1
|
Fu M, Pang L, Wu Z, Wang M, Jin J, Ai S, Li X. Single-cell multi-omics delineates the dynamics of distinct epigenetic codes coordinating mouse gastrulation. BMC Genomics 2025; 26:454. [PMID: 40340740 PMCID: PMC12060302 DOI: 10.1186/s12864-025-11619-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
Gastrulation represents a crucial stage in embryonic development and is tightly controlled by a complex network involving epigenetic reprogramming. However, the molecular coordination among distinct epigenetic layers entailing the progressive restriction of lineage potency remains unclear. Here, we present a multi-omics map of H3K27ac and H3K4me1 single-cell ChIP-seq profiles of mouse embryos collected at six sequential time points. Significant epigenetic priming, as reflected by H3K27ac signals, is evident, yet asynchronous cell fate commitment of each germ layer at distinct histone modification levels are observed. Integrated scRNA-seq and single-cell ChIP-seq analysis unveil a "time lag" transition pattern between enhancer activation and gene expression during germ-layer specification. Notably, by utilizing the H3K27ac and H3K4me1 co-marked active enhancers, we construct a gene regulatory network centered on pivotal transcription factors, highlighting the potential critical role of Cdkn1c in mesoderm lineage specification. Together, our study broadens the current understanding of intricate epigenetic regulatory networks governing mouse gastrulation and sheds light on their relevance to congenital diseases.
Collapse
Affiliation(s)
- Mingzhu Fu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Long Pang
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhenwei Wu
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mei Wang
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Jin Jin
- Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| | - Shanshan Ai
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Department of Cardiology, Heart Center, First People's Hospital of Shunde, Southern Medical University, ShundeDistrict, Foshan, 528300, China.
| | - Xin Li
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
2
|
Stapornwongkul KS, Hahn E, Poliński P, Salamó Palau L, Arató K, Yao L, Williamson K, Gritti N, Anlas K, Osuna Lopez M, Patil KR, Heemskerk I, Ebisuya M, Trivedi V. Glycolytic activity instructs germ layer proportions through regulation of Nodal and Wnt signaling. Cell Stem Cell 2025; 32:744-758.e7. [PMID: 40245870 PMCID: PMC12048219 DOI: 10.1016/j.stem.2025.03.011] [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: 12/20/2023] [Revised: 10/29/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025]
Abstract
Metabolic pathways can influence cell fate decisions, yet their regulative role during embryonic development remains poorly understood. Here, we demonstrate an instructive role of glycolytic activity in regulating signaling pathways involved in mesoderm and endoderm specification. Using a mouse embryonic stem cell (mESC)-based in vitro model for gastrulation, we found that glycolysis inhibition increases ectodermal cell fates at the expense of mesodermal and endodermal lineages. We demonstrate that this relationship is dose dependent, enabling metabolic control of germ layer proportions through exogenous glucose levels. We further show that glycolysis acts as an upstream regulator of Nodal and Wnt signaling and that its influence on cell fate specification can be decoupled from its effects on growth. Finally, we confirm the generality of our findings using a human gastrulation model. Our work underscores the dependence of signaling pathways on metabolic conditions and provides mechanistic insight into the nutritional regulation of cell fate decision-making.
Collapse
Affiliation(s)
- Kristina S Stapornwongkul
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain.
| | - Elisa Hahn
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Patryk Poliński
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Laura Salamó Palau
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Krisztina Arató
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - LiAng Yao
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kate Williamson
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK
| | - Nicola Gritti
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | - Kerim Anlas
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
| | | | - Kiran R Patil
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK
| | - Idse Heemskerk
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Miki Ebisuya
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain; Cluster of Excellence Physics of Life, TU Dresden, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| | - Vikas Trivedi
- European Molecular Biology Laboratory, EMBL Barcelona, C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain; Developmental Biology, EMBL Heidelberg, Heidelberg 69117, Germany.
| |
Collapse
|
3
|
Sagha M. Neural induction: New insight into the default model and an extended four-step model in vertebrate embryos. Dev Dyn 2025. [PMID: 40105405 DOI: 10.1002/dvdy.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 03/20/2025] Open
Abstract
Neural induction is a process by which naïve ectodermal cells differentiate into neural progenitor cells through the inhibition of BMP signaling, a condition typically considered the "default" state in vertebrate embryos. Studies in vertebrate embryos indicate that active FGF/MAPK signaling reduces BMP signaling to facilitate neural induction. Consequently, I propose that FGF stimulation/BMP inhibition more accurately characterizes the default model. Initially, the neuroectoderm is instructed to differentiate into anterior forebrain tissue, with cranial signals stabilizing this outcome. Subsequently, a gradient of caudalizing signals converts the neuroectodermal cells into posterior midbrain, hindbrain, and spinal cord. Furthermore, at the caudal end of the embryo, neuromesodermal progenitor cells are destined to differentiate into both neural progenitor cells and mesodermal cells, aiding in body extension. In light of these observations, I suggest incorporating an additional step, elongation, into the conventional three-step model of neural induction. This updated model encompasses activation, stabilization, transformation, and elongation.
Collapse
Affiliation(s)
- Mohsen Sagha
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
4
|
Gharandouq MH, Ismail MA, Saleh T, Zihlif M, Ababneh NA. Metformin Protects Human Induced Pluripotent Stem Cell (hiPSC)-Derived Neurons from Oxidative Damage Through Antioxidant Mechanisms. Neurotox Res 2025; 43:15. [PMID: 40100475 DOI: 10.1007/s12640-025-00734-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 02/18/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
The antidiabetic drug metformin possesses antioxidant and cell protective effects including in neuronal cells, suggesting its potential use for treating neurodegenerative diseases. This study aimed to assess metformin's effects on viability and antioxidant activity in human-induced pluripotent stem cell (hiPSC)-derived neurons under varying concentrations and stress conditions. Six lines of hiPSC-derived neuronal progenitors derived from healthy human iPSCs were treated with metformin (1-500 µM) on day 18 of differentiation. For mature neurons (day 30), three concentrations (10 µM, 50 µM, and 100 µM) were used to assess cytotoxicity. MG132 proteasomal inhibitor and sodium arsenite (NaArs) were used to investigate oxidative stress, and 50 µM of metformin was tested for its protective effects against oxidative stress in hiPSC-derived neurons. Metformin treatment did not affect cell viability, neuronal differentiation, or trigger reactive oxygen species (ROS) generation in healthy hiPSC-derived motor neurons. Additionally, mitochondrial membrane potential (MMP) loss was not observed at 50 µM metformin. Metformin effectively protected neurons from stress agents and elevated the expression of antioxidant genes when treated with MG132. However, an interplay between MG132 and metformin resulted in lower expression of Nrf2 and NQO1 compared to the MG132 group alone, indicating reduced JC-1 aggregate levels due to MG132 proteasomal inhibition. Metformin upregulated antioxidant genes in hiPSC-derived neurons under stress conditions and protected the cells from oxidative damage.
Collapse
Affiliation(s)
- Mohammad H Gharandouq
- Faculty of Biological Sciences, The University of Jordan, Amman, Jordan
- Cell Therapy Center, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
| | - Mohammad A Ismail
- Cell Therapy Center, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan
- South Australian ImmunoGENomics Cancer Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
- Department of Pharmacology & Therapeutics, College of Medicine & Health Sciences, Arabian Gulf University, Manama, Bahrain
| | - Malik Zihlif
- Department of Pharmaceutical Sciences, School of Medicine, University of Jordan, Amman, Jordan
| | - Nidaa A Ababneh
- Cell Therapy Center, The University of Jordan, Queen Rania Street, Amman, 11942, Jordan.
| |
Collapse
|
5
|
Peinado P, Stazi M, Ballabio C, Margineanu MB, Li Z, Colón CI, Hsieh MS, Pal Choudhuri S, Stastny V, Hamilton S, Le Marois A, Collingridge J, Conrad L, Chen Y, Ng SR, Magendantz M, Bhutkar A, Chen JS, Sahai E, Drapkin BJ, Jacks T, Vander Heiden MG, Kopanitsa MV, Robinson HPC, Li L. Intrinsic electrical activity drives small-cell lung cancer progression. Nature 2025; 639:765-775. [PMID: 39939778 PMCID: PMC11922742 DOI: 10.1038/s41586-024-08575-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/23/2024] [Indexed: 02/14/2025]
Abstract
Elevated or ectopic expression of neuronal receptors promotes tumour progression in many cancer types1,2; neuroendocrine (NE) transformation of adenocarcinomas has also been associated with increased aggressiveness3. Whether the defining neuronal feature, namely electrical excitability, exists in cancer cells and impacts cancer progression remains mostly unexplored. Small-cell lung cancer (SCLC) is an archetypal example of a highly aggressive NE cancer and comprises two major distinct subpopulations: NE cells and non-NE cells4,5. Here we show that NE cells, but not non-NE cells, are excitable, and their action potential firing directly promotes SCLC malignancy. However, the resultant high ATP demand leads to an unusual dependency on oxidative phosphorylation in NE cells. This finding contrasts with the properties of most cancer cells reported in the literature, which are non-excitable and rely heavily on aerobic glycolysis. Additionally, we found that non-NE cells metabolically support NE cells, a process akin to the astrocyte-neuron metabolite shuttle6. Finally, we observed drastic changes in the innervation landscape during SCLC progression, which coincided with increased intratumoural heterogeneity and elevated neuronal features in SCLC cells, suggesting an induction of a tumour-autonomous vicious cycle, driven by cancer cell-intrinsic electrical activity, which confers long-term tumorigenic capability and metastatic potential.
Collapse
Affiliation(s)
- Paola Peinado
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
| | - Marco Stazi
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
| | - Claudio Ballabio
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
| | | | - Zhaoqi Li
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caterina I Colón
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shreoshi Pal Choudhuri
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Victor Stastny
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Seth Hamilton
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Alix Le Marois
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Jodie Collingridge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Linus Conrad
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Yinxing Chen
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sheng Rong Ng
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Margaret Magendantz
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arjun Bhutkar
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jin-Shing Chen
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Benjamin J Drapkin
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - Tyler Jacks
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maksym V Kopanitsa
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
- Charles River Discovery Services, Portishead, UK
| | - Hugh P C Robinson
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Leanne Li
- Cancer Neuroscience Laboratory, Francis Crick Institute, London, UK.
- Koch Institute of Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
6
|
Bergamasco MI, Abeysekera W, Garnham AL, Hu Y, Li-Wai-Suen CS, Sheikh BN, Smyth GK, Thomas T, Voss AK. KAT6B is required for histone 3 lysine 9 acetylation and SOX gene expression in the developing brain. Life Sci Alliance 2025; 8:e202402969. [PMID: 39537341 PMCID: PMC11561263 DOI: 10.26508/lsa.202402969] [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: 07/31/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Heterozygous mutations in the histone lysine acetyltransferase gene KAT6B (MYST4/MORF/QKF) underlie neurodevelopmental disorders, but the mechanistic roles of KAT6B remain poorly understood. Here, we show that loss of KAT6B in embryonic neural stem and progenitor cells (NSPCs) impaired cell proliferation, neuronal differentiation, and neurite outgrowth. Mechanistically, loss of KAT6B resulted in reduced acetylation at histone H3 lysine 9 and reduced expression of key nervous system development genes in NSPCs and the developing cortex, including the SOX gene family, in particular Sox2, which is a key driver of neural progenitor proliferation, multipotency and brain development. In the fetal cortex, KAT6B occupied the Sox2 locus. Loss of KAT6B caused a reduction in Sox2 promoter activity in NSPCs. Sox2 overexpression partially rescued the proliferative defect of Kat6b -/- NSPCs. Collectively, these results elucidate molecular requirements for KAT6B in brain development and identify key KAT6B targets in neural precursor cells and the developing brain.
Collapse
Affiliation(s)
- Maria I Bergamasco
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Waruni Abeysekera
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Yifang Hu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Connie Sn Li-Wai-Suen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Bilal N Sheikh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- School of Mathematics and Statistics, University of Melbourne, Parkville, Australia
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
7
|
Rabeling A, van der Hoven A, Andersen N, Goolam M. Neural Tube Organoids: A Novel System to Study Developmental Timing. Stem Cell Rev Rep 2024; 20:2045-2061. [PMID: 39230820 PMCID: PMC11554929 DOI: 10.1007/s12015-024-10785-5] [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] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
The neural tube (NT) is a transient structure formed during embryogenesis which develops into the brain and spinal cord. While mouse models have been commonly used in place of human embryos to study NT development, species-specific differences limit their applicability. One major difference is developmental timing, with NT formation from the neural plate in 16 days in humans compared to 4 days in mice, as well as differences in the time taken to form neuronal subtypes and complete neurogenesis. Neural tube organoids (NTOs) represent a new way to study NT development in vitro. While mouse and human NTOs have been shown to recapitulate the major developmental events of NT formation; it is unknown whether species-specific developmental timing, also termed allochrony, is also recapitulated. This review summarises current research using both mouse and human NTOs and compares developmental timing events in order to assess if allochrony is maintained in organoids.
