lisowski P, Lickfett S, Rybak-Wolf A, Menacho C, Le S, Pentimalli TM, Notopoulou S, Dykstra W, Oehler D, López-Calcerrada S, Mlody B, Otto M, Wu H, Richter Y, Roth P, Anand R, Kulka LAM, Meierhofer D, Glazar P, Legnini I, Telugu NS, Hahn T, Neuendorf N, Miller DC, Böddrich A, Polzin A, Mayatepek E, Diecke S, Olzscha H, Kirstein J, Ugalde C, Petrakis S, Cambridge S, Rajewsky N, Kühn R, Wanker EE, Priller J, Metzger JJ, Prigione A. Mutant huntingtin impairs neurodevelopment in human brain organoids through CHCHD2-mediated neurometabolic failure. Nat Commun 2024; 15:7027. [PMID: 39174523 PMCID: PMC11341898 DOI: 10.1038/s41467-024-51216-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Abstract
Expansion of the glutamine tract (poly-Q) in the protein huntingtin (HTT) causes the neurodegenerative disorder Huntington's disease (HD). Emerging evidence suggests that mutant HTT (mHTT) disrupts brain development. To gain mechanistic insights into the neurodevelopmental impact of human mHTT, we engineered male induced pluripotent stem cells to introduce a biallelic or monoallelic mutant 70Q expansion or to remove the poly-Q tract of HTT. The introduction of a 70Q mutation caused aberrant development of cerebral organoids with loss of neural progenitor organization. The early neurodevelopmental signature of mHTT highlighted the dysregulation of the protein coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2), a transcription factor involved in mitochondrial integrated stress response. CHCHD2 repression was associated with abnormal mitochondrial morpho-dynamics that was reverted upon overexpression of CHCHD2. Removing the poly-Q tract from HTT normalized CHCHD2 levels and corrected key mitochondrial defects. Hence, mHTT-mediated disruption of human neurodevelopment is paralleled by aberrant neurometabolic programming mediated by dysregulation of CHCHD2, which could then serve as an early interventional target for HD.
Collapse
Affiliation(s)
- Pawel Lisowski
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin, Berlin, Germany
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec n/Warsaw, Poland
| | - Selene Lickfett
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Institute of Anatomy II, Heinrich-Heine-University, Düsseldorf, Germany
| | - Agnieszka Rybak-Wolf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Organoid Platform, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Carmen Menacho
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Stephanie Le
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Tancredi Massimo Pentimalli
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Charité - Universitätsmedizin, Berlin, Germany
| | - Sofia Notopoulou
- Institute of Applied Biosciences (INAB), Centre For Research and Technology Hellas (CERTH), Thessaloniki, Greece
| | - Werner Dykstra
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht, The Netherlands
| | - Daniel Oehler
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | | | - Barbara Mlody
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Centogene, Rostock, Germany
| | - Maximilian Otto
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Haijia Wu
- Institute of Molecular Medicine, Medical School, Hamburg, Germany
| | | | - Philipp Roth
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Linda A M Kulka
- Institute of Physiological Chemistry, Martin-Luther-University, Halle-Wittenberg, Germany
| | - David Meierhofer
- Quantitative RNA Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Petar Glazar
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Quantitative RNA Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Ivano Legnini
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Human Technopole, Milan, Italy
| | - Narasimha Swamy Telugu
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tobias Hahn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Nancy Neuendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Duncan C Miller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Annett Böddrich
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Sebastian Diecke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Heidi Olzscha
- Institute of Molecular Medicine, Medical School, Hamburg, Germany
- Institute of Physiological Chemistry, Martin-Luther-University, Halle-Wittenberg, Germany
| | - Janine Kirstein
- Cell Biology, University of Bremen, Bremen, Germany
- Leibniz Institute on Aging - Fritz-Lipmann Institute, Jena, Germany
| | - Cristina Ugalde
- Instituto de Investigación Hospital 12 de Octubre (i + 12), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Spyros Petrakis
- Institute of Applied Biosciences (INAB), Centre For Research and Technology Hellas (CERTH), Thessaloniki, Greece
| | - Sidney Cambridge
- Institute of Anatomy II, Heinrich-Heine-University, Düsseldorf, Germany
- Dr. Senckenberg Anatomy, Anatomy II, Goethe-University, Frankfurt, Germany
| | - Nikolaus Rajewsky
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Laboratory for Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
- National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Ralf Kühn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Erich E Wanker
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy; School of Medicine and Health, Technical University of Munich and German Center for Mental Health (DZPG), Munich, Germany
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, UK
| | - Jakob J Metzger
- Quantitative Stem Cell Biology, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| | - Alessandro Prigione
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
6
|
Galimberti M, Nucera MR, Bocchi VD, Conforti P, Vezzoli E, Cereda M, Maffezzini C, Iennaco R, Scolz A, Falqui A, Cordiglieri C, Cremona M, Espuny-Camacho I, Faedo A, Felsenfeld DP, Vogt TF, Ranzani V, Zuccato C, Besusso D, Cattaneo E. Huntington's disease cellular phenotypes are rescued non-cell autonomously by healthy cells in mosaic telencephalic organoids. Nat Commun 2024; 15:6534. [PMID: 39095390 PMCID: PMC11297310 DOI: 10.1038/s41467-024-50877-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Huntington's disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.
Collapse
Affiliation(s)
- Maura Galimberti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Maria R Nucera
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Stem Cell Biology Department; Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Vittoria D Bocchi
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Paola Conforti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elena Vezzoli
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- ALEMBIC Advanced Light and Electron Microscopy BioImaging Center, San Raffaele Scientific Institute, DIBIT 1, Via Olgettina 58, 20132, Milan, Italy
| | - Matteo Cereda
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Camilla Maffezzini
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Raffaele Iennaco
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Scolz
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Falqui
- Department of Physics "Aldo Pontremoli", University of Milan, Via Celoria 16, 20133, Milan, Italy
| | - Chiara Cordiglieri
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Martina Cremona
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Swiss Stem Cell Foundation, Via Petrini 2, 6900, Lugano, Switzerland
| | - Ira Espuny-Camacho
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- GIGA-Neuroscience, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, 4000, Liège, Belgium
| | - Andrea Faedo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Axxam, OpenZone, Via Meucci 3, 20091, Bresso, Milan, Italy
| | | | | | - Valeria Ranzani
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Chiara Zuccato
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Dario Besusso
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elena Cattaneo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy.
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy.
| |
Collapse
|
7
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
8
|
Wu J, Fu J. Toward developing human organs via embryo models and chimeras. Cell 2024; 187:3194-3219. [PMID: 38906095 PMCID: PMC11239105 DOI: 10.1016/j.cell.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 02/02/2024] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Developing functional organs from stem cells remains a challenging goal in regenerative medicine. Existing methodologies, such as tissue engineering, bioprinting, and organoids, only offer partial solutions. This perspective focuses on two promising approaches emerging for engineering human organs from stem cells: stem cell-based embryo models and interspecies organogenesis. Both approaches exploit the premise of guiding stem cells to mimic natural development. We begin by summarizing what is known about early human development as a blueprint for recapitulating organogenesis in both embryo models and interspecies chimeras. The latest advances in both fields are discussed before highlighting the technological and knowledge gaps to be addressed before the goal of developing human organs could be achieved using the two approaches. We conclude by discussing challenges facing embryo modeling and interspecies organogenesis and outlining future prospects for advancing both fields toward the generation of human tissues and organs for basic research and translational applications.
Collapse
Affiliation(s)
- Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
9
|
Lundin BF, Knight GT, Fedorchak NJ, Krucki K, Iyer N, Maher JE, Izban NR, Roberts A, Cicero MR, Robinson JF, Iskandar BJ, Willett R, Ashton RS. RosetteArray ® Platform for Quantitative High-Throughput Screening of Human Neurodevelopmental Risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587605. [PMID: 38798648 PMCID: PMC11118315 DOI: 10.1101/2024.04.01.587605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Neural organoids have revolutionized how human neurodevelopmental disorders (NDDs) are studied. Yet, their utility for screening complex NDD etiologies and in drug discovery is limited by a lack of scalable and quantifiable derivation formats. Here, we describe the RosetteArray® platform's ability to be used as an off-the-shelf, 96-well plate assay that standardizes incipient forebrain and spinal cord organoid morphogenesis as micropatterned, 3-D, singularly polarized neural rosette tissues (>9000 per plate). RosetteArrays are seeded from cryopreserved human pluripotent stem cells, cultured over 6-8 days, and immunostained images can be quantified using artificial intelligence-based software. We demonstrate the platform's suitability for screening developmental neurotoxicity and genetic and environmental factors known to cause neural tube defect risk. Given the presence of rosette morphogenesis perturbation in neural organoid models of NDDs and neurodegenerative disorders, the RosetteArray platform could enable quantitative high-throughput screening (qHTS) of human neurodevelopmental risk across regulatory and precision medicine applications.
Collapse
Affiliation(s)
- Brady F. Lundin
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Medical Scientist Training Program, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705 USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Gavin T. Knight
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Neurosetta LLC, 330 N. Orchard Street Rm 4140A, Madison, WI 53715 USA
| | | | - Kevin Krucki
- Neurosetta LLC, 330 N. Orchard Street Rm 4140A, Madison, WI 53715 USA
| | - Nisha Iyer
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jack E. Maher
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Nicholas R. Izban
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Abilene Roberts
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Madeline R. Cicero
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Joshua F. Robinson
- Center of Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bermans J. Iskandar
- Department of Neurological Surgery, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI 53705, USA
| | - Rebecca Willett
- Neurosetta LLC, 330 N. Orchard Street Rm 4140A, Madison, WI 53715 USA
- Departments of Statistics and Computer Science, University of Chicago, Chicago, IL 60637, USA
| | - Randolph S. Ashton
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Neurosetta LLC, 330 N. Orchard Street Rm 4140A, Madison, WI 53715 USA
| |
Collapse
|
10
|
Tamada A, Muguruma K. Recapitulation and investigation of human brain development with neural organoids. IBRO Neurosci Rep 2024; 16:106-117. [PMID: 39007085 PMCID: PMC11240300 DOI: 10.1016/j.ibneur.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Organoids are 3D cultured tissues derived from stem cells that resemble the structure of living organs. Based on the accumulated knowledge of neural development, neural organoids that recapitulate neural tissue have been created by inducing self-organized neural differentiation of stem cells. Neural organoid techniques have been applied to human pluripotent stem cells to differentiate 3D human neural tissues in culture. Various methods have been developed to generate neural tissues of different regions. Currently, neural organoid technology has several significant limitations, which are being overcome in an attempt to create neural organoids that more faithfully recapitulate the living brain. The rapidly advancing neural organoid technology enables the use of living human neural tissue as research material and contributes to our understanding of the development, structure and function of the human nervous system, and is expected to be used to overcome neurological diseases and for regenerative medicine.
