Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.365
Filter
1.
Methods Mol Biol ; 2848: 187-196, 2025.
Article in English | MEDLINE | ID: mdl-39240524

ABSTRACT

In several ocular diseases, degeneration of retinal neurons can lead to permanent blindness. Transplantation of stem cell (SC)-derived RGCs has been proposed as a potential therapy for RGC loss. Although there are reports of successful cases of SC-derived RGC transplantation, achieving long-distance regeneration and functional connectivity remains a challenge. To address these hurdles, retinal organoids are being used to study the regulatory mechanism of stem cell transplantation. Here we present a modified protocol for differentiating human embryonic stem cells (ESCs) into retinal organoids and transplanting organoid-derived RGCs into the murine eyes.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells , Retinal Ganglion Cells , Humans , Animals , Mice , Human Embryonic Stem Cells/cytology , Retinal Ganglion Cells/cytology , Stem Cell Transplantation/methods , Organoids/cytology , Organoids/transplantation , Cell Culture Techniques/methods , Cell- and Tissue-Based Therapy/methods , Retina/cytology , Embryonic Stem Cells/cytology
2.
Nat Commun ; 15(1): 8539, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358345

ABSTRACT

The primed epiblast acts as a transitional stage between the relatively homogeneous naïve epiblast and the gastrulating embryo. Its formation entails coordinated changes in regulatory circuits driven by transcription factors and epigenetic modifications. Using a multi-omic approach in human embryonic stem cell models across the spectrum of peri-implantation development, we demonstrate that the transcription factors ZIC2 and ZIC3 have overlapping but essential roles in opening primed-specific enhancers. Together, they are essential to facilitate progression to and maintain primed pluripotency. ZIC2/3 accomplish this by recruiting SWI/SNF to chromatin and loss of ZIC2/3 or degradation of SWI/SNF both prevent enhancer activation. Loss of ZIC2/3 also results in transcriptome changes consistent with perturbed Polycomb activity and a shift towards the expression of genes linked to differentiation towards the mesendoderm. Additionally, we find an intriguing dependency on the transcriptional machinery for sustained recruitment of ZIC2/3 over a subset of primed-hESC specific enhancers. Taken together, ZIC2 and ZIC3 regulate highly dynamic lineage-specific enhancers and collectively act as key regulators of human primed pluripotency.


Subject(s)
Chromosomal Proteins, Non-Histone , Homeodomain Proteins , Human Embryonic Stem Cells , Nuclear Proteins , Pluripotent Stem Cells , Transcription Factors , Germ Layers/cytology , Germ Layers/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromatin/metabolism , DNA Helicases/metabolism , Enhancer Elements, Genetic , Cell Plasticity , Chromatin Assembly and Disassembly , Transcription, Genetic , Embryonic Development
3.
Nat Commun ; 15(1): 8711, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379383

ABSTRACT

Mediator, a co-regulator complex required for RNA Polymerase II activity, interacts with tissue-specific transcription factors to regulate development and maintain homeostasis. We observe reduced Mediator subunit MED15 expression in endocrine hormone-producing pancreatic islets isolated from people living with type 2 diabetes and sought to understand how MED15 and Mediator control gene expression programs important for the function of insulin-producing ß-cells. Here we show that Med15 is expressed during mouse ß-cell development and maturation. Knockout of Med15 in mouse ß-cells causes defects in ß-cell maturation without affecting ß-cell mass or insulin expression. ChIP-seq and co-immunoprecipitation analyses found that Med15 binds ß-cell transcription factors Nkx6-1 and NeuroD1 to regulate key ß-cell maturation genes. In support of a conserved role during human development, human embryonic stem cell-derived ß-like cells, genetically engineered to express high levels of MED15, express increased levels of maturation markers. We provide evidence of a conserved role for Mediator in ß-cell maturation and demonstrate an additional layer of control that tunes ß-cell transcription factor function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Diabetes Mellitus, Type 2 , Homeodomain Proteins , Insulin-Secreting Cells , Mediator Complex , Mice, Knockout , Insulin-Secreting Cells/metabolism , Animals , Humans , Mediator Complex/metabolism , Mediator Complex/genetics , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Cell Differentiation , Male , Female , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Insulin/metabolism , Adult , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Mice, Inbred C57BL
4.
Nat Commun ; 15(1): 8570, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384784

ABSTRACT

In Multiple Sclerosis (MS), inflammatory demyelinated lesions in the brain and spinal cord lead to neurodegeneration and progressive disability. Remyelination can restore fast saltatory conduction and neuroprotection but is inefficient in MS especially with increasing age, and is not yet treatable with therapies. Intrinsic and extrinsic inhibition of oligodendrocyte progenitor cell (OPC) function contributes to remyelination failure, and we hypothesised that the transplantation of 'improved' OPCs, genetically edited to overcome these obstacles, could improve remyelination. Here, we edit human(h) embryonic stem cell-derived OPCs to be unresponsive to a chemorepellent released from chronic MS lesions, and transplant them into rodent models of chronic lesions. Edited hOPCs display enhanced migration and remyelination compared to controls, regardless of the host age and length of time post-transplant. We show that genetic manipulation and transplantation of hOPCs overcomes the negative environment inhibiting remyelination, with translational implications for therapeutic strategies for people with progressive MS.