Collapse
Affiliation(s)
- Alexa Rabeling
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Amy van der Hoven
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Nathalie Andersen
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Mubeen Goolam
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa.
- UCT Neuroscience Institute, Cape Town, South Africa.
| |
Collapse
|
8
|
Kuribayashi H, Iwagawa T, Murakami A, Kawamura T, Suzuki Y, Watanabe S. NMNAT1 Is Essential for Human iPS Cell Differentiation to the Retinal Lineage. Invest Ophthalmol Vis Sci 2024; 65:37. [PMID: 39446354 PMCID: PMC11512567 DOI: 10.1167/iovs.65.12.37] [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: 12/20/2023] [Accepted: 09/27/2024] [Indexed: 10/27/2024] Open
Abstract
Purpose The gene encoding nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1), a nicotinamide adenine dinucleotide synthetase localized in the cell nucleus, is a causative factor in Leber's congenital amaurosis, which is the earliest onset type of inherited retinal degeneration. We sought to investigate the roles of NMNAT1 in early retinal development. Methods We used human induced pluripotent stem cells (hiPSCs) and established NMNAT1-knockout (KO) hiPSCs using CRISPR/cas9 technology to reveal the roles of NMNAT1 in human retinal development. Results NMNAT1 was not essential for the survival and proliferation of immature hiPSCs; therefore, we subjected NMNAT1-KO hiPSCs to retinal organoid (RO) differentiation culture. The expression levels of immature hiPSC-specific genes decreased in a similar manner after organoid culture initiation up to 2 weeks in the control and NMNAT1-KO. Neuroectoderm-specific genes were induced in the control and NMNAT1-KO organoids within a few days after starting the organoid culture; PAX6 and TUBB3 were higher in NMNAT1-KO organoids up to 7 days than in the control organoids. However, the induction of genes involving retinal early development, such as RAX, which was induced at around day 10 in this culture, was considerably reduced in NMNAT1-KO organoids. Morphological examination also showed failure of retinal primordial structure formation, which became visible at around 2 weeks of the control culture, in the NMNAT1-KO organoids. Decreased intracellular NAD levels and poly(ADP-ribosyl)ation were observed in NMNAT1-KO organoids at 7 to 10 days of the culture. Mass spectrometry analysis of inhibited proteins in the poly(ADP-ribosyl)ation pathway identified poly(ADP-ribosyl)ation of poly(ADP-ribose) polymerase 1 (PARP1) as a major protein. Conclusions These results indicate that NMNAT1 was dispensable for neural lineage differentiation but essential for the commitment of retinal fate differentiation in hiPSCs. The NMNAT1-NAD-PARP1 axis may play a critical role in the appropriate development of human retinal lineage differentiation.
Collapse
Affiliation(s)
- Hiroshi Kuribayashi
- Department of Retinal Development and Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Toshiro Iwagawa
- Department of Retinal Development and Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Akira Murakami
- Department of Ophthalmology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Takeshi Kawamura
- Isotope Science Center, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Science, The University of Tokyo, Bunkyo-ku, Chiba, Japan
| | - Sumiko Watanabe
- Department of Retinal Development and Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
9
|
Niharika, Ureka L, Roy A, Patra SK. Dissecting SOX2 expression and function reveals an association with multiple signaling pathways during embryonic development and in cancer progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189136. [PMID: 38880162 DOI: 10.1016/j.bbcan.2024.189136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
SRY (Sex Determining Region) box 2 (SOX2) is an essential transcription factor that plays crucial roles in activating genes involved in pre- and post-embryonic development, adult tissue homeostasis, and lineage specifications. SOX2 maintains the self-renewal property of stem cells and is involved in the generation of induced pluripotency stem cells. SOX2 protein contains a particular high-mobility group domain that enables SOX2 to achieve the capacity to participate in a broad variety of functions. The information about the involvement of SOX2 with gene regulatory elements, signaling networks, and microRNA is gradually emerging, and the higher expression of SOX2 is functionally relevant to various cancer types. SOX2 facilitates the oncogenic phenotype via cellular proliferation and enhancement of invasive tumor properties. Evidence are accumulating in favor of three dimensional (higher order) folding of chromatin and epigenetic control of the SOX2 gene by chromatin modifications, which implies that the expression level of SOX2 can be modulated by epigenetic regulatory mechanisms, specifically, via DNA methylation and histone H3 modification. In view of this, and to focus further insights into the roles SOX2 plays in physiological functions, involvement of SOX2 during development, precisely, the advances of our knowledge in pre- and post-embryonic development, and interactions of SOX2 in this scenario with various signaling pathways in tumor development and cancer progression, its potential as a therapeutic target against many cancers are summarized and discussed in this article.
Collapse
Affiliation(s)
- Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Lina Ureka
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India.
| |
Collapse
|
10
|
Berasain L, Beati P, Trigila AP, Rubinstein M, Franchini LF. Accelerated evolution in the human lineage led to gain and loss of transcriptional enhancers in the RBFOX1 locus. SCIENCE ADVANCES 2024; 10:eadl1049. [PMID: 38924416 PMCID: PMC11204294 DOI: 10.1126/sciadv.adl1049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
A long-standing goal of evolutionary biology is to decode how changes in gene regulatory networks contribute to human-specific traits. Human accelerated regions (HARs) are prime candidates for driving gene regulatory modifications in human development. The RBFOX1 locus is densely populated with HARs, providing a set of potential regulatory elements that could have changed its expression in the human lineage. Here, we examined the role of RBFOX1-HARs using transgenic zebrafish reporter assays and identified 15 transcriptional enhancers that are active in the developing nervous system, 9 of which displayed differential activity between the human and chimpanzee sequences. The engineered loss of two selected RBFOX1-HARs in knockout mouse models modified Rbfox1 expression at specific developmental stages and tissues in the brain, influencing the expression and splicing of a high number of Rbfox1 target genes. Our results provided insight into the spatial and temporal changes in gene expression driven by RBFOX1-HARs.
Collapse
Affiliation(s)
- Lara Berasain
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) “Dr. Hector N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
| | - Paula Beati
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) “Dr. Hector N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
| | - Anabella P. Trigila
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) “Dr. Hector N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) “Dr. Hector N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1428, Argentina
| | - Lucía F. Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) “Dr. Hector N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina
| |
Collapse
|
11
|
Bitarafan S, Pybus AF, Rivera Moctezuma FG, Adibi M, Franklin TC, Singer AC, Wood LB. Frequency and duration of sensory flicker controls astrocyte and neuron specific transcriptional profiles in 5xFAD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.594705. [PMID: 38826251 PMCID: PMC11142106 DOI: 10.1101/2024.05.20.594705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Current clinical trials are investigating gamma frequency sensory stimulation as a potential therapeutic strategy for Alzheimer's disease, yet we lack a comprehensive picture of the effects of this stimulation on multiple aspects of brain function. While most prior research has focused on gamma frequency sensory stimulation, we previously showed that exposing mice to visual flickering stimulation increased MAPK and NFκB signaling in the visual cortex in a manner dependent on duration and frequency of sensory stimulation exposure. Because these pathways control multiple neuronal and glial functions and are differentially activated based on the duration and frequency of flicker stimulation, we aimed to define the transcriptional effects of different frequencies and durations of flicker stimulation on multiple brain functions. Methods We exposed 5xFAD mice to different frequencies of audio/visual flicker stimulation (constant light, 10Hz, 20Hz, 40Hz) for durations of 0.5hr, 1hr, or 4hr, then used bulk RNAseq to profile transcriptional changes within the visual cortex and hippocampus tissues. Using weighted gene co-expression network analysis, we identified modules of co-expressed genes controlled by frequency and/or duration of stimulation. Results Within the visual cortex, we found that all stimulation frequencies caused fast activation of a module of immune genes within 1hr and slower suppression of synaptic genes after 4hrs of stimulation. Interestingly, all frequencies of stimulation led to slow suppression of astrocyte specific gene sets, while activation of neuronal gene sets was frequency and duration specific. In contrast, in the hippocampus, immune and synaptic modules were suppressed based on the frequency of stimulation. Specifically,10Hz activated a module of genes associated with mitochondrial function, metabolism, and synaptic translation while 10Hz rapidly suppressed a module of genes linked to neurotransmitter activity. Conclusion Collectively, our data indicate that the frequency and duration of flicker stimulation controls immune, neuronal, and metabolic genes in multiple regions of the brain affected by Alzheimer's disease. Flicker stimulation may thus represent a potential therapeutic strategy that can be tuned based on the brain region and the specific cellular process to be modulated.
Collapse
|
12
|
Marelli E, Hughes J, Scotting PJ. SUMO-dependent transcriptional repression by Sox2 inhibits the proliferation of neural stem cells. PLoS One 2024; 19:e0298818. [PMID: 38507426 PMCID: PMC10954124 DOI: 10.1371/journal.pone.0298818] [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: 04/20/2023] [Accepted: 01/30/2024] [Indexed: 03/22/2024] Open
Abstract
Sox2 is known for its roles in maintaining the stem cell state of embryonic stem cells and neural stem cells. In particular, it has been shown to slow the proliferation of these cell types. It is also known for its effects as an activating transcription factor. Despite this, analysis of published studies shows that it represses as many genes as it activates. Here, we identify a new set of target genes that Sox2 represses in neural stem cells. These genes are associated with centrosomes, centromeres and other aspects of cell cycle control. In addition, we show that SUMOylation of Sox2 is necessary for the repression of these genes and for its repressive effects on cell proliferation. Together, these data suggest that SUMO-dependent repression of this group of target genes is responsible for the role of Sox2 in regulating the proliferation of neural stem cells.
Collapse
Affiliation(s)
- Elisa Marelli
- School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| | - Jaime Hughes
- School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| | - Paul J. Scotting
- School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| |
Collapse
|
13
|
Chen Q, Leshkowitz D, Li H, van Impel A, Schulte-Merker S, Amit I, Rizzoti K, Levkowitz G. Neural plate progenitors give rise to both anterior and posterior pituitary cells. Dev Cell 2023; 58:2652-2665.e6. [PMID: 37683631 DOI: 10.1016/j.devcel.2023.08.018] [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: 08/10/2022] [Revised: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023]
Abstract
The pituitary is the master neuroendocrine gland, which regulates body homeostasis. It consists of the anterior pituitary/adenohypophysis harboring hormones producing cells and the posterior pituitary/neurohypophysis, which relays the passage of hormones from the brain to the periphery. It is accepted that the adenohypophysis originates from the oral ectoderm (Rathke's pouch), whereas the neural ectoderm contributes to the neurohypophysis. Single-cell transcriptomics of the zebrafish pituitary showed that cyp26b1-positive astroglial pituicytes of the neurohypophysis and prop1-positive adenohypophyseal progenitors expressed common markers implying lineage relatedness. Genetic tracing identifies that, in contrast to the prevailing dogma, neural plate precursors of zebrafish (her4.3+) and mouse (Sox1+) contribute to both neurohypophyseal and a subset of adenohypophyseal cells. Pituicyte-derived retinoic-acid-degrading enzyme Cyp26b1 fine-tunes differentiation of prop1+ progenitors into hormone-producing cells. These results challenge the notion that adenohypophyseal cells are exclusively derived from non-neural ectoderm and demonstrate that crosstalk between neuro- and adeno-hypophyseal cells affects differentiation of pituitary cells.