Collapse
Affiliation(s)
- Atsushi Tamada
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Keiko Muguruma
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
11
|
Wu S, Hong Y, Chu C, Gan Y, Li X, Tao M, Wang D, Hu H, Zheng Z, Zhu Q, Han X, Zhu W, Xu M, Dong Y, Liu Y, Guo X. Construction of human 3D striato-nigral assembloids to recapitulate medium spiny neuronal projection defects in Huntington's disease. Proc Natl Acad Sci U S A 2024; 121:e2316176121. [PMID: 38771878 PMCID: PMC11145230 DOI: 10.1073/pnas.2316176121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/22/2024] [Indexed: 05/23/2024] Open
Abstract
The striato-nigral (Str-SN) circuit is composed of medium spiny neuronal projections that are mainly sent from the striatum to the midbrain substantial nigra (SN), which is essential for regulating motor behaviors. Dysfunction of the Str-SN circuitry may cause a series of motor disabilities that are associated with neurodegenerative disorders, such as Huntington's disease (HD). Although the etiology of HD is known as abnormally expanded CAG repeats of the huntingtin gene, treatment of HD remains tremendously challenging. One possible reason is the lack of effective HD model that resembles Str-SN circuitry deficits for pharmacological studies. Here, we first differentiated striatum-like organoids from human pluripotent stem cells (hPSCs), containing functional medium spiny neurons (MSNs). We then generated 3D Str-SN assembloids by assembling striatum-like organoids with midbrain SN-like organoids. With AAV-hSYN-GFP-mediated viral tracing, extensive MSN projections from the striatum to the SN are established, which formed synaptic connection with GABAergic neurons in SN organoids and showed the optically evoked inhibitory postsynaptic currents and electronic field potentials by labeling the striatum-like organoids with optogenetic virus. Furthermore, these Str-SN assembloids exhibited enhanced calcium activity compared to that of individual striatal organoids. Importantly, we further demonstrated the reciprocal projection defects in HD iPSC-derived assembloids, which could be ameliorated by treatment of brain-derived neurotrophic factor. Taken together, these findings suggest that Str-SN assembloids could be used for identifying MSN projection defects and could be applied as potential drug test platforms for HD.
Collapse
Affiliation(s)
- Shanshan Wu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yuan Hong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Chu Chu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yixia Gan
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, China
| | - Xinrui Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Mengdan Tao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- School of Biological Science and Medical Engineering Southeast University, Sipailou, Nanjing210096, China
| | - Da Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Hao Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Zhilong Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing211166, China
| | - Qian Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Xiao Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Wanying Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Min Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
- School of Biological Science and Medical Engineering Southeast University, Sipailou, Nanjing210096, China
| | - Xing Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing211166, China
- Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu226001, China
| |
Collapse
|
12
|
Rossant J. Why study human embryo development? Dev Biol 2024; 509:43-50. [PMID: 38325560 DOI: 10.1016/j.ydbio.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/31/2023] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Understanding the processes and mechanisms underlying early human embryo development has become an increasingly active and important area of research. It has potential for insights into important clinical issues such as early pregnancy loss, origins of congenital anomalies and developmental origins of adult disease, as well as fundamental insights into human biology. Improved culture systems for preimplantation embryos, combined with the new tools of single cell genomics and live imaging, are providing new insights into the similarities and differences between human and mouse development. However, access to human embryo material is still restricted and extended culture of early embryos has regulatory and ethical concerns. Stem cell-derived models of different phases of human development can potentially overcome these limitations and provide a scalable source of material to explore the early postimplantation stages of human development. To date, such models are clearly incomplete replicas of normal development but future technological improvements can be envisaged. The ethical and regulatory environment for such studies remains to be fully resolved.
Collapse
Affiliation(s)
- Janet Rossant
- The Gairdner Foundation and the Hospital for Sick Children, University of Toronto, MaRS Centre, Heritage Building, 101 College Street, Suite 335, Toronto, Ontario, M5G 1L7, Canada.
| |
Collapse
|
13
|
Irfan S, Etekochay MO, Atanasov AG, Prasad VP, Kandimalla R, Mofatteh M, V P, Emran TB. Human olfactory neurosphere-derived cells: A unified tool for neurological disease modelling and neurotherapeutic applications. Int J Surg 2024; 110:01279778-990000000-01366. [PMID: 38652180 PMCID: PMC11486950 DOI: 10.1097/js9.0000000000001460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
As one of the leading causes of global mortality and morbidity, various neurological diseases cause social and economic burdens. Despite significant advances in the treatment of neurological diseases, establishing a proper disease model, especially for degenerative and infectious diseases, remains a major challenging issue. For long, mice were the model of choice but suffered from serious drawbacks of differences in anatomical and functional aspects of the nervous system. Furthermore, the collection of post-mortem brain tissues limits their usage in cultured cell lines. Overcoming such limitations has prompted the usage of stem cells derived from the peripheral nervous system, such as the cells of the olfactory mucosa as a preferred choice. These cells can be easily cultured in vitro and retain the receptors of neuronal cells life-long. Such cells have various advantages over embryonic or induced stem cells, including homology, and ease of culture and can be conveniently obtained from diseased individuals through either biopsies or exfoliation. They have continuously helped in understanding the genetic and developmental mechanisms of degenerative diseases like Alzheimer's and Parkinson's disease. Moreover, the mode of infection of various viruses that can lead to post-viral olfactory dysfunction, such as the Zika virus can be monitored through these cells in vitro and their therapeutic development can be fastened.
Collapse
Affiliation(s)
- Saad Irfan
- Animal Science Department, Faculty of Animal and Agriculture Sciences, Universitas Diponegoro, Semarang, Indonesia
| | | | - Atanas G. Atanasov
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria
| | - Vishnu P. Prasad
- Rajiv Gandhi University of Health Sciences, Jayanagar, Bengaluru, Karnataka
| | - Ramesh Kandimalla
- CSIR-Indian Institute of Chemical Technology Uppal Road, Tarnaka, Hyderabad, Telangana State
- Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana, India
| | - Mohammad Mofatteh
- School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Priyanka V
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda, Punjab, India
| | - Talha B. Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
14
|
Alasaadi DN, Alvizi L, Hartmann J, Stillman N, Moghe P, Hiiragi T, Mayor R. Competence for neural crest induction is controlled by hydrostatic pressure through Yap. Nat Cell Biol 2024; 26:530-541. [PMID: 38499770 PMCID: PMC11021196 DOI: 10.1038/s41556-024-01378-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/15/2024] [Indexed: 03/20/2024]
Abstract
Embryonic induction is a key mechanism in development that corresponds to an interaction between a signalling and a responding tissue, causing a change in the direction of differentiation by the responding tissue. Considerable progress has been achieved in identifying inductive signals, yet how tissues control their responsiveness to these signals, known as competence, remains poorly understood. While the role of molecular signals in competence has been studied, how tissue mechanics influence competence remains unexplored. Here we investigate the role of hydrostatic pressure in controlling competence in neural crest cells, an embryonic cell population. We show that neural crest competence decreases concomitantly with an increase in the hydrostatic pressure of the blastocoel, an embryonic cavity in contact with the prospective neural crest. By manipulating hydrostatic pressure in vivo, we show that this increase leads to the inhibition of Yap signalling and impairs Wnt activation in the responding tissue, which would be required for neural crest induction. We further show that hydrostatic pressure controls neural crest induction in amphibian and mouse embryos and in human cells, suggesting a conserved mechanism across vertebrates. Our work sets out how tissue mechanics can interplay with signalling pathways to regulate embryonic competence.
Collapse
Affiliation(s)
- Delan N Alasaadi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Lucas Alvizi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Jonas Hartmann
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Namid Stillman
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Prachiti Moghe
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Collaboration for joint PhD degree between the European Molecular Biology Laboratory (EMBL) and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Takashi Hiiragi
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK.
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.
| |
Collapse
|
15
|
Xue X, Kim YS, Ponce-Arias AI, O'Laughlin R, Yan RZ, Kobayashi N, Tshuva RY, Tsai YH, Sun S, Zheng Y, Liu Y, Wong FCK, Surani A, Spence JR, Song H, Ming GL, Reiner O, Fu J. A patterned human neural tube model using microfluidic gradients. Nature 2024; 628:391-399. [PMID: 38408487 PMCID: PMC11006583 DOI: 10.1038/s41586-024-07204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/16/2024] [Indexed: 02/28/2024]
Abstract
The human nervous system is a highly complex but organized organ. The foundation of its complexity and organization is laid down during regional patterning of the neural tube, the embryonic precursor to the human nervous system. Historically, studies of neural tube patterning have relied on animal models to uncover underlying principles. Recently, models of neurodevelopment based on human pluripotent stem cells, including neural organoids1-5 and bioengineered neural tube development models6-10, have emerged. However, such models fail to recapitulate neural patterning along both rostral-caudal and dorsal-ventral axes in a three-dimensional tubular geometry, a hallmark of neural tube development. Here we report a human pluripotent stem cell-based, microfluidic neural tube-like structure, the development of which recapitulates several crucial aspects of neural patterning in brain and spinal cord regions and along rostral-caudal and dorsal-ventral axes. This structure was utilized for studying neuronal lineage development, which revealed pre-patterning of axial identities of neural crest progenitors and functional roles of neuromesodermal progenitors and the caudal gene CDX2 in spinal cord and trunk neural crest development. We further developed dorsal-ventral patterned microfluidic forebrain-like structures with spatially segregated dorsal and ventral regions and layered apicobasal cellular organizations that mimic development of the human forebrain pallium and subpallium, respectively. Together, these microfluidics-based neurodevelopment models provide three-dimensional lumenal tissue architectures with in vivo-like spatiotemporal cell differentiation and organization, which will facilitate the study of human neurodevelopment and disease.