Collapse
Affiliation(s)
- Qiyu Chen
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dena Leshkowitz
- Life Science Core Facilities, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Hanjie Li
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Present address: CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Andreas van Impel
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, UK
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel.
| |
Collapse
|
14
|
Songsaad AT, Thairat S, Seemaung P, Thongsuk A, Balit T, Ruangsawasdi N, Phruksaniyom C, Gonmanee T, White KL, Thonabulsombat C. Characterization of neural stem cells derived from human stem cells from the apical papilla undergoing three-dimensional neurosphere induction. J Appl Oral Sci 2023; 31:e20230209. [PMID: 37970885 PMCID: PMC10697670 DOI: 10.1590/1678-7757-2023-0209] [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: 06/09/2023] [Revised: 09/03/2023] [Accepted: 09/29/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVES The endogenous repairing based on the activation of neural stem cells (NSCs) is impaired by neurodegenerative diseases. The present study aims to characterize human stem cells from the apical papilla (hSCAPs) with features of mesenchymal stem cells (MSCs) and to demonstrate the neuronal differentiation of hSCAPs into NSCs through the formation of three-dimensional (3D) neurospheres, verifying the structural, immunophenotyping, self-renewal, gene expression and neuronal activities of these cells to help further improve NSCs transplantation. METHODOLOGY The hSCAPs were isolated from healthy impacted human third molar teeth and characterized as MSCs. They were then induced into 3D-neurospheres using a specific neural induction medium. Subsequently, the intra-neurospheral cells were confirmed to be NSCs by the identification of Nissl substance and the analysis of immunofluorescence staining, self-renewal ability, and gene expression of the cells. Moreover, the neuronal activity was investigated using intracellular calcium oscillation. RESULTS The isolated cells from the human apical papilla expressed many markers of MSCs, such as self-renewal ability and multilineage differentiation. These cells were thus characterized as MSCs, specifically as hSCAPs. The neurospheres induced from hSCAPs exhibited a 3D-floating spheroidal shape and larger neurospheres, and consisted of a heterogeneous population of intra-neurospheral cells. Further investigation showed that these intra-neurospheral cells had Nissl body staining and also expressed both Nestin and SOX2. They presented a self-renewal ability as well, which was observed after their disaggregation. Their gene expression profiling also exhibited a significant amount of NSC markers (NES, SOX1, and PAX6). Lastly, a large and dynamic change of the fluorescent signal that indicated calcium ions (Ca2+) was detected in the intracellular calcium oscillation, which indicated the neuronal activity of NSCs-derived hSCAPs. CONCLUSIONS The hSCAPs exhibited properties of MSCs and could differentiate into NSCs under 3D-neurosphere generation. The present findings suggest that NSCs-derived hSCAPs may be used as an alternative candidates for cell-based therapy, which uses stem cell transplantation to further treat neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Sarut Thairat
- Mahidol University, Faculty of Dentistry, Oral Tissues, Cells and Molecular Biology Analysis and Research Center, Bangkok, Thailand
| | - Peeratchai Seemaung
- Mahidol University, Faculty of Science, Department of Anatomy, Bangkok, Thailand
| | - Amarin Thongsuk
- Mahidol University, Faculty of Science, Department of Anatomy, Bangkok, Thailand
| | - Tatcha Balit
- Mahidol University, Faculty of Science, Department of Anatomy, Bangkok, Thailand
| | - Nisarat Ruangsawasdi
- Mahidol University, Faculty of Dentistry, Department of Pharmacology, Bangkok, Thailand
| | | | - Thanasup Gonmanee
- Mahidol University, Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Samut Prakan, Thailand
| | - Kenneth L White
- Utah State University, College of Agriculture and Applied Sciences, Department of Animal, Dairy, and Veterinary Sciences, Utah, The United States of America
| | | |
Collapse
|
15
|
Xu T, Cao L, Duan J, Li Y, Li Y, Hu Z, Li S, Zhang M, Wang G, Guo F, Lu J. Uncovering the role of FOXA2 in the Development of Human Serotonin Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303884. [PMID: 37679064 PMCID: PMC10646255 DOI: 10.1002/advs.202303884] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/08/2023] [Indexed: 09/09/2023]
Abstract
Directed differentiation of serotonin neurons (SNs) from human pluripotent stem cells (hPSCs) provides a valuable tool for uncovering the mechanism of human SN development and the associated neuropsychiatric disorders. Previous studies report that FOXA2 is expressed by serotonergic progenitors (SNPs) and functioned as a serotonergic fate determinant in mouse. However, in the routine differentiation experiments, it is accidentally found that less SNs and more non-neuronal cells are obtained from SNP stage with higher percentage of FOXA2-positive cells. This phenomenon prompted them to question the role of FOXA2 as an intrinsic fate determinant for human SN differentiation. Herein, by direct differentiation of engineered hPSCs into SNs, it is found that the SNs are not derived from FOXA2-lineage cells; FOXA2-knockout hPSCs can still differentiate into mature and functional SNs with typical serotonergic identity; FOXA2 overexpression suppresses the SN differentiation, indicating that FOXA2 is not intrinsically required for human SN differentiation. Furthermore, repressing FOXA2 expression by retinoic acid (RA) and dynamically modulating Sonic Hedgehog (SHH) signaling pathway promotes human SN differentiation. This study uncovers the role of FOXA2 in human SN development and improves the differentiation efficiency of hPSCs into SNs by repressing FOXA2 expression.
Collapse
Affiliation(s)
- Ting Xu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Jinjin Duan
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yingqi Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - You Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Zhangsen Hu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Shuanqing Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Meihui Zhang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Guanhao Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Fei Guo
- Key Laboratory of Receptor ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
- Suzhou Institute of Tongji UniversitySuzhou215101China
| |
Collapse
|
16
|
Oh SY, Na SB, Kang YK, Do JT. In Vitro Embryogenesis and Gastrulation Using Stem Cells in Mice and Humans. Int J Mol Sci 2023; 24:13655. [PMID: 37686459 PMCID: PMC10563085 DOI: 10.3390/ijms241713655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
During early mammalian embryonic development, fertilized one-cell embryos develop into pre-implantation blastocysts and subsequently establish three germ layers through gastrulation during post-implantation development. In recent years, stem cells have emerged as a powerful tool to study embryogenesis and gastrulation without the need for eggs, allowing for the generation of embryo-like structures known as synthetic embryos or embryoids. These in vitro models closely resemble early embryos in terms of morphology and gene expression and provide a faithful recapitulation of early pre- and post-implantation embryonic development. Synthetic embryos can be generated through a combinatorial culture of three blastocyst-derived stem cell types, such as embryonic stem cells, trophoblast stem cells, and extraembryonic endoderm cells, or totipotent-like stem cells alone. This review provides an overview of the progress and various approaches in studying in vitro embryogenesis and gastrulation in mice and humans using stem cells. Furthermore, recent findings and breakthroughs in synthetic embryos and gastruloids are outlined. Despite ethical considerations, synthetic embryo models hold promise for understanding mammalian (including humans) embryonic development and have potential implications for regenerative medicine and developmental research.
Collapse
Affiliation(s)
| | | | | | - Jeong Tae Do
- Department of Stem Cell Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Republic of Korea; (S.Y.O.); (S.B.N.); (Y.K.K.)
| |
Collapse
|
17
|
Suppinger S, Zinner M, Aizarani N, Lukonin I, Ortiz R, Azzi C, Stadler MB, Vianello S, Palla G, Kohler H, Mayran A, Lutolf MP, Liberali P. Multimodal characterization of murine gastruloid development. Cell Stem Cell 2023; 30:867-884.e11. [PMID: 37209681 PMCID: PMC10241222 DOI: 10.1016/j.stem.2023.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/16/2023] [Accepted: 04/25/2023] [Indexed: 05/22/2023]
Abstract
Gastruloids are 3D structures generated from pluripotent stem cells recapitulating fundamental principles of embryonic pattern formation. Using single-cell genomic analysis, we provide a resource mapping cell states and types during gastruloid development and compare them with the in vivo embryo. We developed a high-throughput handling and imaging pipeline to spatially monitor symmetry breaking during gastruloid development and report an early spatial variability in pluripotency determining a binary response to Wnt activation. Although cells in the gastruloid-core revert to pluripotency, peripheral cells become primitive streak-like. These two populations subsequently break radial symmetry and initiate axial elongation. By performing a compound screen, perturbing thousands of gastruloids, we derive a phenotypic landscape and infer networks of genetic interactions. Finally, using a dual Wnt modulation, we improve the formation of anterior structures in the existing gastruloid model. This work provides a resource to understand how gastruloids develop and generate complex patterns in vitro.
Collapse
Affiliation(s)
- Simon Suppinger
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; University of Basel, 4001 Basel, Switzerland
| | - Marietta Zinner
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland
| | - Nadim Aizarani
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland
| | - Ilya Lukonin
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; Roche Institute of Human Biology, 4058 Basel, Switzerland
| | - Raphael Ortiz
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland
| | - Chiara Azzi
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; Babraham Institute, Cambridge CB22 3AT, UK
| | - Michael B Stadler
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; University of Basel, 4001 Basel, Switzerland; Swiss Institute of Bioinformatics, 4058 Basel, Switzerland
| | - Stefano Vianello
- School of Life Sciences, Federal Institute of Technology EPFL, 1015 Lausanne, Switzerland
| | - Giovanni Palla
- Institute of Computational Biology, Helmholtz Center Munich, 85764 Munich, Germany; TUM School of Life Sciences Weihenstephan, Technical University of Munich, 80333 Munich, Germany
| | - Hubertus Kohler
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland
| | - Alexandre Mayran
- School of Life Sciences, Federal Institute of Technology EPFL, 1015 Lausanne, Switzerland
| | - Matthias P Lutolf
- Roche Institute of Human Biology, 4058 Basel, Switzerland; School of Life Sciences, Federal Institute of Technology EPFL, 1015 Lausanne, Switzerland
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; University of Basel, 4001 Basel, Switzerland.
| |
Collapse
|
18
|
Osorio MJ, Mariani JN, Zou L, Schanz SJ, Heffernan K, Cornwell A, Goldman SA. Glial progenitor cells of the adult human white and grey matter are contextually distinct. Glia 2023; 71:524-540. [PMID: 36334067 PMCID: PMC10100527 DOI: 10.1002/glia.24291] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
Genomic analyses have revealed heterogeneity among glial progenitor cells (GPCs), but the compartment selectivity of human GPCs (hGPCs) is unclear. Here, we asked if GPCs of human grey and white brain matter are distinct in their architecture and associated gene expression. RNA profiling of NG2-defined hGPCs derived from adult human neocortex and white matter differed in their expression of genes involved in Wnt, NOTCH, BMP and TGFβ signaling, suggesting compartment-selective biases in fate and self-renewal. White matter hGPCs over-expressed the BMP antagonists BAMBI and CHRDL1, suggesting their tonic suppression of astrocytic fate relative to cortical hGPCs, whose relative enrichment of cytoskeletal genes presaged their greater morphological complexity. In human glial chimeric mice, cortical hGPCs assumed larger and more complex morphologies than white matter hGPCs, and both were more complex than their mouse counterparts. These findings suggest that human grey and white matter GPCs comprise context-specific pools with distinct functional biases.
Collapse
Affiliation(s)
- Maria Joana Osorio
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA.,Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - John N Mariani
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Lisa Zou
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Steven J Schanz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Kate Heffernan
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Adam Cornwell
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA.,Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Anand GM, Megale HC, Murphy SH, Weis T, Lin Z, He Y, Wang X, Liu J, Ramanathan S. Controlling organoid symmetry breaking uncovers an excitable system underlying human axial elongation. Cell 2023; 186:497-512.e23. [PMID: 36657443 PMCID: PMC10122509 DOI: 10.1016/j.cell.2022.12.043] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/28/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023]
Abstract
The human embryo breaks symmetry to form the anterior-posterior axis of the body. As the embryo elongates along this axis, progenitors in the tail bud give rise to tissues that generate spinal cord, skeleton, and musculature. This raises the question of how the embryo achieves axial elongation and patterning. While ethics necessitate in vitro studies, the variability of organoid systems has hindered mechanistic insights. Here, we developed a bioengineering and machine learning framework that optimizes organoid symmetry breaking by tuning their spatial coupling. This framework enabled reproducible generation of axially elongating organoids, each possessing a tail bud and neural tube. We discovered that an excitable system composed of WNT/FGF signaling drives elongation by inducing a neuromesodermal progenitor-like signaling center. We discovered that instabilities in the excitable system are suppressed by secreted WNT inhibitors. Absence of these inhibitors led to ectopic tail buds and branches. Our results identify mechanisms governing stable human axial elongation.