Collapse
Affiliation(s)
- Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yung Su Kim
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Alfredo-Isaac Ponce-Arias
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Richard O'Laughlin
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Robin Zhexuan Yan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Norio Kobayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Rami Yair Tshuva
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yue Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Frederick C K Wong
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Azim Surani
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Stokar-Regenscheit N, Bell L, Berridge B, Rudmann D, Tagle D, Hargrove-Grimes P, Schaudien D, Hahn K, Kühnlenz J, Ashton RS, Tseng M, Reichelt M, Laing ST, Kiyota T, Chamanza R, Sura R, Tomlinson L. Complex In Vitro Model Characterization for Context of Use in Toxicologic Pathology: Use Cases by Collaborative Teams of Biologists, Bioengineers, and Pathologists. Toxicol Pathol 2024; 52:123-137. [PMID: 38888280 DOI: 10.1177/01926233241253811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Complex in vitro models (CIVMs) offer the potential to increase the clinical relevance of preclinical efficacy and toxicity assessments and reduce the reliance on animals in drug development. The European Society of Toxicologic Pathology (ESTP) and Society for Toxicologic Pathology (STP) are collaborating to highlight the role of pathologists in the development and use of CIVM. Pathologists are trained in comparative animal medicine which enhances their understanding of mechanisms of human and animal diseases, thus allowing them to bridge between animal models and humans. This skill set is important for CIVM development, validation, and data interpretation. Ideally, diverse teams of scientists, including engineers, biologists, pathologists, and others, should collaboratively develop and characterize novel CIVM, and collectively assess their precise use cases (context of use). Implementing a morphological CIVM evaluation should be essential in this process. This requires robust histological technique workflows, image analysis techniques, and needs correlation with translational biomarkers. In this review, we demonstrate how such tissue technologies and analytics support the development and use of CIVM for drug efficacy and safety evaluations. We encourage the scientific community to explore similar options for their projects and to engage with health authorities on the use of CIVM in benefit-risk assessment.
Collapse
Affiliation(s)
- Nadine Stokar-Regenscheit
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Luisa Bell
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | - Danilo Tagle
- National Center for Advancing Translational Sciences/National Institutes of Health, Bethesda, Maryland, USA
| | - Passley Hargrove-Grimes
- National Center for Advancing Translational Sciences/National Institutes of Health, Bethesda, Maryland, USA
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Kerstin Hahn
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Julia Kühnlenz
- Bayer SAS, CropScience, Pathology & Mechanistic Toxicology, Sophia Antipolis, France
| | - Randolph S Ashton
- University of Wisconsin-Madison, Madison, Wisconsin, USA
- Neurosetta LLC, Madison, Wisconsin, USA
| | - Min Tseng
- Genentech, South San Francisco, California, USA
| | | | | | | | | | | | - Lindsay Tomlinson
- Pfizer Inc., Drug Safety Research and Development, Cambridge, Massachusetts, USA
| |
Collapse
|
17
|
Beirute-Herrera J, López-Amo Calvo B, Edenhofer F, Esk C. The promise of genetic screens in human in vitro brain models. Biol Chem 2024; 405:13-24. [PMID: 37697643 DOI: 10.1515/hsz-2023-0174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023]
Abstract
Advances of in vitro culture models have allowed unprecedented insights into human neurobiology. At the same time genetic screening has matured into a robust and accessible experimental strategy allowing for the simultaneous study of many genes in parallel. The combination of both technologies is a newly emerging tool for neuroscientists, opening the door to identifying causal cell- and tissue-specific developmental and disease mechanisms. However, with complex experimental genetic screening set-ups new challenges in data interpretation and experimental scope arise that require a deep understanding of the benefits and challenges of individual approaches. In this review, we summarize the literature that applies genetic screening to in vitro brain models, compare experimental strengths and weaknesses and point towards future directions of these promising approaches.
Collapse
Affiliation(s)
- Julianne Beirute-Herrera
- Institute of Molecular Biology, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
- Center for Molecular Biosciences, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
| | - Beatriz López-Amo Calvo
- Institute of Molecular Biology, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
- Center for Molecular Biosciences, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
| | - Frank Edenhofer
- Institute of Molecular Biology, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
- Center for Molecular Biosciences, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
| | - Christopher Esk
- Institute of Molecular Biology, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
- Center for Molecular Biosciences, University Innsbruck, Technikerstr. 25, A-6020 Innsbruck, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Dr. Bohr Gasse 3, 1030 Vienna, Austria
| |
Collapse
|
18
|
Zheng Y. Stem Cell-Derived Microfluidic Amniotic Sac Embryoid (μPASE). Methods Mol Biol 2024; 2767:75-84. [PMID: 36749485 DOI: 10.1007/7651_2022_470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The microfluidic amniotic sac embryoid (μPASE) is a human pluripotent stem cell (hPSC)-derived multicellular human embryo-like structure with molecular and morphological features resembling the progressive development of the early post-implantation human embryonic sac. The microfluidic device is specifically designed to control the formation of hPSC clusters and expose the clusters to different morphogen environments, allowing the development of μPASEs in a highly controllable, reproducible, and scalable fashion. The μPASE model displays human embryonic developmental landmarks such as lumenogenesis of the epiblast, amniotic cavity formation, and the specification of primordial germ cells and gastrulating cells (or mesendoderm cells). Here, we provide detailed instructions needed to reproduce μPASEs, including the immunofluorescence staining and cell retrieval protocols for characterizing μPASEs obtained under different experimental conditions.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| |
Collapse
|
19
|
Isomursu A, Alanko J, Hernández-Pérez S, Saukkonen K, Saari M, Mattila PK, Ivaska J. Dynamic Micropatterning Reveals Substrate-Dependent Differences in the Geometric Control of Cell Polarization and Migration. SMALL METHODS 2024; 8:e2300719. [PMID: 37926786 DOI: 10.1002/smtd.202300719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/03/2023] [Indexed: 11/07/2023]
Abstract
Cells are highly dynamic and adopt variable shapes and sizes. These variations are biologically important but challenging to investigate in a spatiotemporally controlled manner. Micropatterning, confining cells on microfabricated substrates with defined geometries and molecular compositions, is a powerful tool for controlling cell shape and interactions. However, conventional binary micropatterns are static and fail to address dynamic changes in cell polarity, spreading, and migration. Here, a method for dynamic micropatterning is reported, where the non-adhesive surface surrounding adhesive micropatterns is rapidly converted to support specific cell-matrix interactions while allowing simultaneous imaging of the cells. The technique is based on ultraviolet photopatterning of biotinylated polyethylene glycol-grafted poly-L-lysine, and it is simple, inexpensive, and compatible with a wide range of streptavidin-conjugated ligands. Experiments using biotinylation-based dynamic micropatterns reveal that distinct extracellular matrix ligands and bivalent integrin-clustering antibodies support different degrees of front-rear polarity in human glioblastoma cells, which correlates to altered directionality and persistence upon release and migration on fibronectin. Unexpectedly, however, neither an asymmetric cell shape nor centrosome orientation can fully predict the future direction of migration. Taken together, biotinylation-based dynamic micropatterns allow easily accessible and highly customizable control over cell morphology and motility.
Collapse
Affiliation(s)
- Aleksi Isomursu
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Jonna Alanko
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Sara Hernández-Pérez
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, 20014, Finland
- Department of Life Technologies, University of Turku, Turku, 20520, Finland
| | - Karla Saukkonen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Markku Saari
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Pieta K Mattila
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, 20014, Finland
- Department of Life Technologies, University of Turku, Turku, 20520, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- Department of Life Technologies, University of Turku, Turku, 20520, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, 20520, Finland
- Western Finnish Cancer Center (FICAN West), University of Turku, Turku, 20520, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, 00014, Finland
| |
Collapse
|
20
|
Louessard M, Cailleret M, Jarrige M, Bigarreau J, Lenoir S, Dufour N, Rey M, Saudou F, Deglon N, Perrier AL. Mono- and Biallelic Inactivation of Huntingtin Gene in Patient-Specific Induced Pluripotent Stem Cells Reveal HTT Roles in Striatal Development and Neuronal Functions. J Huntingtons Dis 2024; 13:41-53. [PMID: 38427495 DOI: 10.3233/jhd-231509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Mutations in the Huntingtin (HTT) gene cause Huntington's disease (HD), a neurodegenerative disorder. As a scaffold protein, HTT is involved in numerous cellular functions, but its normal and pathogenic functions during human forebrain development are poorly understood. Objective To investigate the developmental component of HD, with a specific emphasis on understanding the functions of wild-type and mutant HTT alleles during forebrain neuron development in individuals carrying HD mutations. Methods We used CRISPR/Cas9 gene-editing technology to disrupt the ATG region of the HTT gene via non-homologous end joining to produce mono- or biallelic HTT knock-out human induced pluripotent stem cell (iPSC) clones. Results We showed that the loss of wild-type, mutant, or both HTT isoforms does not affect the pluripotency of iPSCs or their transition into neural cells. However, we observed that HTT loss causes division impairments in forebrain neuro-epithelial cells and alters maturation of striatal projection neurons (SPNs) particularly in the acquisition of DARPP32 expression, a key functional marker of SPNs. Finally, young post-mitotic neurons derived from HTT-/- human iPSCs display cellular dysfunctions observed in adult HD neurons. Conclusions We described a novel collection of isogenic clones with mono- and biallelic HTT inactivation that complement existing HD-hiPSC isogenic series to explore HTT functions and test therapeutic strategies in particular HTT-lowering drugs. Characterizing neural and neuronal derivatives from human iPSCs of this collection, we show evidence that HTT loss or mutation has impacts on neuro-epithelial and striatal neurons maturation, and on basal DNA damage and BDNF axonal transport in post-mitotic neurons.