Collapse
Affiliation(s)
- Giridhar M Anand
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Heitor C Megale
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sean H Murphy
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Theresa Weis
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Zuwan Lin
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA
| | - Yichun He
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02138, USA
| | - Jia Liu
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Sharad Ramanathan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
20
|
Wijnand A, Verhelst H. Anti-SOX1 Antibodies in a 3-Year-old Girl, Post-Varicella. Child Neurol Open 2023; 10:2329048X231200613. [PMID: 37745634 PMCID: PMC10517599 DOI: 10.1177/2329048x231200613] [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: 04/25/2023] [Revised: 08/04/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Anti-SRY-related HMG-box gene 1 (SOX1) antibodies were initially described in adults with paraneoplastic neurological disorders, where they are considered high-risk onconeural autoantibodies. Only two pediatric cases of anti-SOX1 antibodies have been reported: a 17-year-old adolescent presenting with paraneoplastic limbic encephalitis due to Hodgkin lymphoma and a 12-year-old girl presenting with non-paraneoplastic encephalitis. We present a unique case of anti-SOX1 antibodies in a 3-year-old girl, post-varicella infection. Initially, she presented with ataxia and dysmetria, with subsequent reports from parents of urinary incontinence and significant behavior changes. Additionally, reflexes in the lower limbs were absent. Anti-SOX1 antibodies tested positive in both serum and cerebrospinal fluid. Oncological screening at presentation and a seven-month follow-up showed no malignancies. The patient exhibited favorable clinical progress without requiring treatment. At the seven-month follow-up, serum antibodies tested negative. This case report broadens the known clinical spectrum, being the first description of post-varicella anti-SOX1 antibodies.
Collapse
Affiliation(s)
- Aline Wijnand
- Department of Child and Adolescent Psychiatry, Ghent University Hospital, Ghent, Belgium
| | - Helene Verhelst
- Department of Pediatrics, Division of Pediatric Neurology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
21
|
The Effects of Co-Culture of Embryonic Stem Cells with Neural Stem Cells on Differentiation. Curr Issues Mol Biol 2022; 44:6104-6116. [PMID: 36547077 PMCID: PMC9776753 DOI: 10.3390/cimb44120416] [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: 11/21/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Researching the technology for in vitro differentiation of embryonic stem cells (ESCs) into neural lineages is very important in developmental biology, regenerative medicine, and cell therapy. Thus, studies on in vitro differentiation of ESCs into neural lineages by co-culture are expected to improve our understanding of this process. A co-culture system has long been used to study interactions between cell populations, improve culture efficiency, and establish synthetic interactions between populations. In this study, we investigated the effect of a co-culture of ESCs with neural stem cells (NSCs) in two-dimensional (2D) or three-dimensional (3D) culture conditions. Furthermore, we examined the effect of an NSC-derived conditioned medium (CM) on ESC differentiation. OG2-ESCs lost the specific morphology of colonies and Oct4-GFP when co-cultured with NSC. Additionally, real-time PCR analysis showed that ESCs co-cultured with NSCs expressed higher levels of ectoderm markers Pax6 and Sox1 under both co-culture conditions. However, the differentiation efficiency of CM was lower than that of the non-conditioned medium. Collectively, our results show that co-culture with NSCs promotes the differentiation of ESCs into the ectoderm.
Collapse
|
22
|
Carvalho MS, Nogueira DE, Cabral JM, Rodrigues CA. Neural progenitor cell-derived extracellular matrix as a new platform for neural differentiation of human induced pluripotent stem cells. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100070. [PMID: 36824374 PMCID: PMC9934470 DOI: 10.1016/j.bbiosy.2022.100070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
The culture microenvironment has been demonstrated to regulate stem cell fate and to be a crucial aspect for quality-controlled stem cell maintenance and differentiation to a specific lineage. In this context, extracellular matrix (ECM) proteins are particularly important to mediate the interactions between the cells and the culture substrate. Human induced pluripotent stem cells (hiPSCs) are usually cultured as anchorage-dependent cells and require adhesion to an ECM substrate to support their survival and proliferation in vitro. Matrigel, a common substrate for hiPSC culture is a complex and undefined mixture of ECM proteins which are expensive and not well suited to clinical application. Decellularized cell-derived ECM has been shown to be a promising alternative to the common protein coatings used in stem cell culture. However, very few studies have used this approach as a niche for neural differentiation of hiPSCs. Here, we developed a new stem cell culture system based on decellularized cell-derived ECM from neural progenitor cells (NPCs) for expansion and neural differentiation of hiPSCs, as an alternative to Matrigel and poly-l-ornithine/laminin-coated well plates. Interestingly, hiPSCs were able to grow and maintain their pluripotency when cultured on decellularized ECM from NPCs (NPC ECM). Furthermore, NPC ECM enhanced the neural differentiation of hiPSCs compared to poly-l-ornithine/laminin-coated wells, which are used in most neural differentiation protocols, presenting a statistically significant enhancement of neural gene expression markers, such as βIII-Tubulin and MAP2. Taken together, our results demonstrate that NPC ECM provides a functional microenvironment, mimicking the neural niche, which may have interesting future applications for the development of new strategies in neural stem cell research.
Collapse
Affiliation(s)
- Marta S. Carvalho
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Corresponding author.
| | - Diogo E.S. Nogueira
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Joaquim M.S. Cabral
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Carlos A.V. Rodrigues
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
23
|
Amadei G, Handford CE, Qiu C, De Jonghe J, Greenfeld H, Tran M, Martin BK, Chen DY, Aguilera-Castrejon A, Hanna JH, Elowitz MB, Hollfelder F, Shendure J, Glover DM, Zernicka-Goetz M. Embryo model completes gastrulation to neurulation and organogenesis. Nature 2022; 610:143-153. [PMID: 36007540 PMCID: PMC9534772 DOI: 10.1038/s41586-022-05246-3] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 08/17/2022] [Indexed: 11/23/2022]
Abstract
Embryonic stem (ES) cells can undergo many aspects of mammalian embryogenesis in vitro1-5, but their developmental potential is substantially extended by interactions with extraembryonic stem cells, including trophoblast stem (TS) cells, extraembryonic endoderm stem (XEN) cells and inducible XEN (iXEN) cells6-11. Here we assembled stem cell-derived embryos in vitro from mouse ES cells, TS cells and iXEN cells and showed that they recapitulate the development of whole natural mouse embryo in utero up to day 8.5 post-fertilization. Our embryo model displays headfolds with defined forebrain and midbrain regions and develops a beating heart-like structure, a trunk comprising a neural tube and somites, a tail bud containing neuromesodermal progenitors, a gut tube, and primordial germ cells. This complete embryo model develops within an extraembryonic yolk sac that initiates blood island development. Notably, we demonstrate that the neurulating embryo model assembled from Pax6-knockout ES cells aggregated with wild-type TS cells and iXEN cells recapitulates the ventral domain expansion of the neural tube that occurs in natural, ubiquitous Pax6-knockout embryos. Thus, these complete embryoids are a powerful in vitro model for dissecting the roles of diverse cell lineages and genes in development. Our results demonstrate the self-organization ability of ES cells and two types of extraembryonic stem cells to reconstitute mammalian development through and beyond gastrulation to neurulation and early organogenesis.
Collapse
Affiliation(s)
- Gianluca Amadei
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Biology, University of Padua, Padua, Italy
| | - Charlotte E Handford
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Joachim De Jonghe
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Francis Crick Institute, London, UK
| | - Hannah Greenfeld
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Martin Tran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Dong-Yuan Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | | | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - David M Glover
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| |
Collapse
|
24
|
Feng Y, Zhong ZW, Xu Y, Zhang ZY, Ao LL, Yang Z, Wang YL, Jiang YH. Characterization of the transcription factor Sox3 regulating the gonadal development of pearlscale angelfish (Centropyge vrolikii). FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:1193-1207. [PMID: 35963922 DOI: 10.1007/s10695-022-01110-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/03/2022] [Indexed: 06/15/2023]
Abstract
As a member of the Sox gene family, Sox3 plays a vital role in gonadal development and gametogenesis. Nevertheless, the exact expression pattern of this gene in fish is still unknown. Here, we identified the Sox3 gene of Centropyge vrolikii, namely, Cv-Sox3. The Cv-Sox3 mRNA expression in the ovary and testis was detected by reverse transcription-polymerase chain reaction (RT-PCR) analysis, and the mRNA expression level of Cv-Sox3 in the ovary in the resting stage was significantly higher than that in other tissues. The phylogenetic tree and alignment of multiple sequences were constructed to analyze the evolutionary relationships of Cv-Sox3. Cv-Sox3 was relatively conserved in the evolution of teleost fish, indicating the importance and similarity of its function. The in situ hybridization results demonstrate that Cv-Sox3 was present in the follicle cells and cytoplasm of oocytes in the ovary of different stages, and the positive signals occurred in germ cells of the testis. After interfering with Cv-Sox3, the growth rate of ovarian cells in culture became slow, and the expression of ovary-bias-related genes Cyp19a and Foxl2 significantly increased. Meanwhile, the expression of testis-bias-related genes Dmrt1, Sox9, Cyp11a, Amh, and Sox8 significantly decreased. These results suggest that Cv-Sox3 gene might be expressed in the germ cells of male and female gonads during gonadal development. This study provides a precise expression pattern of Cv-Sox3 and demonstrates that Cv-Sox3 might play a significant role in the reproductive regulation of C. vrolikii. In this study, Sox3 of C. vrolikii (Cv-Sox3) was cloned to understand the expression pattern in the gonadal development, which is expressed in germ cells, involved in the process of gonadal development. The results demonstrated that Cv-Sox3 may play a significant role in the reproductive regulation of C. vrolikii.
Collapse
Affiliation(s)
- Yan Feng
- Key Laboratory of Healthy Mariculture for East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- National Demonstration Center for Experimental Aquatic Science and Technology Education, Jimei University, Xiamen, 361021, China
| | - Zhao-Wei Zhong
- Key Laboratory of Healthy Mariculture for East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- National Demonstration Center for Experimental Aquatic Science and Technology Education, Jimei University, Xiamen, 361021, China
| | - Yan Xu
- Key Laboratory of Healthy Mariculture for East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- National Demonstration Center for Experimental Aquatic Science and Technology Education, Jimei University, Xiamen, 361021, China
| | - Ze-Yu Zhang
- College of Food and Biological Engineering, Jimei University, Xiamen, 361021, Fujian, China
| | - Lu-Lu Ao
- Key Laboratory of Healthy Mariculture for East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- National Demonstration Center for Experimental Aquatic Science and Technology Education, Jimei University, Xiamen, 361021, China
| | - Zhen Yang
- Key Laboratory of Healthy Mariculture for East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
- National Demonstration Center for Experimental Aquatic Science and Technology Education, Jimei University, Xiamen, 361021, China
| | - Yi-Lei Wang
- Key Laboratory of Healthy Mariculture for East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China.
- National Demonstration Center for Experimental Aquatic Science and Technology Education, Jimei University, Xiamen, 361021, China.
| | - Yong-Hua Jiang
- Key Laboratory of Healthy Mariculture for East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China.
- National Demonstration Center for Experimental Aquatic Science and Technology Education, Jimei University, Xiamen, 361021, China.
| |
Collapse
|
25
|
Wei X, Fu S, Li H, Liu Y, Wang S, Feng W, Yang Y, Liu X, Zeng YY, Cheng M, Lai Y, Qiu X, Wu L, Zhang N, Jiang Y, Xu J, Su X, Peng C, Han L, Lou WPK, Liu C, Yuan Y, Ma K, Yang T, Pan X, Gao S, Chen A, Esteban MA, Yang H, Wang J, Fan G, Liu L, Chen L, Xu X, Fei JF, Gu Y. Single-cell Stereo-seq reveals induced progenitor cells involved in axolotl brain regeneration. Science 2022; 377:eabp9444. [PMID: 36048929 DOI: 10.1126/science.abp9444] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The molecular mechanism underlying brain regeneration in vertebrates remains elusive. We performed spatial enhanced resolution omics sequencing (Stereo-seq) to capture spatially resolved single-cell transcriptomes of axolotl telencephalon sections during development and regeneration. Annotated cell types exhibited distinct spatial distribution, molecular features, and functions. We identified an injury-induced ependymoglial cell cluster at the wound site as a progenitor cell population for the potential replenishment of lost neurons, through a cell state transition process resembling neurogenesis during development. Transcriptome comparisons indicated that these induced cells may originate from local resident ependymoglial cells. We further uncovered spatially defined neurons at the lesion site that may regress to an immature neuron-like state. Our work establishes spatial transcriptome profiles of an anamniote tetrapod brain and decodes potential neurogenesis from ependymoglial cells for development and regeneration, thus providing mechanistic insights into vertebrate brain regeneration.
Collapse
Affiliation(s)
- Xiaoyu Wei
- BGI-Hangzhou, Hangzhou 310012, China.,BGI-Shenzhen, Shenzhen 518103, China
| | - Sulei Fu
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Hanbo Li
- BGI-Shenzhen, Shenzhen 518103, China.,BGI-Qingdao, Qingdao 266555, China.,Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, Qingdao 266555, China
| | - Yang Liu
- BGI-Shenzhen, Shenzhen 518103, China
| | - Shuai Wang
- BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weimin Feng
- BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunzhi Yang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | | | - Yan-Yun Zeng
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Mengnan Cheng
- BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiwei Lai
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xiaojie Qiu
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Liang Wu
- BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Yujia Jiang
- BGI-Shenzhen, Shenzhen 518103, China.,BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Jiangshan Xu
- BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Cheng Peng
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Lei Han
- BGI-Shenzhen, Shenzhen 518103, China.,Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China.,Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Wilson Pak-Kin Lou
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Chuanyu Liu
- BGI-Shenzhen, Shenzhen 518103, China.,Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Yue Yuan
- BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Tao Yang
- BGI-Shenzhen, Shenzhen 518103, China
| | - Xiangyu Pan
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | | | - Ao Chen
- BGI-Shenzhen, Shenzhen 518103, China.,Department of Biology, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Miguel A Esteban
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.,Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518103, China.,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518103, China.,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | | | - Longqi Liu
- BGI-Hangzhou, Hangzhou 310012, China.,BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.,Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Liang Chen
- Hubei Key Laboratory of Cell Homeostasis, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518103, China.,Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | - Ji-Feng Fei
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Ying Gu
- BGI-Hangzhou, Hangzhou 310012, China.,BGI-Shenzhen, Shenzhen 518103, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.,Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| |
Collapse
|
26
|
Jeong S, Park SM, Jo NR, Kwon JS, Lee J, Kim K, Go SM, Cai L, Ahn D, Lee SD, Hyun SH, Choi KC, Jeung EB. Pre-validation of an alternative test method for prediction of developmental neurotoxicity. Food Chem Toxicol 2022; 164:113070. [PMID: 35483486 DOI: 10.1016/j.fct.2022.113070] [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: 01/17/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 11/26/2022]
Abstract
Exposure to neurodevelopmental toxicants can cause permanent brain injury. Hance, determining the neurotoxicity of unknown substances is essential for the safety of substance. As an alternative method to animal studies, developmental neurotoxicity test (DNT) and the first discriminant function (DF) were established in previous study. This study aimed to increase the predictability of the DNT method and perform a mobility test. Two endpoints of 29 newly investigated substances were used to establish a second-generation DF (2nd GDF). As two endpoints, the half-inhibitory concentration of the cell viability (IC50) was determined using a cell counting kit-8 assay. The half-inhibitory concentration of differentiation (ID50) was determined by measuring the green fluorescent protein (GFP) intensity in 46C cells. The substances were treated dose-dependently to measure IC50 and ID50. The 2nd GDF classified 29 chemicals accurately as toxic and non-toxic. Four participants of three independent laboratories were enrolled to test the mobility. The results of the test set were highly accurate in reproducibility (100% of accuracy, sensitivity, and specificity) and mobility (accuracy 93.33%, sensitivity 90.91%, and specificity 100%). In conclusion, the protocol is transferable, reproducible, and accurate. Therefore, this could be a standardizing method for determining a neurotoxicant as an alternative for animal experiments.
Collapse
Affiliation(s)
- SunHwa Jeong
- Laboratory of Veterinary Biochemistry and Molecular Biology (LVBMB), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Seon-Mi Park
- Laboratory of Veterinary Biochemistry and Molecular Biology (LVBMB), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Na Rea Jo
- Department of Information and Statistics (DIS), College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Jin-Sook Kwon
- Laboratory of Veterinary Biochemistry and Molecular Biology (LVBMB), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Jimin Lee
- Laboratory of Veterinary Biochemistry and Molecular Biology (LVBMB), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - KangMin Kim
- Laboratory of Veterinary Biochemistry and Molecular Biology (LVBMB), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Seon Myeong Go
- Laboratory of Veterinary Biochemistry and Molecular Biology (LVBMB), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Lian Cai
- Laboratory of Veterinary Embryology and Biotechnology (LVEB), College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Dohee Ahn
- Laboratory of Biochemistry and Immunology (LBI), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Sung Duck Lee
- Department of Information and Statistics (DIS), College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (LVEB), College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology (LBI), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology (LVBMB), College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
27
|
Yaghmaeian Salmani B, Balderson B, Bauer S, Ekman H, Starkenberg A, Perlmann T, Piper M, Bodén M, Thor S. Selective requirement for polycomb repressor complex 2 in the generation of specific hypothalamic neuronal subtypes. Development 2022; 149:274592. [PMID: 35245348 PMCID: PMC8959139 DOI: 10.1242/dev.200076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/18/2022] [Indexed: 11/20/2022]
Abstract
The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the polycomb repressor complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3, a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g. Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that most neuronal subtypes were still generated in Eed mutants. However, we observed an increase in glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, hypocretin and Tac2-Pax6 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, but points to a unique sensitivity of particular neuronal subtypes to a disrupted epigenomic landscape. Summary: Polycomb repressor complex 2 inactivation results in selective effects on mouse hypothalamic development, increasing glutamatergic/GABA cells, while reducing dopamine, Hcrt and Tac2-Pax6 cells.
Collapse
Affiliation(s)
- Behzad Yaghmaeian Salmani
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
- Department of Cell and Molecular Biology, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Brad Balderson
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Susanne Bauer
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Helen Ekman
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Annika Starkenberg
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Michael Piper
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Mikael Bodén
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
28
|
Dady A, Davidson L, Halley PA, Storey KG. Human spinal cord in vitro differentiation pace is initially maintained in heterologous embryonic environments. eLife 2022; 11:e67283. [PMID: 35188104 PMCID: PMC8929931 DOI: 10.7554/elife.67283] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 02/02/2022] [Indexed: 11/23/2022] Open
Abstract
Species-specific differentiation pace in vitro indicates that some aspects of neural differentiation are governed by cell intrinsic properties. Here we describe a novel in vitro human neural-rosette assay that recapitulates dorsal spinal cord differentiation but proceeds more rapidly than in the human embryo, suggesting that it lacks endogenous signalling dynamics. To test whether in vitro conditions represent an intrinsic differentiation pace, human iPSC-derived neural rosettes were challenged by grafting into the faster differentiating chicken embryonic neural tube iso-chronically, or hetero-chronically into older embryos. In both contexts in vitro differentiation pace was initially unchanged, while long-term analysis revealed iso-chronic slowed and hetero-chronic conditions promoted human neural differentiation. Moreover, hetero-chronic conditions did not alter the human neural differentiation programme, which progressed to neurogenesis, while the host embryo advanced into gliogenesis. This study demonstrates that intrinsic properties limit human differentiation pace, and that timely extrinsic signals are required for progression through an intrinsic human neural differentiation programme.
Collapse
Affiliation(s)
- Alwyn Dady
- Division of Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Lindsay Davidson
- Division of Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Pamela A Halley
- Division of Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Kate G Storey
- Division of Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| |
Collapse
|
29
|
Xi J, Xu Y, Guo Z, Li J, Wu Y, Sun Q, Wang Y, Chen M, Zhu S, Bian S, Kang J. LncRNA SOX1-OT V1 acts as a decoy of HDAC10 to promote SOX1-dependent hESC neuronal differentiation. EMBO Rep 2022; 23:e53015. [PMID: 34927789 PMCID: PMC8811645 DOI: 10.15252/embr.202153015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 02/05/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are abundantly expressed in the nervous system, but their regulatory roles in neuronal differentiation are poorly understood. Using a human embryonic stem cell (hESC)-based 2D neural differentiation approach and a 3D cerebral organoid system, we show that SOX1-OT variant 1 (SOX1-OT V1), a SOX1 overlapping noncoding RNA, plays essential roles in both dorsal cortical neuron differentiation and ventral GABAergic neuron differentiation by facilitating SOX1 expression. SOX1-OT V1 physically interacts with HDAC10 through its 5' region, acts as a decoy to block HDAC10 binding to the SOX1 promoter, and thus maintains histone acetylation levels at the SOX1 promoter. SOX1 in turn activates ASCL1 expression and promotes neuronal differentiation. Taken together, we identify a SOX1-OT V1/HDAC10-SOX1-ASCL1 axis, which promotes neurogenesis, highlighting a role for lncRNAs in hESC neuronal differentiation.
Collapse
Affiliation(s)
- Jiajie Xi
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yanxin Xu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zhenming Guo
- Institute for Regenerative MedicineShanghai East HospitalSchool of Life Sciences and TechnologyFrontier Science Center for Stem Cell ResearchTongji UniversityShanghaiChina
| | - Jianguo Li
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yukang Wu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Qiaoyi Sun
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yuxi Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Mengxia Chen
- Institute for Regenerative MedicineShanghai East HospitalSchool of Life Sciences and TechnologyFrontier Science Center for Stem Cell ResearchTongji UniversityShanghaiChina
| | - Songcheng Zhu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Shan Bian
- Institute for Regenerative MedicineShanghai East HospitalSchool of Life Sciences and TechnologyFrontier Science Center for Stem Cell ResearchTongji UniversityShanghaiChina
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant HospitalShanghai Key Laboratory of Maternal Fetal MedicineShanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchNational Stem Cell Translational Resource CenterSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| |
Collapse
|
30
|
Bérenger-Currias NM, Mircea M, Adegeest E, van den Berg PR, Feliksik M, Hochane M, Idema T, Tans SJ, Semrau S. A gastruloid model of the interaction between embryonic and extra-embryonic cell types. J Tissue Eng 2022; 13:20417314221103042. [PMID: 35707767 PMCID: PMC9189523 DOI: 10.1177/20417314221103042] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022] Open
Abstract
Stem-cell derived in vitro systems, such as organoids or embryoids, hold great
potential for modeling in vivo development. Full control over their initial
composition, scalability, and easily measurable dynamics make those systems
useful for studying specific developmental processes in isolation. Here we
report the formation of gastruloids consisting of mouse embryonic stem cells
(mESCs) and extraembryonic endoderm (XEN) cells. These XEN-enhanced gastruloids
(XEGs) exhibit the formation of neural epithelia, which are absent in
gastruloids derived from mESCs only. By single-cell RNA-seq, imaging, and
differentiation experiments, we demonstrate the neural characteristics of the
epithelial tissue. We further show that the mESCs induce the differentiation of
the XEN cells to a visceral endoderm-like state. Finally, we demonstrate that
local inhibition of WNT signaling and production of a basement membrane by the
XEN cells underlie the formation of the neuroepithelial tissue. In summary, we
establish XEGs to explore heterotypic cellular interactions and their
developmental consequences in vitro.
Collapse
Affiliation(s)
- Noémie Mlp Bérenger-Currias
- Department of Physics, Leiden University, Leiden, The Netherlands.,Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft, The Netherlands
| | - Maria Mircea
- Department of Physics, Leiden University, Leiden, The Netherlands
| | - Esmée Adegeest
- Department of Physics, Leiden University, Leiden, The Netherlands
| | | | - Marleen Feliksik
- Department of Physics, Leiden University, Leiden, The Netherlands
| | - Mazène Hochane
- Department of Physics, Leiden University, Leiden, The Netherlands
| | - Timon Idema
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft, The Netherlands
| | - Sander J Tans
- Delft University of Technology, Department of Bionanoscience, Kavli Institute of Nanoscience, Delft, The Netherlands.,AMOLF, Amsterdam, The Netherlands
| | - Stefan Semrau
- Department of Physics, Leiden University, Leiden, The Netherlands
| |
Collapse
|
31
|
Russo L, Sladitschek HL, Neveu PA. Multi-layered regulation of neuroectoderm differentiation by retinoic acid in a primitive streak-like context. Stem Cell Reports 2022; 17:231-244. [PMID: 35063128 PMCID: PMC8828549 DOI: 10.1016/j.stemcr.2021.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The formation of the primitive streak (PS) and the subsequent induction of neuroectoderm are hallmarks of gastrulation. Combining an in vitro reconstitution of this process based on mouse embryonic stem cells (mESCs) with a collection of knockouts in reporter mESC lines, we identified retinoic acid (RA) as a critical mediator of early neural induction triggered by TGFβ or Wnt signaling inhibition. Single-cell RNA sequencing analysis captured the temporal unfolding of cell type diversification, up to the emergence of somite and neural fates. In the absence of the RA-synthesizing enzyme Aldh1a2, a sensitive RA reporter revealed a hitherto unidentified residual RA signaling that specified neural fate. Genetic evidence showed that the RA-degrading enzyme Cyp26a1 protected PS-like cells from neural induction, even in the absence of TGFβ and Wnt antagonists. Overall, we characterized a multi-layered control of RA levels that regulates early neural differentiation in an in vitro PS-like system. In vitro reconstitution of neural induction by primitive streak-like cells Retinoic acid mediates neural induction triggered by TGFβ or Wnt signaling inhibition A sensitive activity reporter reveals Aldh1a2-independent retinoic acid signaling Cyp26a1 protects primitive streak-like cells from neural induction
Collapse
Affiliation(s)
- Luigi Russo
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; Joint PhD Degree from EMBL and Heidelberg University, Faculty of Biosciences, 69120 Heidelberg, Germany
| | - Hanna L Sladitschek
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Pierre A Neveu
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.