Collapse
Affiliation(s)
- Morgane Louessard
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: Mécanismes, Thérapies, Imagerie, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Michel Cailleret
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Margot Jarrige
- CECS/AFM, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Julie Bigarreau
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| | - Sophie Lenoir
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Noëlle Dufour
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: Mécanismes, Thérapies, Imagerie, Fontenay-aux-Roses, France
| | - Maria Rey
- Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Department of Clinical Neurosciences (DNC), and Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies, Lausanne, Switzerland
| | - Frédéric Saudou
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Nicole Deglon
- Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Department of Clinical Neurosciences (DNC), and Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies, Lausanne, Switzerland
| | - Anselme L Perrier
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: Mécanismes, Thérapies, Imagerie, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, Univ Evry, Institut des Cellules Souches pour le Traitement et l'étude des Maladies Monogéniques, Corbeil-Essonne, France
| |
Collapse
|
21
|
Mirdass C, Catala M, Bocel M, Nedelec S, Ribes V. Stem cell-derived models of spinal neurulation. Emerg Top Life Sci 2023; 7:423-437. [PMID: 38087891 DOI: 10.1042/etls20230087] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Neurulation is a critical step in early embryonic development, giving rise to the neural tube, the primordium of the central nervous system in amniotes. Understanding this complex, multi-scale, multi-tissue morphogenetic process is essential to provide insights into normal development and the etiology of neural tube defects. Innovations in tissue engineering have fostered the generation of pluripotent stem cell-based in vitro models, including organoids, that are emerging as unique tools for delving into neurulation mechanisms, especially in the context of human development. Each model captures specific aspects of neural tube morphogenesis, from epithelialization to neural tissue elongation, folding and cavitation. In particular, the recent models of human and mouse trunk morphogenesis, such as gastruloids, that form a spinal neural plate-like or neural tube-like structure are opening new avenues to study normal and pathological neurulation. Here, we review the morphogenetic events generating the neural tube in the mammalian embryo and questions that remain unanswered. We discuss the advantages and limitations of existing in vitro models of neurulation and possible future technical developments.
Collapse
Affiliation(s)
- Camil Mirdass
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Institut du Fer à Moulin, 75005 Paris, France
- Inserm, UMR-S 1270, 75005 Paris, France
- Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Martin Catala
- Institut de Biologie Paris Seine (IBPS) - Developmental Biology Laboratory, UMR7622 CNRS, INSERM ERL 1156, Sorbonne Université, 9 Quai Saint-Bernard, 75005 Paris, France
| | - Mikaëlle Bocel
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Stéphane Nedelec
- Institut du Fer à Moulin, 75005 Paris, France
- Inserm, UMR-S 1270, 75005 Paris, France
- Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Vanessa Ribes
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| |
Collapse
|
22
|
Lee JH, Shaker MR, Park SH, Sun W. Transcriptional Signature of Valproic Acid-Induced Neural Tube Defects in Human Spinal Cord Organoids. Int J Stem Cells 2023; 16:385-393. [PMID: 37643760 PMCID: PMC10686804 DOI: 10.15283/ijsc23012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/15/2023] [Accepted: 06/20/2023] [Indexed: 08/31/2023] Open
Abstract
In vertebrates, the entire central nervous system is derived from the neural tube, which is formed through a conserved early developmental morphogenetic process called neurulation. Although the perturbations in neurulation caused by genetic or environmental factors lead to neural tube defects (NTDs), the most common congenital malformation and the precise molecular pathological cascades mediating NTDs are not well understood. Recently, we have developed human spinal cord organoids (hSCOs) that recapitulate some aspects of human neurulation and observed that valproic acid (VPA) could cause neurulation defects in an organoid model. In this study, we identified and verified the significant changes in cell-cell junctional genes/proteins in VPA-treated organoids using transcriptomic and immunostaining analysis. Furthermore, VPA-treated mouse embryos exhibited impaired gene expression and NTD phenotypes, similar to those observed in the hSCO model. Collectively, our data demonstrate that hSCOs provide a valuable biological resource for dissecting the molecular pathways underlying the currently unknown human neurulation process using destructive biological analysis tools.
Collapse
Affiliation(s)
- Ju-Hyun Lee
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Mohammed R. Shaker
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Si-Hyung Park
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Thompson LM, Orr HT. HD and SCA1: Tales from two 30-year journeys since gene discovery. Neuron 2023; 111:3517-3530. [PMID: 37863037 PMCID: PMC10842341 DOI: 10.1016/j.neuron.2023.09.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/21/2023] [Accepted: 09/26/2023] [Indexed: 10/22/2023]
Abstract
One of the more transformative findings in human genetics was the discovery that the expansion of unstable nucleotide repeats underlies a group of inherited neurological diseases. A subset of these unstable repeat neurodegenerative diseases is due to the expansion of a CAG trinucleotide repeat encoding a stretch of glutamines, i.e., the polyglutamine (polyQ) repeat neurodegenerative diseases. Among the CAG/polyQ repeat diseases are Huntington's disease (HD) and spinocerebellar ataxia type 1 (SCA1), in which the expansions are within widely expressed proteins. Although both HD and SCA1 are autosomal dominantly inherited, and both typically cause mid- to late-life-onset movement disorders with cognitive decline, they each are characterized by distinct clinical characteristics and predominant sites of neuropathology. Importantly, the respective affected proteins, Huntingtin (HTT, HD) and Ataxin 1 (ATXN1, SCA1), have unique functions and biological properties. Here, we review HD and SCA1 with a focus on how their disease-specific and shared features may provide informative insights.
Collapse
Affiliation(s)
- Leslie M Thompson
- Department of Psychiatry and Human Behavior, Department of Neurobiology and Behavior, Department of Biological Chemistry, Institute of Memory Impairments and Neurological Disorders, Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota, Minneapolis and Saint Paul, MN 55455, USA.
| |
Collapse
|
24
|
Jordan R, Ford-Scheimer SL, Alarcon RM, Atala A, Borenstein JT, Brimacombe KR, Cherry S, Clevers H, Davis MI, Funnell SGP, Gehrke L, Griffith LG, Grossman AC, Hartung T, Ingber DE, Kleinstreuer NC, Kuo CJ, Lee EM, Mummery CL, Pickett TE, Ramani S, Rosado-Olivieri EA, Struble EB, Wan Z, Williams MS, Hall MD, Ferrer M, Markossian S. Report of the Assay Guidance Workshop on 3-Dimensional Tissue Models for Antiviral Drug Development. J Infect Dis 2023; 228:S337-S354. [PMID: 37669225 PMCID: PMC10547463 DOI: 10.1093/infdis/jiad334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
The National Center for Advancing Translational Sciences (NCATS) Assay Guidance Manual (AGM) Workshop on 3D Tissue Models for Antiviral Drug Development, held virtually on 7-8 June 2022, provided comprehensive coverage of critical concepts intended to help scientists establish robust, reproducible, and scalable 3D tissue models to study viruses with pandemic potential. This workshop was organized by NCATS, the National Institute of Allergy and Infectious Diseases, and the Bill and Melinda Gates Foundation. During the workshop, scientific experts from academia, industry, and government provided an overview of 3D tissue models' utility and limitations, use of existing 3D tissue models for antiviral drug development, practical advice, best practices, and case studies about the application of available 3D tissue models to infectious disease modeling. This report includes a summary of each workshop session as well as a discussion of perspectives and challenges related to the use of 3D tissues in antiviral drug discovery.
Collapse
Affiliation(s)
- Robert Jordan
- Bill and Melinda Gates Foundation, Seattle, Washington, USA
| | - Stephanie L Ford-Scheimer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Rodolfo M Alarcon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Kyle R Brimacombe
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Mindy I Davis
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Simon G P Funnell
- UK Health Security Agency, Salisbury, United Kingdom
- Quadram Institute Bioscience, Norwich, United Kingdom
| | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Abigail C Grossman
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Thomas Hartung
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Donald E Ingber
- Harvard Medical School, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- Harvard School of Engineering and Applied Sciences, Cambridge, Massachusetts, USA
- Boston Children's Hospital, Boston, Massachusetts, USA
| | - Nicole C Kleinstreuer
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle, North Carolina, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | - Emily M Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | | | - Thames E Pickett
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Evi B Struble
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mark S Williams
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Sarine Markossian
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
25
|
Sullivan AE, Santos SD. The ever-growing world of gastruloids: autogenous models of mammalian embryogenesis. Curr Opin Genet Dev 2023; 82:102102. [PMID: 37604096 DOI: 10.1016/j.gde.2023.102102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 08/23/2023]
Abstract
During early development, extrinsic cues prompt a collection of pluripotent cells to begin the extensive process of cellular differentiation that gives rise to all tissues in the mammalian embryo, a process known as gastrulation. Advances in stem cell biology have resulted in the generation of stem cell-based in vitro models of mammalian gastrulation called gastruloids. Gastruloids and subsequent gastruloid-based models are tractable, scalable and more accessible than mammalian embryos. As such, they have opened an unprecedented avenue for modelling in vitro self-organisation, patterning and fate specification. This review focuses on discussing the recent advances of this rapidly moving research area, clarifying what structures they model and the underlying signal hierarchy. We highlight the exciting potential of these models and where the field might be heading.
Collapse
Affiliation(s)
- Adrienne E Sullivan
- Quantitative Cell Biology Laboratory, The Francis Crick Institute, 1-Midland Road, NW1 1AT London, UK.
| | - Silvia Dm Santos
- Quantitative Cell Biology Laboratory, The Francis Crick Institute, 1-Midland Road, NW1 1AT London, UK.
| |
Collapse
|
26
|
Huang K, Li Q, Xue Y, Wang Q, Chen Z, Gu Z. Application of colloidal photonic crystals in study of organoids. Adv Drug Deliv Rev 2023; 201:115075. [PMID: 37625595 DOI: 10.1016/j.addr.2023.115075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 07/09/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
As alternative disease models, other than 2D cell lines and patient-derived xenografts, organoids have preferable in vivo physiological relevance. However, both endogenous and exogenous limitations impede the development and clinical translation of these organoids. Fortunately, colloidal photonic crystals (PCs), which benefit from favorable biocompatibility, brilliant optical manipulation, and facile chemical decoration, have been applied to the engineering of organoids and have achieved the desirable recapitulation of the ECM niche, well-defined geometrical onsets for initial culture, in situ multiphysiological parameter monitoring, single-cell biomechanical sensing, and high-throughput drug screening with versatile functional readouts. Herein, we review the latest progress in engineering organoids fabricated from colloidal PCs and provide inputs for future research.