| |
Collapse
|
32
|
Wilkins HM, Troutwine BR, Menta BW, Manley SJ, Strope TA, Lysaker CR, Swerdlow RH. Mitochondrial Membrane Potential Influences Amyloid-β Protein Precursor Localization and Amyloid-β Secretion. J Alzheimers Dis 2022; 85:381-394. [PMID: 34806611 PMCID: PMC9212216 DOI: 10.3233/jad-215280] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Amyloid-β (Aβ), which derives from the amyloid-β protein precursor (AβPP), forms plaques and serves as a fluid biomarker in Alzheimer's disease (AD). How Aβ forms from AβPP is known, but questions relating to AβPP and Aβ biology remain unanswered. AD patients show mitochondrial dysfunction, and an Aβ/AβPP mitochondria relationship exists. OBJECTIVE We considered how mitochondrial biology may impact AβPP and Aβ biology. METHODS SH-SY5Y cells were transfected with AβPP constructs. After treatment with FCCP (uncoupler), Oligomycin (ATP synthase inhibitor), or starvation Aβ levels were measured. β-secretase (BACE1) expression was measured. Mitochondrial localized full-length AβPP was also measured. All parameters listed were measured in ρ0 cells on an SH-SY5Y background. iPSC derived neurons were also used to verify key results. RESULTS We showed that mitochondrial depolarization routes AβPP to, while hyperpolarization routes AβPP away from, the organelle. Mitochondrial AβPP and cell Aβ secretion inversely correlate, as cells with more mitochondrial AβPP secrete less Aβ, and cells with less mitochondrial AβPP secrete more Aβ. An inverse relationship between secreted/extracellular Aβ and intracellular Aβ was observed. CONCLUSION Our findings indicate mitochondrial function alters AβPP localization and suggest enhanced mitochondrial activity promotes Aβ secretion while depressed mitochondrial activity minimizes Aβ secretion. Our data complement other studies that indicate a mitochondrial, AβPP, and Aβ nexus, and could help explain why cerebrospinal fluid Aβ is lower in those with AD. Our data further suggest Aβ secretion could serve as a biomarker of cell or tissue mitochondrial function.
Collapse
Affiliation(s)
- Heather M. Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Benjamin R. Troutwine
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
| | - Blaise W. Menta
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Sharon J. Manley
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
| | - Taylor A. Strope
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Colton R. Lysaker
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Russell H. Swerdlow
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
33
|
Galiakberova AA, Surin AM, Bakaeva ZV, Sharipov RR, Zhang D, Dorovskoy DA, Shakirova KM, Fisenko AP, Dashinimaev EB. IPSC-Derived Human Neurons with GCaMP6s Expression Allow In Vitro Study of Neurophysiological Responses to Neurochemicals. Neurochem Res 2021; 47:952-966. [PMID: 34855047 PMCID: PMC8891101 DOI: 10.1007/s11064-021-03497-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022]
Abstract
The study of human neurons and their interaction with neurochemicals is difficult due to the inability to collect primary biomaterial. However, recent advances in the cultivation of human stem cells, methods for their neuronal differentiation and chimeric fluorescent calcium indicators have allowed the creation of model systems in vitro. In this paper we report on the development of a method to obtain human neurons with the GCaMP6s calcium indicator, based on a human iPSC line with the TetON–NGN2 transgene complex. The protocol we developed allows us quickly, conveniently and efficiently obtain significant amounts of human neurons suitable for the study of various neurochemicals and their effects on specific neurophysiological activity, which can be easily registered using fluorescence microscopy. In the neurons we obtained, glutamate (Glu) induces rises in [Ca2+]i which are caused by ionotropic receptors for Glu, predominantly of the NMDA-type. Taken together, these facts allow us to consider the model we have created to be a useful and successful development of this technology.
Collapse
Affiliation(s)
- A A Galiakberova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, Moscow, Russia, 117997.
- Faculty of Biology, Lomonosov Moscow State University, GSP-1, Leninskie Gory, Moscow, Russia, 119991.
| | - A M Surin
- Laboratory of Neurobiology, "National Medical Research Center of Children's Health", Russian Ministry of Health, Lomonosov Avenue, Moscow, Russia, 119991
- Laboratory of Pathology of Ion Transport and Intracellular Signaling, Institute of General Pathology and Pathophysiology, Baltiyskaya St., Moscow, Russia, 125315
| | - Z V Bakaeva
- Laboratory of Neurobiology, "National Medical Research Center of Children's Health", Russian Ministry of Health, Lomonosov Avenue, Moscow, Russia, 119991
- Department of General Biology and Physiology, Gorodovikov Kalmyk State University, Pushkin St., Elista, Russia, 358000
| | - R R Sharipov
- Laboratory of Pathology of Ion Transport and Intracellular Signaling, Institute of General Pathology and Pathophysiology, Baltiyskaya St., Moscow, Russia, 125315
| | - Dongxing Zhang
- Moscow Institute of Physics and Technology (State University), Institutskiy per., 141701, Dolgoprudny, Russia
| | - D A Dorovskoy
- Moscow Institute of Physics and Technology (State University), Institutskiy per., 141701, Dolgoprudny, Russia
| | - K M Shakirova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, Moscow, Russia, 117997
| | - A P Fisenko
- Laboratory of Neurobiology, "National Medical Research Center of Children's Health", Russian Ministry of Health, Lomonosov Avenue, Moscow, Russia, 119991
| | - E B Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, Moscow, Russia, 117997
- Moscow Institute of Physics and Technology (State University), Institutskiy per., 141701, Dolgoprudny, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov St., Moscow, Russia, 119334
| |
Collapse
|
34
|
Walter J, Bolognin S, Poovathingal SK, Magni S, Gérard D, Antony PMA, Nickels SL, Salamanca L, Berger E, Smits LM, Grzyb K, Perfeito R, Hoel F, Qing X, Ohnmacht J, Bertacchi M, Jarazo J, Ignac T, Monzel AS, Gonzalez-Cano L, Krüger R, Sauter T, Studer M, de Almeida LP, Tronstad KJ, Sinkkonen L, Skupin A, Schwamborn JC. The Parkinson's-disease-associated mutation LRRK2-G2019S alters dopaminergic differentiation dynamics via NR2F1. Cell Rep 2021; 37:109864. [PMID: 34686322 DOI: 10.1016/j.celrep.2021.109864] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 07/27/2021] [Accepted: 09/29/2021] [Indexed: 11/19/2022] Open
Abstract
Increasing evidence suggests that neurodevelopmental alterations might contribute to increase the susceptibility to develop neurodegenerative diseases. We investigate the occurrence of developmental abnormalities in dopaminergic neurons in a model of Parkinson's disease (PD). We monitor the differentiation of human patient-specific neuroepithelial stem cells (NESCs) into dopaminergic neurons. Using high-throughput image analyses and single-cell RNA sequencing, we observe that the PD-associated LRRK2-G2019S mutation alters the initial phase of neuronal differentiation by accelerating cell-cycle exit with a concomitant increase in cell death. We identify the NESC-specific core regulatory circuit and a molecular mechanism underlying the observed phenotypes. The expression of NR2F1, a key transcription factor involved in neurogenesis, decreases in LRRK2-G2019S NESCs, neurons, and midbrain organoids compared to controls. We also observe accelerated dopaminergic differentiation in vivo in NR2F1-deficient mouse embryos. This suggests a pathogenic mechanism involving the LRRK2-G2019S mutation, where the dynamics of dopaminergic differentiation are modified via NR2F1.
Collapse
Affiliation(s)
- Jonas Walter
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Silvia Bolognin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Suresh K Poovathingal
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Stefano Magni
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Deborah Gérard
- Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Paul M A Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Sarah L Nickels
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Luis Salamanca
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Emanuel Berger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Lisa M Smits
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Rita Perfeito
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal
| | - Fredrik Hoel
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Xiaobing Qing
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Jochen Ohnmacht
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | | | - Javier Jarazo
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Tomasz Ignac
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Anna S Monzel
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Laura Gonzalez-Cano
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Transversal Translational Medicine, Luxembourg Institute of Health, Strassen, Luxembourg; Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg
| | - Thomas Sauter
- Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, 06108 Nice, France
| | - Luis Pereira de Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Karl J Tronstad
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Lasse Sinkkonen
- Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Center for Research of Biological Systems, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg.
| |
Collapse
|
35
|
Chen ACH, Huang W, Fong SW, Chan C, Lee KC, Yeung WSB, Lee YL. Hyperglycemia Altered DNA Methylation Status and Impaired Pancreatic Differentiation from Embryonic Stem Cells. Int J Mol Sci 2021; 22:10729. [PMID: 34639069 PMCID: PMC8509790 DOI: 10.3390/ijms221910729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 11/16/2022] Open
Abstract
The prevalence of type 2 diabetes (T2D) is rapidly increasing across the globe. Fetal exposure to maternal diabetes was correlated with higher prevalence of impaired glucose tolerance and T2D later in life. Previous studies showed aberrant DNA methylation patterns in pancreas of T2D patients. However, the underlying mechanisms remained largely unknown. We utilized human embryonic stem cells (hESC) as the in vitro model for studying the effects of hyperglycemia on DNA methylome and early pancreatic differentiation. Culture in hyperglycemic conditions disturbed the pancreatic lineage potential of hESC, leading to the downregulation of expression of pancreatic markers PDX1, NKX6-1 and NKX6-2 after in vitro differentiation. Genome-wide DNA methylome profiling revealed over 2000 differentially methylated CpG sites in hESC cultured in hyperglycemic condition when compared with those in control glucose condition. Gene ontology analysis also revealed that the hypermethylated genes were enriched in cell fate commitment. Among them, NKX6-2 was validated and its hypermethylation status was maintained upon differentiation into pancreatic progenitor cells. We also established mouse ESC lines at both physiological glucose level (PG-mESC) and conventional hyperglycemia glucose level (HG-mESC). Concordantly, DNA methylome analysis revealed the enrichment of hypermethylated genes related to cell differentiation in HG-mESC, including Nkx6-1. Our results suggested that hyperglycemia dysregulated the epigenome at early fetal development, possibly leading to impaired pancreatic development.
Collapse
Affiliation(s)
- Andy Chun Hang Chen
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong, Shenzhen Hospital, Shenzhen 518000, China;
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Wen Huang
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Sze Wan Fong
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Chris Chan
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Kai Chuen Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - William Shu Biu Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong, Shenzhen Hospital, Shenzhen 518000, China;
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Yin Lau Lee
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong, Shenzhen Hospital, Shenzhen 518000, China;
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| |
Collapse
|
36
|
Wei YL, Wen B, Gao JZ, Chen ZZ. Brain transcriptome analysis reveals genes involved in parental care behaviour in discus fish (Symphysodon haraldi). Gen Comp Endocrinol 2021; 309:113793. [PMID: 33887271 DOI: 10.1016/j.ygcen.2021.113793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
Parental care is common in mammals and allows offspring to obtain milk, a substance rich in a range of nutritional and non-nutritional factors crucial to the survival of newborns. The discus fish Symphysodon spp., an Amazonian cichlid, shows an unusual behaviour: Free-swimming fry bite on their parents' skin mucus for growth and development during the first month after hatching. This is similar to the breastfeeding behaviour of mammals, but little is known about the regulatory mechanism by which discus secrete 'milk' and the related genes involved in parental care. Here, transcriptome sequencing was performed by using the brain tissues of female discus fish in parental and non-parental care. The results showed that a total of 86 differentially expressed genes (71 up-regulated genes and 15 down-regulated genes) were obtained by comparing parental with non-parental discus fish, including up-regulated LAPTM, FOXB, SOX1S, OTX2 and NR1F2, and down-regulated EDNRB, PRKCD, H1-5 and HBE. Through functional enrichment analysis, a total of 20 pathways were identified, e.g., estrogen signaling pathway, inflammatory mediator regulation of TRP channels, vascular smooth muscle contraction, GnRH signaling pathway, neurotrophin signaling pathway, NOD-like receptor signaling pathway, Jak-STAT signaling pathway, Fc gamma R-mediated phagocytosis, serotonergic synapse, autophagy-animal and cytokine-cytokine receptor interaction. These pathways and related genes might play important roles in the regulation of discus 'milk' secretion.