Collapse
Affiliation(s)
- Kai Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Qiwei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yufei Xue
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Qiong Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, Jiangsu 215163, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
27
|
Lin F, Li X, Sun S, Li Z, Lv C, Bai J, Song L, Han Y, Li B, Fu J, Shao Y. Mechanically enhanced biogenesis of gut spheroids with instability-driven morphomechanics. Nat Commun 2023; 14:6016. [PMID: 37758697 PMCID: PMC10533890 DOI: 10.1038/s41467-023-41760-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Region-specific gut spheroids are precursors for gastrointestinal and pulmonary organoids that hold great promise for fundamental studies and translations. However, efficient production of gut spheroids remains challenging due to a lack of control and mechanistic understanding of gut spheroid morphogenesis. Here, we report an efficient biomaterial system, termed micropatterned gut spheroid generator (μGSG), to generate gut spheroids from human pluripotent stem cells through mechanically enhanced tissue morphogenesis. We show that μGSG enhances the biogenesis of gut spheroids independent of micropattern shape and size; instead, mechanically enforced cell multilayering and crowding is demonstrated as a general, geometry-insensitive mechanism that is necessary and sufficient for promoting spheroid formation. Combining experimental findings and an active-phase-field morphomechanics theory, our study further reveals an instability-driven mechanism and a mechanosensitive phase diagram governing spheroid pearling and fission in μGSG. This work unveils mechanobiological paradigms based on tissue architecture and surface tension for controlling tissue morphogenesis and advancing organoid technology.
Collapse
Affiliation(s)
- Feng Lin
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Xia Li
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Shiyu Sun
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Zhongyi Li
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Chenglin Lv
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Jianbo Bai
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Lin Song
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Yizhao Han
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yue Shao
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, 100084, China.
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
| |
Collapse
|
28
|
Liorni N, Napoli A, Castellana S, Giallongo S, Řeháková D, Re OL, Koutná I, Mazza T, Vinciguerra M. Integrative CUT&Tag-RNA-Seq analysis of histone variant macroH2A1-dependent orchestration of human induced pluripotent stem cell reprogramming. Epigenomics 2023; 15:863-877. [PMID: 37846557 DOI: 10.2217/epi-2023-0267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
Aim: Human induced pluripotent stem cells (iPSCs) are inefficiently derived from somatic cells by overexpression of defined transcription factors. Overexpression of H2A histone variant macroH2A1.1, but not macroH2A1.2, leads to increased iPSC reprogramming by unclear mechanisms. Materials & methods: Cleavage under targets and tagmentation (CUT&Tag) allows robust epigenomic profiling of a low cell number. We performed an integrative CUT&Tag-RNA-Seq analysis of macroH2A1-dependent orchestration of iPSCs reprogramming using human endothelial cells. Results: We demonstrate wider genome occupancy, predicted transcription factors binding, and gene expression regulated by macroH2A1.1 during reprogramming, compared to macroH2A1.2. MacroH2A1.1, previously associated with neurodegenerative pathologies, specifically activated ectoderm/neural processes. Conclusion: CUT&Tag and RNA-Seq data integration is a powerful tool to investigate the epigenetic mechanisms occurring during cell reprogramming.
Collapse
Affiliation(s)
- Niccolò Liorni
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza,71013, San Giovanni Rotondo, Italy
| | - Alessandro Napoli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza,71013, San Giovanni Rotondo, Italy
| | - Stefano Castellana
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza,71013, San Giovanni Rotondo, Italy
| | - Sebastiano Giallongo
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Department of Biomedical & Biotechnological Sciences, University of Catania, 95123, Catania, Italy
| | - Daniela Řeháková
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Institute of Experimental Biology, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Oriana Lo Re
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Department of Translational Stem Cell Biology, Research Institute, Medical University of Varna (RIMUV), 9002, Varna, Bulgaria
| | - Irena Koutná
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Department of Histology & Embryology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza,71013, San Giovanni Rotondo, Italy
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Department of Translational Stem Cell Biology, Research Institute, Medical University of Varna (RIMUV), 9002, Varna, Bulgaria
- Faculty of Health, Liverpool John Moores University, L2 2ER, Liverpool, UK
| |
Collapse
|
29
|
Laundos TL, Li S, Cheang E, De Santis R, Piccolo FM, Brivanlou AH. Huntingtin CAG-expansion mutation results in a dominant negative effect. Front Cell Dev Biol 2023; 11:1252521. [PMID: 37727506 PMCID: PMC10505792 DOI: 10.3389/fcell.2023.1252521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/14/2023] [Indexed: 09/21/2023] Open
Abstract
Introduction: Huntington's disease (HD) remains an incurable and fatal neurodegenerative disease long after CAG-expansion mutation in the huntingtin gene (HTT) was identified as the cause. The underlying pathological mechanism, whether HTT loss of function or gain of toxicity results from mutation, remains a matter of debate. Methods: In this study, we genetically modulated wild-type or mutant HTT expression levels in isogenic human embryonic stem cells to systematically investigate their contribution to HD-specific phenotypes. Results: Using highly reproducible and quantifiable in vitro micropattern-based assays, we observed comparable phenotypes with HD mutation and HTT depletion. However, halving endogenous wild-type HTT levels did not strongly recapitulate the HD phenotypes, arguing against a classical loss of function mechanism. Remarkably, expression of CAG-expanded HTT in non-HD cells induced HD like phenotypes akin to HTT depletion. Discussion: By corollary, these results indicate a dominant negative effect of mutated HTT on its wild-type counterpart. Complementation with additional copies of wild-type HTT ameliorated the HD-associated phenotypes, strongly supporting a classical dominant negative mechanism. Understanding the molecular basis of this dominant negative effect will guide the development of efficient clinical strategies to counteract the deleterious impact of mutant HTT on the wild-type HTT function.
Collapse
Affiliation(s)
- Tiago L. Laundos
- Laboratory of Synthetic Embryology, The Rockefeller University, New York City, NY, United States
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Shu Li
- Laboratory of Synthetic Embryology, The Rockefeller University, New York City, NY, United States
| | - Eric Cheang
- Laboratory of Synthetic Embryology, The Rockefeller University, New York City, NY, United States
| | - Riccardo De Santis
- Laboratory of Synthetic Embryology, The Rockefeller University, New York City, NY, United States
| | - Francesco M. Piccolo
- Laboratory of Synthetic Embryology, The Rockefeller University, New York City, NY, United States
| | - Ali H. Brivanlou
- Laboratory of Synthetic Embryology, The Rockefeller University, New York City, NY, United States
| |
Collapse
|
30
|
Qian W, Good MC. Peeking under the hood of early embryogenesis: Using tools and synthetic biology to understand native control systems and sculpt tissues. Semin Cell Dev Biol 2023; 141:43-49. [PMID: 35525819 PMCID: PMC9633583 DOI: 10.1016/j.semcdb.2022.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/13/2022] [Indexed: 10/18/2022]
Abstract
Early embryogenesis requires rapid division of pluripotent blastomeres, regulated genome activation, precise spatiotemporal signaling to pattern cell fate, and morphogenesis to shape primitive tissue architectures. The complexity of this process has inspired researchers to move beyond simple genetic perturbation into engineered devices and synthetic biology tools to permit temporal and spatial manipulation of the control systems guiding development. By precise alteration of embryo organization, it is now possible to advance beyond basic analytical strategies and directly test the sufficiency of models for developmental regulation. Separately, advances in micropatterning and embryoid culture have facilitated the bottom-up construction of complex embryo tissues allowing ex vivo systems to recapitulate even later stages of development. Embryos fertilized and grown ex vivo offer an excellent opportunity to exogenously perturb fundamental pathways governing embryogenesis. Here we review the technologies developed to thermally modulate the embryo cell cycle, and optically regulate morphogen and signaling pathways in space and time, specifically in the blastula embryo. Additionally, we highlight recent advances in cell patterning in two and three dimensions that have helped reveal the self-organizing properties and gene regulatory networks guiding early embryo organization.
Collapse
Affiliation(s)
- Wenchao Qian
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Cell and Molecular Biology Graduate Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew C. Good
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA,Cell and Molecular Biology Graduate Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Lead Contact,Correspondence: (M.C.G), Address: 421 Curie Blvd, 1151 Biomedical Research Building, Philadelphia PA 19104
| |
Collapse
|
31
|
Zhai J, Xu Y, Wan H, Yan R, Guo J, Skory R, Yan L, Wu X, Sun F, Chen G, Zhao W, Yu K, Li W, Guo F, Plachta N, Wang H. Neurulation of the cynomolgus monkey embryo achieved from 3D blastocyst culture. Cell 2023; 186:2078-2091.e18. [PMID: 37172562 DOI: 10.1016/j.cell.2023.04.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/15/2022] [Accepted: 04/12/2023] [Indexed: 05/15/2023]
Abstract
Neural tube (NT) defects arise from abnormal neurulation and result in the most common birth defects worldwide. Yet, mechanisms of primate neurulation remain largely unknown due to prohibitions on human embryo research and limitations of available model systems. Here, we establish a three-dimensional (3D) prolonged in vitro culture (pIVC) system supporting cynomolgus monkey embryo development from 7 to 25 days post-fertilization. Through single-cell multi-omics analyses, we demonstrate that pIVC embryos form three germ layers, including primordial germ cells, and establish proper DNA methylation and chromatin accessibility through advanced gastrulation stages. In addition, pIVC embryo immunofluorescence confirms neural crest formation, NT closure, and neural progenitor regionalization. Finally, we demonstrate that the transcriptional profiles and morphogenetics of pIVC embryos resemble key features of similarly staged in vivo cynomolgus and human embryos. This work therefore describes a system to study non-human primate embryogenesis through advanced gastrulation and early neurulation.
Collapse
Affiliation(s)
- Jinglei Zhai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanhong Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haifeng Wan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Rui Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jing Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Robin Skory
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Long Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Xulun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Fengyuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Gang Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Wentao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Kunyuan Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Fan Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Nicolas Plachta
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|
32
|
Cassel de Camps C, Mok S, Ashby E, Li C, Lépine P, Durcan TM, Moraes C. Compressive molding of engineered tissues via thermoresponsive hydrogel devices. LAB ON A CHIP 2023; 23:2057-2067. [PMID: 36916609 DOI: 10.1039/d3lc00007a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Biofabrication of tissues requires sourcing appropriate combinations of cells, and then arranging those cells into a functionally-useful construct. Recently, organoids with diverse cell populations have shown great promise as building blocks from which to assemble more complex structures. However, organoids typically adopt spherical or uncontrolled morphologies, which intrinsically limit the tissue structures that can be produced using this bioassembly technique. Here, we develop microfabricated smart hydrogel platforms in thermoresponsive poly(N-isopropylacrylamide) to compressively mold microtissues such as spheroids or organoids into customized forms, on demand. These Compressive Hydrogel Molders (CHyMs) compact at cell culture temperatures to force loaded tissues into a new shape, and then expand to release the tissues for downstream applications. As a first demonstration, breast cancer spheroids were biaxially compacted in cylindrical cavities, and uniaxially compacted in rectangular ones. Spheroid shape changes persisted after the tissues were released from the CHyMs. We then demonstrate long-term molding of spherical brain organoids in ring-shaped CHyMs over one week. Fused bridges formed only when brain organoids were encased in Matrigel, and the resulting ring-shaped organoids expressed tissue markers that correspond with expected differentiation profiles. These results demonstrate that tissues differentiate appropriately even during long-term molding in a CHyM. This platform hence provides a new tool to shape pre-made tissues as desired, via temporary compression and release, allowing an exploration of alternative organoid geometries as building blocks for bioassembly applications.
Collapse
Affiliation(s)
| | - Stephanie Mok
- Department of Chemical Engineering, McGill University, Montréal, H3A 0C5 QC, Canada
| | - Emily Ashby
- Department of Chemical Engineering, McGill University, Montréal, H3A 0C5 QC, Canada
| | - Chen Li
- Department of Chemical Engineering, McGill University, Montréal, H3A 0C5 QC, Canada
| | - Paula Lépine
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University, 3801 University Street, Montréal, H3A 2B4 QC, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University, 3801 University Street, Montréal, H3A 2B4 QC, Canada
| | - Christopher Moraes
- Department of Biomedical Engineering, McGill University, Montréal, H3A 2B4 QC, Canada.
- Department of Chemical Engineering, McGill University, Montréal, H3A 0C5 QC, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, H3A 1A3 QC, Canada
- Division of Experimental Medicine, McGill University, Montréal, H4A 3J1, QC, Canada
| |
Collapse
|
33
|
Rosado-Olivieri EA, Razooky B, Le Pen J, De Santis R, Barrows D, Sabry Z, Hoffmann HH, Park J, Carroll TS, Poirier JT, Rice CM, Brivanlou AH. Organotypic human lung bud microarrays identify BMP-dependent SARS-CoV-2 infection in lung cells. Stem Cell Reports 2023; 18:1107-1122. [PMID: 37084725 PMCID: PMC10116630 DOI: 10.1016/j.stemcr.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/23/2023] Open
Abstract
Although lung disease is the primary clinical outcome in COVID-19 patients, how SARS-CoV-2 induces lung pathology remains elusive. Here we describe a high-throughput platform to generate self-organizing and commensurate human lung buds derived from hESCs cultured on micropatterned substrates. Lung buds resemble human fetal lungs and display proximodistal patterning of alveolar and airway tissue directed by KGF. These lung buds are susceptible to infection by SARS-CoV-2 and endemic coronaviruses and can be used to track cell type-specific cytopathic effects in hundreds of lung buds in parallel. Transcriptomic comparisons of infected lung buds and postmortem tissue of COVID-19 patients identified an induction of BMP signaling pathway. BMP activity renders lung cells more susceptible to SARS-CoV-2 infection and its pharmacological inhibition impairs infection by this virus. These data highlight the rapid and scalable access to disease-relevant tissue using lung buds that recapitulate key features of human lung morphogenesis and viral infection biology.
Collapse
Affiliation(s)
- E A Rosado-Olivieri
- Laboratory of Synthetic Embryology, the Rockefeller University, New York, NY, USA
| | - B Razooky
- Laboratory of Virology and Infectious Diseases, the Rockefeller University, New York, NY, USA
| | - J Le Pen
- Laboratory of Virology and Infectious Diseases, the Rockefeller University, New York, NY, USA
| | - R De Santis
- Laboratory of Synthetic Embryology, the Rockefeller University, New York, NY, USA
| | - D Barrows
- Bioinformatics Resource Center, the Rockefeller University, New York, NY, USA
| | - Z Sabry
- Laboratory of Synthetic Embryology, the Rockefeller University, New York, NY, USA
| | - H-H Hoffmann
- Laboratory of Virology and Infectious Diseases, the Rockefeller University, New York, NY, USA
| | - J Park
- Laboratory of Virology and Infectious Diseases, the Rockefeller University, New York, NY, USA
| | - T S Carroll
- Bioinformatics Resource Center, the Rockefeller University, New York, NY, USA
| | - J T Poirier
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - C M Rice
- Laboratory of Virology and Infectious Diseases, the Rockefeller University, New York, NY, USA.
| | - A H Brivanlou
- Laboratory of Synthetic Embryology, the Rockefeller University, New York, NY, USA.
| |
Collapse
|
34
|
Fabbri M, Ginoza M, Assen L, Jongsma K, Isasi R. Modeling policy development: examining national governance of stem cell-based embryo models. Regen Med 2023; 18:155-168. [PMID: 36601984 DOI: 10.2217/rme-2022-0136] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Researchers can now coax human pluripotent stem cells to imitate the structure and spontaneous self-organization of the developing human embryo. Although these stem cell-based embryo models present an advantageous alternative to embryo research, they also raise ethical and policy challenges. In 2021, the International Society for Stem Cell Research revised its Guidelines for Stem Cell Research and Clinical Translation, providing contemporaneous best practices for ethical conduct in the field. The Guidelines complement national governance frameworks; however, they also contain contentious and aspirational norms that might catalyze change in research practice and in the enactment of national policies. Using a sample of 11 research-intensive countries, the authors compare research policy frameworks against the International Society for Stem Cell Research Guidelines to showcase how developments in global and national policies might affect stem cell-based embryo model research governance and illustrate fertile areas for ethical reflection and policy development.
Collapse
Affiliation(s)
- Morris Fabbri
- Dr John T Macdonald Foundation Department of Human Genetics, John P Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Lars Assen
- University Medical Center Utrecht, Utrecht, 3584, The Netherlands
| | - Karin Jongsma
- University Medical Center Utrecht, Utrecht, 3584, The Netherlands
| | - Rosario Isasi
- Dr John T Macdonald Foundation Department of Human Genetics, John P Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Dr John T Macdonald Foundation Department of Human Genetics, John P Hussman Institute for Human Genomics, Interdisciplinary Stem Cell Institute, Miami, FL 33136, USA
| |
Collapse
|
35
|
Jeong E, Choi S, Cho SW. Recent Advances in Brain Organoid Technology for Human Brain Research. ACS APPLIED MATERIALS & INTERFACES 2023; 15:200-219. [PMID: 36468535 DOI: 10.1021/acsami.2c17467] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Brain organoids are self-assembled three-dimensional aggregates with brain-like cell types and structures and have emerged as new model systems that can be used to investigate human neurodevelopment and neurological disorders. However, brain organoids are not as mature and functional as real human brains due to limitations of the culture system with insufficient developmental patterning signals and a lack of components that are important for brain development and function, such as the non-neural population and vasculature. In addition, establishing the desired brain-like environment and monitoring the complex neural networks and physiological functions of the brain organoids remain challenging. The current protocols to generate brain organoids also have problems with heterogeneity and batch variation due to spontaneous self-organization of brain organoids into complex architectures of the brain. To address these limitations of current brain organoid technologies, various engineering platforms, such as extracellular matrices, fluidic devices, three-dimensional bioprinting, bioreactors, polymeric scaffolds, microelectrodes, and biochemical sensors, have been employed to improve neuronal development and maturation, reduce structural heterogeneity, and facilitate functional analysis and monitoring. In this review, we provide an overview of the latest engineering techniques that overcome these limitations in the production and application of brain organoids.
Collapse
Affiliation(s)
- Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Suah Choi
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
36
|
Hoang P, Sun S, Tarris BA, Ma Z. Controlling Morphology and Functions of Cardiac Organoids by Two-Dimensional Geometrical Templates. Cells Tissues Organs 2023; 212:64-73. [PMID: 35008091 PMCID: PMC9271134 DOI: 10.1159/000521787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/21/2021] [Indexed: 11/19/2022] Open
Abstract
Traditionally, tissue-specific organoids are generated as 3D aggregates of stem cells embedded in Matrigel or hydrogels, and the aggregates eventually end up a spherical shape and suspended in the matrix. Lack of geometrical control of organoid formation makes these spherical organoids limited for modeling the tissues with complex shapes. To address this challenge, we developed a new method to generate 3D spatial-organized cardiac organoids from 2D micropatterned human induced pluripotent stem cell (hiPSC) colonies, instead of directly from 3D stem cell aggregates. This new approach opens the possibility to create cardiac organoids that are templated by 2D non-spherical geometries, which potentially provides us a deeper understanding of biophysical controls on developmental organogenesis. Here, we designed 2D geometrical templates with quadrilateral shapes and pentagram shapes that had same total area but different geometrical shapes. Using this templated substrate, we grew cardiac organoids from hiPSCs and collected a series of parameters to characterize morphological and functional properties of the cardiac organoids. In quadrilateral templates, we found that increasing the aspect ratio impaired cardiac tissue 3D self-assembly, but the elongated geometry improved the cardiac contractile functions. However, in pentagram templates, cardiac organoid structure and function were optimized with a specific geometry of an ideal star shape. This study will shed a light on "organogenesis-by-design" by increasing the intricacy of starting templates from external geometrical cues to improve the organoid morphogenesis and functionality.
Collapse
Affiliation(s)
- Plansky Hoang
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York, USA.,BioInspired Syracuse Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Shiyang Sun
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York, USA.,BioInspired Syracuse Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Bearett A Tarris
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York, USA.,BioInspired Syracuse Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Zhen Ma
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York, USA.,BioInspired Syracuse Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| |
Collapse
|
37
|
Lim RG, Al-Dalahmah O, Wu J, Gold MP, Reidling JC, Tang G, Adam M, Dansu DK, Park HJ, Casaccia P, Miramontes R, Reyes-Ortiz AM, Lau A, Hickman RA, Khan F, Paryani F, Tang A, Ofori K, Miyoshi E, Michael N, McClure N, Flowers XE, Vonsattel JP, Davidson S, Menon V, Swarup V, Fraenkel E, Goldman JE, Thompson LM. Huntington disease oligodendrocyte maturation deficits revealed by single-nucleus RNAseq are rescued by thiamine-biotin supplementation. Nat Commun 2022; 13:7791. [PMID: 36543778 PMCID: PMC9772349 DOI: 10.1038/s41467-022-35388-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
The complexity of affected brain regions and cell types is a challenge for Huntington's disease (HD) treatment. Here we use single nucleus RNA sequencing to investigate molecular pathology in the cortex and striatum from R6/2 mice and human HD post-mortem tissue. We identify cell type-specific and -agnostic signatures suggesting oligodendrocytes (OLs) and oligodendrocyte precursors (OPCs) are arrested in intermediate maturation states. OL-lineage regulators OLIG1 and OLIG2 are negatively correlated with CAG length in human OPCs, and ATACseq analysis of HD mouse NeuN-negative cells shows decreased accessibility regulated by OL maturation genes. The data implicates glucose and lipid metabolism in abnormal cell maturation and identify PRKCE and Thiamine Pyrophosphokinase 1 (TPK1) as central genes. Thiamine/biotin treatment of R6/1 HD mice to compensate for TPK1 dysregulation restores OL maturation and rescues neuronal pathology. Our insights into HD OL pathology spans multiple brain regions and link OL maturation deficits to abnormal thiamine metabolism.