Collapse
Affiliation(s)
- Yu-Ling Wei
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Bin Wen
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| | - Jian-Zhong Gao
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Zai-Zhong Chen
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
37
|
Kaji DA, Montero AM, Patel R, Huang AH. Transcriptional profiling of mESC-derived tendon and fibrocartilage cell fate switch. Nat Commun 2021; 12:4208. [PMID: 34244516 PMCID: PMC8270956 DOI: 10.1038/s41467-021-24535-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
The transcriptional regulators underlying induction and differentiation of dense connective tissues such as tendon and related fibrocartilaginous tissues (meniscus and annulus fibrosus) remain largely unknown. Using an iterative approach informed by developmental cues and single cell RNA sequencing (scRNA-seq), we establish directed differentiation models to generate tendon and fibrocartilage cells from mouse embryonic stem cells (mESCs) by activation of TGFβ and hedgehog pathways, achieving 90% induction efficiency. Transcriptional signatures of the mESC-derived cells recapitulate embryonic tendon and fibrocartilage signatures from the mouse tail. scRNA-seq further identify retinoic acid signaling as a critical regulator of cell fate switch between TGFβ-induced tendon and fibrocartilage lineages. Trajectory analysis by RNA sequencing define transcriptional modules underlying tendon and fibrocartilage fate induction and identify molecules associated with lineage-specific differentiation. Finally, we successfully generate 3-dimensional engineered tissues using these differentiation protocols and show activation of mechanotransduction markers with dynamic tensile loading. These findings provide a serum-free approach to generate tendon and fibrocartilage cells and tissues at high efficiency for modeling development and disease.
Collapse
Affiliation(s)
- Deepak A Kaji
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angela M Montero
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roosheel Patel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
38
|
Stevanovic M, Drakulic D, Lazic A, Ninkovic DS, Schwirtlich M, Mojsin M. SOX Transcription Factors as Important Regulators of Neuronal and Glial Differentiation During Nervous System Development and Adult Neurogenesis. Front Mol Neurosci 2021; 14:654031. [PMID: 33867936 PMCID: PMC8044450 DOI: 10.3389/fnmol.2021.654031] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The SOX proteins belong to the superfamily of transcription factors (TFs) that display properties of both classical TFs and architectural components of chromatin. Since the cloning of the Sox/SOX genes, remarkable progress has been made in illuminating their roles as key players in the regulation of multiple developmental and physiological processes. SOX TFs govern diverse cellular processes during development, such as maintaining the pluripotency of stem cells, cell proliferation, cell fate decisions/germ layer formation as well as terminal cell differentiation into tissues and organs. However, their roles are not limited to development since SOX proteins influence survival, regeneration, cell death and control homeostasis in adult tissues. This review summarized current knowledge of the roles of SOX proteins in control of central nervous system development. Some SOX TFs suspend neural progenitors in proliferative, stem-like state and prevent their differentiation. SOX proteins function as pioneer factors that occupy silenced target genes and keep them in a poised state for activation at subsequent stages of differentiation. At appropriate stage of development, SOX members that maintain stemness are down-regulated in cells that are competent to differentiate, while other SOX members take over their functions and govern the process of differentiation. Distinct SOX members determine down-stream processes of neuronal and glial differentiation. Thus, sequentially acting SOX TFs orchestrate neural lineage development defining neuronal and glial phenotypes. In line with their crucial roles in the nervous system development, deregulation of specific SOX proteins activities is associated with neurodevelopmental disorders (NDDs). The overview of the current knowledge about the link between SOX gene variants and NDDs is presented. We outline the roles of SOX TFs in adult neurogenesis and brain homeostasis and discuss whether impaired adult neurogenesis, detected in neurodegenerative diseases, could be associated with deregulation of SOX proteins activities. We present the current data regarding the interaction between SOX proteins and signaling pathways and microRNAs that play roles in nervous system development. Finally, future research directions that will improve the knowledge about distinct and various roles of SOX TFs in health and diseases are presented and discussed.
Collapse
Affiliation(s)
- Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.,Faculty of Biology, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Danijela Drakulic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Andrijana Lazic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Danijela Stanisavljevic Ninkovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Schwirtlich
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marija Mojsin
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
39
|
Yang L, Su Z, Wang Z, Li Z, Shang Z, Du H, Liu G, Qi D, Yang Z, Xu Z, Zhang Z. Transcriptional profiling reveals the transcription factor networks regulating the survival of striatal neurons. Cell Death Dis 2021; 12:262. [PMID: 33712552 PMCID: PMC7955055 DOI: 10.1038/s41419-021-03552-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 01/31/2023]
Abstract
The striatum is structurally highly diverse, and its organ functionality critically depends on normal embryonic development. Although several studies have been conducted on the gene functional changes that occur during striatal development, a system-wide analysis of the underlying molecular changes is lacking. Here, we present a comprehensive transcriptome profile that allows us to explore the trajectory of striatal development and identify the correlation between the striatal development and Huntington's disease (HD). Furthermore, we applied an integrative transcriptomic profiling approach based on machine learning to systematically map a global landscape of 277 transcription factor (TF) networks. Most of these TF networks are linked to biological processes, and some unannotated genes provide information about the corresponding mechanisms. For example, we found that the Meis2 and Six3 were crucial for the survival of striatal neurons, which were verified using conditional knockout (CKO) mice. Finally, we used RNA-Seq to speculate their downstream targets.
Collapse
Affiliation(s)
- Lin Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Zihao Su
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Ziwu Wang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Zicong Shang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Heng Du
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Dashi Qi
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China.
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Shanghai Key Lab of Birth Defect, Children's Hospital, Fudan University, Shanghai, 200032, P.R. China.
| |
Collapse
|
40
|
SOX1 Is a Backup Gene for Brain Neurons and Glioma Stem Cell Protection and Proliferation. Mol Neurobiol 2021; 58:2634-2642. [PMID: 33481176 DOI: 10.1007/s12035-020-02240-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022]
Abstract
Failed neuroprotection leads to the initiation of several diseases. SOX1 plays many roles in embryogenesis, oncogenesis, and male sex determination, and can promote glioma stem cell proliferation, invasion, and migration due to its high expression in glioblastoma cells. The functional versatility of the SOX1 gene in malignancy, epilepsy, and Parkinson's disease, as well as its adverse effects on dopaminergic neurons, makes it an interesting research focus. Hence, we collate the most important discoveries relating to the neuroprotective effects of SOX1 in brain cancer and propose hypothesis worthy of SOX1's role in the survival of senescent neuronal cells, its roles in fibroblast cell proliferation, and cell fat for neuroprotection, and the discharge of electrical impulses for homeostasis. Increase in electrical impulses transmitted by senescent cells affects the synthesis of neurotransmitters, which will modify the brain cell metabolism and microenvironment.
Collapse
|
41
|
An integrative atlas of chicken long non-coding genes and their annotations across 25 tissues. Sci Rep 2020; 10:20457. [PMID: 33235280 PMCID: PMC7686352 DOI: 10.1038/s41598-020-77586-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Long non-coding RNAs (LNC) regulate numerous biological processes. In contrast to human, the identification of LNC in farm species, like chicken, is still lacunar. We propose a catalogue of 52,075 chicken genes enriched in LNC (http://www.fragencode.org/), built from the Ensembl reference extended using novel LNC modelled here from 364 RNA-seq and LNC from four public databases. The Ensembl reference grew from 4,643 to 30,084 LNC, of which 59% and 41% with expression ≥ 0.5 and ≥ 1 TPM respectively. Characterization of these LNC relatively to the closest protein coding genes (PCG) revealed that 79% of LNC are in intergenic regions, as in other species. Expression analysis across 25 tissues revealed an enrichment of co-expressed LNC:PCG pairs, suggesting co-regulation and/or co-function. As expected LNC were more tissue-specific than PCG (25% vs. 10%). Similarly to human, 16% of chicken LNC hosted one or more miRNA. We highlighted a new chicken LNC, hosting miR155, conserved in human, highly expressed in immune tissues like miR155, and correlated with immunity-related PCG in both species. Among LNC:PCG pairs tissue-specific in the same tissue, we revealed an enrichment of divergent pairs with the PCG coding transcription factors, as for example LHX5, HXD3 and TBX4, in both human and chicken.
Collapse
|
42
|
Zhao S, Duan K, Ai Z, Niu B, Chen Y, Kong R, Li T. Generation of cortical neurons through large-scale expanding neuroepithelial stem cell from human pluripotent stem cells. Stem Cell Res Ther 2020; 11:431. [PMID: 33008480 PMCID: PMC7532602 DOI: 10.1186/s13287-020-01939-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/22/2020] [Accepted: 09/17/2020] [Indexed: 02/08/2023] Open
Abstract
Background Considerable progress has been made in converting human pluripotent stem cells (hPSCs) into cortical neurons for disease modeling and regenerative medicine. However, these procedures are hard to provide sufficient cells for their applications. Using a combination of small-molecules and growth factors, we previously identified one condition which can rapidly induce hPSCs into neuroepithelial stem cells (NESCs). Here, we developed a scalable suspension culture system, which largely yields high-quality NESC-spheres and subsequent cortical neurons. Methods The NESC medium was first optimized, and the suspension culture system was then enlarged from plates to stirred bioreactors for large-scale production of NESC-spheres by a stirring speed of 60 rpm. During the expansion, the quality of NESC-spheres was evaluated. The differentiation potential of NESC-spheres into cortical neurons was demonstrated by removing bFGF and two pathway inhibitors from the NESC medium. Cellular immunofluorescence staining, global transcriptome, and single-cell RNA sequencing analysis were used to identify the characteristics, identities, purities, or homogeneities of NESC-spheres or their differentiated cells, respectively. Results The optimized culture system is more conducive to large-scale suspension production of NESCs. These largely expanded NESC-spheres maintain unlimited self-renewal ability and NESC state by retaining their uniform sizes, high cell vitalities, and robust expansion abilities. After long-term expansion, NESC-spheres preserve high purity, homogeneity, and normal diploid karyotype. These expanded NESC-spheres on a large scale have strong differentiation potential and effectively produce mature cortical neurons. Conclusions We developed a serum-free, defined, and low-cost culture system for large-scale expansion of NESCs in stirred suspension bioreactors. The stable and controllable 3D system supports long-term expansion of high-quality and homogeneous NESC-spheres. These NESC-spheres can be used to efficiently give rise to cortical neurons for cell therapy, disease modeling, and drug screening in future.
Collapse
Affiliation(s)
- Shumei Zhao
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Kui Duan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Zongyong Ai
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Baohua Niu
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Yanying Chen
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Ruize Kong
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Tianqing Li
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China. .,Xi'an ChaoYue Stem Cell Co, Ltd, Xi'an, China.
| |
Collapse
|
43
|
Liu X, Fang Z, Wen J, Tang F, Liao B, Jing N, Lai D, Jin Y. SOX1 Is Required for the Specification of Rostral Hindbrain Neural Progenitor Cells from Human Embryonic Stem Cells. iScience 2020; 23:101475. [PMID: 32905879 PMCID: PMC7486433 DOI: 10.1016/j.isci.2020.101475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 07/11/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023] Open
Abstract
Region-specific neural progenitor cells (NPCs) can be generated from human embryonic stem cells (hESCs) by modulating signaling pathways. However, how intrinsic transcriptional factors contribute to the neural regionalization is not well characterized. Here, we generate region-specific NPCs from hESCs and find that SOX1 is highly expressed in NPCs with the rostral hindbrain identity. Moreover, we find that OTX2 inhibits SOX1 expression, displaying exclusive expression between the two factors. Furthermore, SOX1 knockout (KO) leads to the upregulation of midbrain genes and downregulation of rostral hindbrain genes, indicating that SOX1 is required for specification of rostral hindbrain NPCs. Our SOX1 chromatin immunoprecipitation sequencing analysis reveals that SOX1 binds to the distal region of GBX2 to activate its expression. Overexpression of GBX2 largely abrogates SOX1-KO-induced aberrant gene expression. Taken together, this study uncovers previously unappreciated role of SOX1 in early neural regionalization and provides new information for the precise control of the OTX2/GBX2 interface.