Collapse
Affiliation(s)
- Ryan G Lim
- UCI MIND, University of California Irvine, Irvine, CA, USA
| | - Osama Al-Dalahmah
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Maxwell P Gold
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Guomei Tang
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Miriam Adam
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David K Dansu
- Advanced Science Research Center at the City University of New York, New York, NY, USA
| | - Hye-Jin Park
- Advanced Science Research Center at the City University of New York, New York, NY, USA
| | - Patrizia Casaccia
- Advanced Science Research Center at the City University of New York, New York, NY, USA
| | | | - Andrea M Reyes-Ortiz
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Alice Lau
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
| | - Richard A Hickman
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Fatima Khan
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Fahad Paryani
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Alice Tang
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Kenneth Ofori
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily Miyoshi
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Neethu Michael
- Department of Pathology, University of California Irvine, Irvine, CA, USA
| | - Nicolette McClure
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Xena E Flowers
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, New York, NY, USA
| | - Jean Paul Vonsattel
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, New York, NY, USA
| | - Shawn Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton, NJ, USA
| | - Vilas Menon
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Vivek Swarup
- UCI MIND, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, New York, NY, USA.
| | - Leslie M Thompson
- UCI MIND, University of California Irvine, Irvine, CA, USA.
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA.
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Sue and Bill Gross Stem Cell Center University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
38
|
Humbert S, Barnat M. Huntington's disease and brain development. C R Biol 2022; 345:77-90. [PMID: 36847466 DOI: 10.5802/crbiol.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Huntington's disease is a rare inherited neurological disorder that generally manifests in mild-adulthood. The disease is characterized by the dysfunction and the degeneration of specific brain structures leading progressively to psychiatric, cognitive and motor disorders. The disease is caused by a mutation in the gene coding for huntingtin and, although it appears in adulthood, embryos carry the mutated gene from their development in utero. Studies based on mouse models and human stem cells have reported altered developmental mechanisms in disease conditions. However, does the mutation affect development in humans? Focusing on the early stages of brain development in human fetuses carrying the HD mutation, we have identified abnormalities in the development of the neocortex, the structure that ensure higher cerebral functions. Altogether, these studies suggests that developmental defects could contribute to the onset symptoms in adults, changing the perspective on disease and thus the health care of patients.
Collapse
|
39
|
Ku T, Ren Z, Yang R, Liu QS, Sang N, Faiola F, Zhou Q, Jiang G. Abnormal neural differentiation in response to graphene quantum dots through histone modification interference. ENVIRONMENT INTERNATIONAL 2022; 170:107572. [PMID: 36228552 DOI: 10.1016/j.envint.2022.107572] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
Graphene quantum dots (GQDs) have been broadly applied in biomedicine in recent years, and their environmental exposure and toxicological impacts have raised increasing concerns. The nanosafety assessment on the nervous system is one of the most important aspects, and potential effects of GQDs on neurodevelopment and the underlying mechanism are still elusive. In this study, the neural developmental toxicities of OH-GQDs and NH2-GQDs were investigated using the mouse embryonic stem cells (mESCs). The results revealed that OH-GQDs significantly inhibited the ectoderm development, and reduced the neural precursor formation and neurogenesis during the neural differentiation of the mESCs. The exploration on the mechanism uncovered that the increased enrichment of H3K27me3 at the promoter region of the Smad6 gene was involved in histone modification-activated BMP signal pathway, which consequently influenced its regulatory effects on neural differentiation. Additionally, OH-GQDs elicited a stronger effect on inducing the imbalance of histone modification, and resulted in higher latency of neural differentiation disturbance than did NH2-GQDs, suggesting surface functionalization-specific effects of GQDs on neurodevelopmental toxicity. This study would provide new insights in not only the adverse effects of GQDs on neurodevelopment, but also the influence from the chemical modification of GQDs on their bioactivities.
Collapse
Affiliation(s)
- Tingting Ku
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, 030006, China
| | - Zhihua Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, 030006, China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Environment and Health, Jianghan University, Wuhan 430056, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
40
|
Abstract
The complex process by which a single-celled zygote develops into a viable embryo is nothing short of a miraculous wonder of the natural world. Elucidating how this process is orchestrated in humans has long eluded the grasp of scientists due to ethical and practical limitations. Thankfully, pluripotent stem cells that resemble early developmental cell types possess the ability to mimic specific embryonic events. As such, murine and human stem cells have been leveraged by scientists to create in vitro models that aim to recapitulate different stages of early mammalian development. Here, we examine the wide variety of stem cell-based embryo models that have been developed to recapitulate and study embryonic events, from pre-implantation development through to early organogenesis. We discuss the applications of these models, key considerations regarding their importance within the field, and how such models are expected to grow and evolve to achieve exciting new milestones in the future.
Collapse
Affiliation(s)
- Aidan H. Terhune
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeyoon Bok
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
41
|
Metzger JJ, Pereda C, Adhikari A, Haremaki T, Galgoczi S, Siggia ED, Brivanlou AH, Etoc F. Deep-learning analysis of micropattern-based organoids enables high-throughput drug screening of Huntington's disease models. CELL REPORTS METHODS 2022; 2:100297. [PMID: 36160045 PMCID: PMC9500000 DOI: 10.1016/j.crmeth.2022.100297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 02/06/2022] [Accepted: 08/19/2022] [Indexed: 11/19/2022]
Abstract
Organoids are carrying the promise of modeling complex disease phenotypes and serving as a powerful basis for unbiased drug screens, potentially offering a more efficient drug-discovery route. However, unsolved technical bottlenecks of reproducibility and scalability have prevented the use of current organoids for high-throughput screening. Here, we present a method that overcomes these limitations by using deep-learning-driven analysis for phenotypic drug screens based on highly standardized micropattern-based neural organoids. This allows us to distinguish between disease and wild-type phenotypes in complex tissues with extremely high accuracy as well as quantify two predictors of drug success: efficacy and adverse effects. We applied our approach to Huntington's disease (HD) and discovered that bromodomain inhibitors revert complex phenotypes induced by the HD mutation. This work demonstrates the power of combining machine learning with phenotypic drug screening and its successful application to reveal a potentially new druggable target for HD.
Collapse
Affiliation(s)
- Jakob J. Metzger
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY 10065, USA
| | - Carlota Pereda
- RUMI Scientific, Alexandria LaunchLabs, New York, NY 10016, USA
| | - Arjun Adhikari
- RUMI Scientific, Alexandria LaunchLabs, New York, NY 10016, USA
| | - Tomomi Haremaki
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
- RUMI Scientific, Alexandria LaunchLabs, New York, NY 10016, USA
| | - Szilvia Galgoczi
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Eric D. Siggia
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY 10065, USA
| | - Ali H. Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Fred Etoc
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
- RUMI Scientific, Alexandria LaunchLabs, New York, NY 10016, USA
| |
Collapse
|
42
|
Zheng Y, Yan RZ, Sun S, Kobayashi M, Xiang L, Yang R, Goedel A, Kang Y, Xue X, Esfahani SN, Liu Y, Resto Irizarry AM, Wu W, Li Y, Ji W, Niu Y, Chien KR, Li T, Shioda T, Fu J. Single-cell analysis of embryoids reveals lineage diversification roadmaps of early human development. Cell Stem Cell 2022; 29:1402-1419.e8. [PMID: 36055194 PMCID: PMC9499422 DOI: 10.1016/j.stem.2022.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/08/2022] [Accepted: 08/11/2022] [Indexed: 01/03/2023]
Abstract
Despite its clinical and fundamental importance, our understanding of early human development remains limited. Stem cell-derived, embryo-like structures (or embryoids) allowing studies of early development without using natural embryos can potentially help fill the knowledge gap of human development. Herein, transcriptome at the single-cell level of a human embryoid model was profiled at different time points. Molecular maps of lineage diversifications from the pluripotent human epiblast toward the amniotic ectoderm, primitive streak/mesoderm, and primordial germ cells were constructed and compared with in vivo primate data. The comparative transcriptome analyses reveal a critical role of NODAL signaling in human mesoderm and primordial germ cell specification, which is further functionally validated. Through comparative transcriptome analyses and validations with human blastocysts and in vitro cultured cynomolgus embryos, we further proposed stringent criteria for distinguishing between human blastocyst trophectoderm and early amniotic ectoderm cells.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Robin Zhexuan Yan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mutsumi Kobayashi
- Massachusetts General Hospital Center for Cancer Research, Charlestown, MA 02129, USA
| | - Lifeng Xiang
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China
| | - Ran Yang
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Alexander Goedel
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Yu Kang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sajedeh Nasr Esfahani
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yue Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Weisheng Wu
- BRCF Bioinformatics Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yunxiu Li
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Yuyu Niu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Kenneth R Chien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Toshihiro Shioda
- Massachusetts General Hospital Center for Cancer Research, Charlestown, MA 02129, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
43
|
Iyer NR, Ashton RS. Bioengineering the human spinal cord. Front Cell Dev Biol 2022; 10:942742. [PMID: 36092702 PMCID: PMC9458954 DOI: 10.3389/fcell.2022.942742] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Three dimensional, self-assembled organoids that recapitulate key developmental and organizational events during embryogenesis have proven transformative for the study of human central nervous system (CNS) development, evolution, and disease pathology. Brain organoids have predominated the field, but human pluripotent stem cell (hPSC)-derived models of the spinal cord are on the rise. This has required piecing together the complex interactions between rostrocaudal patterning, which specifies axial diversity, and dorsoventral patterning, which establishes locomotor and somatosensory phenotypes. Here, we review how recent insights into neurodevelopmental biology have driven advancements in spinal organoid research, generating experimental models that have the potential to deepen our understanding of neural circuit development, central pattern generation (CPG), and neurodegenerative disease along the body axis. In addition, we discuss the application of bioengineering strategies to drive spinal tissue morphogenesis in vitro, current limitations, and future perspectives on these emerging model systems.
Collapse
Affiliation(s)
- Nisha R. Iyer
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Wisconsin Institute for Discovery, University of Wisconsin—Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
| | - Randolph S. Ashton
- Wisconsin Institute for Discovery, University of Wisconsin—Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
| |
Collapse
|
44
|
Zhang SY, Zhao J, Ni JJ, Li H, Quan ZZ, Qing H. Application and prospects of high-throughput screening for in vitro neurogenesis. World J Stem Cells 2022; 14:393-419. [PMID: 35949394 PMCID: PMC9244953 DOI: 10.4252/wjsc.v14.i6.393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/07/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, high-throughput screening (HTS) has made great contributions to new drug discovery. HTS technology is equipped with higher throughput, minimized platforms, more automated and computerized operating systems, more efficient and sensitive detection devices, and rapid data processing systems. At the same time, in vitro neurogenesis is gradually becoming important in establishing models to investigate the mechanisms of neural disease or developmental processes. However, challenges remain in generating more mature and functional neurons with specific subtypes and in establishing robust and standardized three-dimensional (3D) in vitro models with neural cells cultured in 3D matrices or organoids representing specific brain regions. Here, we review the applications of HTS technologies on in vitro neurogenesis, especially aiming at identifying the essential genes, chemical small molecules and adaptive microenvironments that hold great prospects for generating functional neurons or more reproductive and homogeneous 3D organoids. We also discuss the developmental tendency of HTS technology, e.g., so-called next-generation screening, which utilizes 3D organoid-based screening combined with microfluidic devices to narrow the gap between in vitro models and in vivo situations both physiologically and pathologically.
Collapse
Affiliation(s)
- Shu-Yuan Zhang
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Juan Zhao
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Jun-Jun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhen-Zhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
45
|
Rosario Isasi, Ginoza M, Jongsma K, Lars Assen, Morris Fabbri. Mending the gaps: ethically sensitive cells and the evolution of European stem cell policy. Regen Med 2022; 17:581-595. [PMID: 35670098 DOI: 10.2217/rme-2022-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The past decades witnessed the slow evolution of Europe's heterogeneous stem cell (SC) policy and substantial scientific advances in the field. Parallel to these developments, professional organizations have grown in influence. With the recently revised International Society for Stem Cell Research's Guidelines as a backdrop, we address the evolution of SC policies in 46 European countries and discuss how they fare against evolving ethical standards, societal views, and scientific advances. We identify areas of convergence, divergence, and the suitability of extant governance mechanisms to meet their stewardship roles. Europe represents a rich case study as it encompasses a wide range of policy approaches present worldwide. Comparative studies provide an opportunity to promote insight into national frameworks and to foster international harmonization.
Collapse
Affiliation(s)
- Rosario Isasi
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, Interdisciplinary Stem Cell Institute
| | - Margaret Ginoza
- University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | | | - Lars Assen
- University Medical Center Utrecht, Utrecht, NL 3584
| | - Morris Fabbri
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
46
|
Abstract
Embryoids and organoids hold great promise for human biology and medicine. Herein, we discuss conceptual and technological frameworks useful for developing high-fidelity embryoids and organoids that display tissue- and organ-level phenotypes and functions, which are critically needed for decoding developmental programs and improving translational applications. Through dissecting the layers of inputs controlling mammalian embryogenesis, we review recent progress in reconstructing multiscale structural orders in embryoids and organoids. Bioengineering tools useful for multiscale, multimodal structural engineering of tissue- and organ-level cellular organization and microenvironment are also discussed to present integrative, bioengineering-directed approaches to achieve next-generation, high-fidelity embryoids and organoids.
Collapse
Affiliation(s)
- Yue Shao
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China; State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
47
|
Romero-Morales AI, Gama V. Revealing the Impact of Mitochondrial Fitness During Early Neural Development Using Human Brain Organoids. Front Mol Neurosci 2022; 15:840265. [PMID: 35571368 PMCID: PMC9102998 DOI: 10.3389/fnmol.2022.840265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial homeostasis -including function, morphology, and inter-organelle communication- provides guidance to the intrinsic developmental programs of corticogenesis, while also being responsive to environmental and intercellular signals. Two- and three-dimensional platforms have become useful tools to interrogate the capacity of cells to generate neuronal and glia progeny in a background of metabolic dysregulation, but the mechanistic underpinnings underlying the role of mitochondria during human neurogenesis remain unexplored. Here we provide a concise overview of cortical development and the use of pluripotent stem cell models that have contributed to our understanding of mitochondrial and metabolic regulation of early human brain development. We finally discuss the effects of mitochondrial fitness dysregulation seen under stress conditions such as metabolic dysregulation, absence of developmental apoptosis, and hypoxia; and the avenues of research that can be explored with the use of brain organoids.
Collapse
Affiliation(s)
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
48
|
Piccolo FM, Kastan NR, Haremaki T, Tian Q, Laundos TL, De Santis R, Beaudoin AJ, Carroll TS, Luo JD, Gnedeva K, Etoc F, Hudspeth AJ, Brivanlou AH. Role of YAP in early ectodermal specification and a Huntington's Disease model of human neurulation. eLife 2022; 11:e73075. [PMID: 35451959 PMCID: PMC9033270 DOI: 10.7554/elife.73075] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
The Hippo pathway, a highly conserved signaling cascade that functions as an integrator of molecular signals and biophysical states, ultimately impinges upon the transcription coactivator Yes-associated protein 1 (YAP). Hippo-YAP signaling has been shown to play key roles both at the early embryonic stages of implantation and gastrulation, and later during neurogenesis. To explore YAP's potential role in neurulation, we used self-organizing neuruloids grown from human embryonic stem cells on micropatterned substrates. We identified YAP activation as a key lineage determinant, first between neuronal ectoderm and nonneuronal ectoderm, and later between epidermis and neural crest, indicating that YAP activity can enhance the effect of BMP4 stimulation and therefore affect ectodermal specification at this developmental stage. Because aberrant Hippo-YAP signaling has been implicated in the pathology of Huntington's Disease (HD), we used isogenic mutant neuruloids to explore the relationship between signaling and the disease. We found that HD neuruloids demonstrate ectopic activation of gene targets of YAP and that pharmacological reduction of YAP's transcriptional activity can partially rescue the HD phenotype.
Collapse
Affiliation(s)
- Francesco M Piccolo
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Nathaniel R Kastan
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
- Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew YorkUnited States
| | - Tomomi Haremaki
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Qingyun Tian
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Tiago L Laundos
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do PortoPortoPortugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do PortoPortoPortugal
- INEB - Instituto de Engenharia Biomédica, Universidade do PortoPortoPortugal
| | - Riccardo De Santis
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Andrew J Beaudoin
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Ji-Dung Luo
- Bioinformatics Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Ksenia Gnedeva
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
- Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew YorkUnited States
| | - Fred Etoc
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| | - AJ Hudspeth
- Howard Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
- Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew YorkUnited States
| | - Ali H Brivanlou
- Laboratory of of Stem Cell Biology and Molecular Embryology, The Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
49
|
Abstract
Developing organs are shaped, in part, by physical interaction with their environment in the embryo. In recent years, technical advances in live-cell imaging and material science have greatly expanded our understanding of the mechanical forces driving organ formation. Here, we provide a broad overview of the types of forces generated during embryonic development and then focus on a subset of organs underlying our senses: the eyes, inner ears, nose and skin. The epithelia in these organs emerge from a common origin: the ectoderm germ layer; yet, they arrive at unique and complex forms over developmental time. We discuss exciting recent animal studies that show a crucial role for mechanical forces in, for example, the thickening of sensory placodes, the coiling of the cochlea and the lengthening of hair. Finally, we discuss how microfabricated organoid systems can now provide unprecedented insights into the physical principles of human development.
Collapse
Affiliation(s)
- Anh Phuong Le
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Kim
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Karl R. Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
50
|
Lee JH, Shin H, Shaker MR, Kim HJ, Park SH, Kim JH, Lee N, Kang M, Cho S, Kwak TH, Kim JW, Song MR, Kwon SH, Han DW, Lee S, Choi SY, Rhyu IJ, Kim H, Geum D, Cho IJ, Sun W. Production of human spinal-cord organoids recapitulating neural-tube morphogenesis. Nat Biomed Eng 2022; 6:435-448. [PMID: 35347276 DOI: 10.1038/s41551-022-00868-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/15/2022] [Indexed: 12/12/2022]
Abstract
Human spinal-cord-like tissues induced from human pluripotent stem cells are typically insufficiently mature and do not mimic the morphological features of neurulation. Here, we report a three-dimensional culture system and protocol for the production of human spinal-cord-like organoids (hSCOs) recapitulating the neurulation-like tube-forming morphogenesis of the early spinal cord. The hSCOs exhibited neurulation-like tube-forming morphogenesis, cellular differentiation into the major types of spinal-cord neurons as well as glial cells, and mature synaptic functional activities, among other features of the development of the spinal cord. We used the hSCOs to screen for antiepileptic drugs that can cause neural-tube defects. hSCOs may also facilitate the study of the development of the human spinal cord and the modelling of diseases associated with neural-tube defects.
Collapse
Affiliation(s)
- Ju-Hyun Lee
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Mohammed R Shaker
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Kim
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Si-Hyung Park
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - June Hoan Kim
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Namwon Lee
- InterMinds Inc., Seongnam, Republic of Korea
| | - Minjin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Subin Cho
- Department of Bio-Information Science, Ewha Womans University, Seoul, Republic of Korea
| | - Tae Hwan Kwak
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jong Woon Kim
- Department of Obstetrics and Gynecology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Seung-Hae Kwon
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Dong Wook Han
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sanghyuk Lee
- Department of Bio-Information Science, Ewha Womans University, Seoul, Republic of Korea.,Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Im Joo Rhyu
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyun Kim
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Dongho Geum
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,School of Electrical and Electronics Engineering, Yonsei University, Seoul, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|