Collapse
Affiliation(s)
- Xinyuan Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zhuoqing Fang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Jing Wen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Fan Tang
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - Bing Liao
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
| | - Dongmei Lai
- International Peace Maternity and Child Health Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200030, China
| | - Ying Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
- Basic Clinical Research Center, Renji Hospital, Shanghai JiaoTong University School of Medicine, 160 Pujian Road, Shanghai 200127, China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
| |
Collapse
|
44
|
Sur A, Renfro A, Bergmann PJ, Meyer NP. Investigating cellular and molecular mechanisms of neurogenesis in Capitella teleta sheds light on the ancestor of Annelida. BMC Evol Biol 2020; 20:84. [PMID: 32664907 PMCID: PMC7362552 DOI: 10.1186/s12862-020-01636-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/03/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Diverse architectures of nervous systems (NSs) such as a plexus in cnidarians or a more centralized nervous system (CNS) in insects and vertebrates are present across Metazoa, but it is unclear what selection pressures drove evolution and diversification of NSs. One underlying aspect of this diversity lies in the cellular and molecular mechanisms driving neurogenesis, i.e. generation of neurons from neural precursor cells (NPCs). In cnidarians, vertebrates, and arthropods, homologs of SoxB and bHLH proneural genes control different steps of neurogenesis, suggesting that some neurogenic mechanisms may be conserved. However, data are lacking for spiralian taxa. RESULTS To that end, we characterized NPCs and their daughters at different stages of neurogenesis in the spiralian annelid Capitella teleta. We assessed cellular division patterns in the neuroectoderm using static and pulse-chase labeling with thymidine analogs (EdU and BrdU), which enabled identification of NPCs that underwent multiple rounds of division. Actively-dividing brain NPCs were found to be apically-localized, whereas actively-dividing NPCs for the ventral nerve cord (VNC) were found apically, basally, and closer to the ventral midline. We used lineage tracing to characterize the changing boundary of the trunk neuroectoderm. Finally, to start to generate a genetic hierarchy, we performed double-fluorescent in-situ hybridization (FISH) and single-FISH plus EdU labeling for neurogenic gene homologs. In the brain and VNC, Ct-soxB1 and Ct-neurogenin were expressed in a large proportion of apically-localized, EdU+ NPCs. In contrast, Ct-ash1 was expressed in a small subset of apically-localized, EdU+ NPCs and subsurface, EdU- cells, but not in Ct-neuroD+ or Ct-elav1+ cells, which also were subsurface. CONCLUSIONS Our data suggest a putative genetic hierarchy with Ct-soxB1 and Ct-neurogenin at the top, followed by Ct-ash1, then Ct-neuroD, and finally Ct-elav1. Comparison of our data with that from Platynereis dumerilii revealed expression of neurogenin homologs in proliferating NPCs in annelids, which appears different than the expression of vertebrate neurogenin homologs in cells that are exiting the cell cycle. Furthermore, differences between neurogenesis in the head versus trunk of C. teleta suggest that these two tissues may be independent developmental modules, possibly with differing evolutionary trajectories.
Collapse
Affiliation(s)
- A. Sur
- Department of Biology, Clark University, 950 Main Street, Worcester, MA 01610 USA
| | - A. Renfro
- Department of Biology, Clark University, 950 Main Street, Worcester, MA 01610 USA
| | - P. J. Bergmann
- Department of Biology, Clark University, 950 Main Street, Worcester, MA 01610 USA
| | - N. P. Meyer
- Department of Biology, Clark University, 950 Main Street, Worcester, MA 01610 USA
| |
Collapse
|
45
|
Rao C, Malaguti M, Mason JO, Lowell S. The transcription factor E2A drives neural differentiation in pluripotent cells. Development 2020; 147:dev184093. [PMID: 32487737 PMCID: PMC7328008 DOI: 10.1242/dev.184093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 05/26/2020] [Indexed: 12/21/2022]
Abstract
The intrinsic mechanisms that link extracellular signalling to the onset of neural differentiation are not well understood. In pluripotent mouse cells, BMP blocks entry into the neural lineage via transcriptional upregulation of inhibitor of differentiation (Id) factors. We have previously identified the major binding partner of Id proteins in pluripotent cells as the basic helix-loop-helix (bHLH) transcription factor (TF) E2A. Id1 can prevent E2A from forming heterodimers with bHLH TFs or from forming homodimers. Here, we show that overexpression of a forced E2A homodimer is sufficient to drive robust neural commitment in pluripotent cells, even under non-permissive conditions. Conversely, we find that E2A null cells display a defect in their neural differentiation capacity. E2A acts as an upstream activator of neural lineage genes, including Sox1 and Foxd4, and as a repressor of Nodal signalling. Our results suggest a crucial role for E2A in establishing neural lineage commitment in pluripotent cells.
Collapse
Affiliation(s)
- Chandrika Rao
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Mattias Malaguti
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - John O Mason
- Centre for Discovery Brain Sciences, University of Edinburgh, 15 George Square, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
46
|
Castro VL, Reyes JF, Reyes-Nava NG, Paz D, Quintana AM. Hcfc1a regulates neural precursor proliferation and asxl1 expression in the developing brain. BMC Neurosci 2020; 21:27. [PMID: 32522152 PMCID: PMC7288482 DOI: 10.1186/s12868-020-00577-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Background Precise regulation of neural precursor cell (NPC) proliferation and differentiation is essential to ensure proper brain development and function. The HCFC1 gene encodes a transcriptional co-factor that regulates cell proliferation, and previous studies suggest that HCFC1 regulates NPC number and differentiation. However, the molecular mechanism underlying these cellular deficits has not been completely characterized. Methods Here we created a zebrafish harboring mutations in the hcfc1a gene (the hcfc1aco60/+ allele), one ortholog of HCFC1, and utilized immunohistochemistry and RNA-sequencing technology to understand the function of hcfc1a during neural development. Results The hcfc1aco60/+ allele results in an increased number of NPCs and increased expression of neuronal and glial markers. These neural developmental deficits are associated with larval hypomotility and the abnormal expression of asxl1, a polycomb transcription factor, which we identified as a downstream effector of hcfc1a. Inhibition of asxl1 activity and/or expression in larvae harboring the hcfc1aco60/+ allele completely restored the number of NPCs to normal levels. Conclusion Collectively, our data demonstrate that hcfc1a regulates NPC number, NPC proliferation, motor behavior, and brain development.
Collapse
Affiliation(s)
- Victoria L Castro
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Joel F Reyes
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Nayeli G Reyes-Nava
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - David Paz
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Anita M Quintana
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
47
|
Mishra P, Cohen RI, Zhao N, Moghe PV. Fluorescence-based actin turnover dynamics of stem cells as a profiling method for stem cell functional evolution, heterogeneity and phenotypic lineage parsing. Methods 2020; 190:44-54. [PMID: 32473293 DOI: 10.1016/j.ymeth.2020.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Stem cells are widely explored in regenerative medicine as a source to produce diverse cell types. Despite the wide usage of stem cells like mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs), there is a lack of robust methods to rapidly discern the phenotypic and functional heterogeneity of stem cells. The organization of actin cytoskeleton has been previously used to discern divergent stem cell differentiation pathways. In this paper, we highlight the versatility of a cell profiling method for actin turnover dynamics. Actin filaments in live stem cells are labeled using SiR-actin, a cell permeable fluorogenic probe, to determine the endogenous actin turnover. Live MSC imaging after days of induction successfully demonstrated lineage specific change in actin turnover. Next, we highlighted the differences in the cellular heterogeneity of actin dynamics during adipogenic or osteogenic MSC differentiation. Next, we applied the method to differentiating iPSCs in culture, and detected a progressive slowdown in actin turnover during differentiation upon stimulation with neural or cardiac media. Finally, as a proof of concept, the actin dynamic profiling was used to isolate MSCs via flow cytometry prior to sorting into three distinct sub-populations with low, intermediate or high actin dynamics. A greater fraction of MSCs with more rapid actin dynamics demonstrated increased inclination for adipogenesis, whereas, slower actin dynamics correlated with increased osteogenesis. Together, these results show that actin turnover can serve as a versatile biomarker to not only track cellular phenotypic heterogeneity but also harvest live cells with potential for differential phenotypic fates.
Collapse
Affiliation(s)
- Prakhar Mishra
- Molecular Biosciences Graduate Program in Cell and Developmental Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Ricky I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Nanxia Zhao
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
48
|
Desai D, Pethe P. Polycomb repressive complex 1: Regulators of neurogenesis from embryonic to adult stage. J Cell Physiol 2020; 235:4031-4045. [PMID: 31608994 DOI: 10.1002/jcp.29299] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/27/2019] [Indexed: 02/05/2023]
Abstract
Development of vertebrate nervous system is a complex process which involves differential gene expression and disruptions in this process or in the mature brain, may lead to neurological disorders and diseases. Extensive work that spanned several decades using rodent models and recent work on stem cells have helped uncover the intricate process of neuronal differentiation and maturation. There are various morphological changes, genetic and epigenetic modifications which occur during normal mammalian neural development, one of the chromatin modifications that controls vital gene expression are the posttranslational modifications on histone proteins, that controls accessibility of translational machinery. Among the histone modifiers, polycomb group proteins (PcGs), such as Ezh2, Eed and Suz12 form large protein complexes-polycomb repressive complex 2 (PRC2); while Ring1b and Bmi1 proteins form core of PRC1 along with accessory proteins such as Cbx, Hph, Rybp and Pcgfs catalyse histone modifications such as H3K27me3 and H2AK119ub1. PRC1 proteins are known to play critical role in X chromosome inactivation in females but they also repress the expression of key developmental genes and tightly regulate the mammalian neuronal development. In this review we have discussed the signalling pathways, morphogens and nuclear factors that initiate, regulate and maintain cells of the nervous system. Further, we have extensively reviewed the recent literature on the role of Ring1b and Bmi1 in mammalian neuronal development and differentiation; as well as highlighted questions that are still unanswered.
Collapse
Affiliation(s)
- Divya Desai
- Department of Biological Sciences, Sunandan Divatia School of Science (SDSOS), Narsee Monjee Institute of Management Studies (NMIMS) deemed-to-be University, Mumbai, India
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International University (SIU), Pune, India
| |
Collapse
|
49
|
Lukmanto D, Khanh VC, Shirota S, Kato T, Takasaki MM, Ohneda O. Dynamic Changes of Mouse Embryonic Stem Cell-Derived Neural Stem Cells Under In Vitro Prolonged Culture and Hypoxic Conditions. Stem Cells Dev 2019; 28:1434-1450. [DOI: 10.1089/scd.2019.0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Donny Lukmanto
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Vuong Cat Khanh
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Saori Shirota
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Toshiki Kato
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Mami Matsuo Takasaki
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Osamu Ohneda
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
50
|
Shparberg RA, Glover HJ, Morris MB. Modeling Mammalian Commitment to the Neural Lineage Using Embryos and Embryonic Stem Cells. Front Physiol 2019; 10:705. [PMID: 31354503 PMCID: PMC6637848 DOI: 10.3389/fphys.2019.00705] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/20/2019] [Indexed: 12/21/2022] Open
Abstract
Early mammalian embryogenesis relies on a large range of cellular and molecular mechanisms to guide cell fate. In this highly complex interacting system, molecular circuitry tightly controls emergent properties, including cell differentiation, proliferation, morphology, migration, and communication. These molecular circuits include those responsible for the control of gene and protein expression, as well as metabolism and epigenetics. Due to the complexity of this circuitry and the relative inaccessibility of the mammalian embryo in utero, mammalian neural commitment remains one of the most challenging and poorly understood areas of developmental biology. In order to generate the nervous system, the embryo first produces two pluripotent populations, the inner cell mass and then the primitive ectoderm. The latter is the cellular substrate for gastrulation from which the three multipotent germ layers form. The germ layer definitive ectoderm, in turn, is the substrate for multipotent neurectoderm (neural plate and neural tube) formation, representing the first morphological signs of nervous system development. Subsequent patterning of the neural tube is then responsible for the formation of most of the central and peripheral nervous systems. While a large number of studies have assessed how a competent neurectoderm produces mature neural cells, less is known about the molecular signatures of definitive ectoderm and neurectoderm and the key molecular mechanisms driving their formation. Using pluripotent stem cells as a model, we will discuss the current understanding of how the pluripotent inner cell mass transitions to pluripotent primitive ectoderm and sequentially to the multipotent definitive ectoderm and neurectoderm. We will focus on the integration of cell signaling, gene activation, and epigenetic control that govern these developmental steps, and provide insight into the novel growth factor-like role that specific amino acids, such as L-proline, play in this process.
Collapse
Affiliation(s)
| | | | - Michael B. Morris
- Embryonic Stem Cell Laboratory, Discipline of Physiology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|