Subject(s)
Multiple Sclerosis , Oligodendrocyte Precursor Cells , Remyelination , Animals , Remyelination/genetics , Humans , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/cytology , Multiple Sclerosis/therapy , Multiple Sclerosis/genetics , CRISPR-Cas Systems , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Rats , Mice , Gene Editing/methods , Disease Models, Animal , Myelin Sheath/metabolism , Female , Oligodendroglia/cytology , Oligodendroglia/metabolism , Male , Cell Movement/genetics , Encephalomyelitis, Autoimmune, Experimental/therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Cell Differentiation
5.
Stem Cell Res Ther ; 15(1): 344, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39380037

ABSTRACT

BACKGROUND: Lung injury and pulmonary fibrosis (PF), frequently arising as sequelae of severe and acute lung disease, currently face a dearth of effective therapeutic potions. Mesenchymal stem cells (MSCs) with immunomodulatory and tissue repair functions have immense potential to treat lung injury and PF. However, the optimal route of administration, timing, and frequency of dosing remain elusive. Human embryonic stem cell-derived immunity-and-matrix-regulatory cells (IMRCs) have shown therapeutic potential for lung injury and PF. METHODS: To ascertain the optimal therapeutic regimen for IMRCs in PF, we conducted an experimental study. Utilizing a mouse model of PF induced by bleomycin (BLM), IMRCs were administered via either a single or double intravenous (IV) or intratracheal (IT) injection on the first and seventh days post-BLM induction. RESULTS: Our findings revealed that IV infusion of IMRCs surpassed IT infusion in enhancing survival rates, facilitating body weight recovery, and optimizing Ashcroft and Szapiel scores among the model mice. Notably, IV administration exhibited a more profound ability to mitigate lung inflammation and fibrosis. Moreover, earlier and more frequent administrations of IMRCs were found to be advantageous in enhancing their therapeutic effects. Specifically, early administration with two IV infusions significantly improved body weight, lung organ coefficient, pulmonary ventilation and diffusion functions, and PF. This was accompanied by an increase in alveolar type I and II epithelial cells and a suppression of macrophage infiltration via CD24. CONCLUSION: Collectively, these results suggested that IMRCs infusion ameliorated lung injury by promoting lung regeneration and inhibiting macrophage infiltration in a route, time, and frequency-dependent manner.


Subject(s)
Bleomycin , Human Embryonic Stem Cells , Lung Injury , Pulmonary Fibrosis , Animals , Mice , Humans , Human Embryonic Stem Cells/cytology , Pulmonary Fibrosis/therapy , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/chemically induced , Lung Injury/therapy , Lung Injury/pathology , Disease Models, Animal , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL
6.
STAR Protoc ; 5(3): 103297, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39243376

ABSTRACT

Gene editing of human pluripotent stem cells is a promising approach for developing targeted gene therapies for metabolic diseases. Here, we present a protocol for generating a CRISPR-Cas12a gene knockout of protein tyrosine phosphatases in human embryonic stem cells. We describe steps for differentiating the edited clones into pancreatic islet-like spheroids rich in ß-like cells. We then detail procedures for implanting these spheroids under the murine kidney capsule for in vivo maturation.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Insulin-Secreting Cells , Pluripotent Stem Cells , Protein Tyrosine Phosphatases , Humans , Gene Editing/methods , CRISPR-Cas Systems/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Mice , Animals , Cell Differentiation/genetics , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism
7.
Cell Rep ; 43(9): 114700, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39235941

ABSTRACT

Genomic imprinting involves differential DNA methylation and gene expression between homologous paternal and maternal loci. It remains unclear, however, whether DNA replication also shows parent-of-origin-specific patterns at imprinted or other genomic regions. Here, we investigate genome-wide asynchronous DNA replication utilizing uniparental human embryonic stem cells containing either maternal-only (parthenogenetic) or paternal-only (androgenetic) DNA. Four clusters of imprinted genes exhibited differential replication timing based on parent of origin, while the remainder of the genome, 99.82%, showed no significant replication asynchrony between parental origins. Active alleles in imprinted gene clusters replicated earlier than their inactive counterparts. At the Prader-Willi syndrome locus, replication asynchrony spanned virtually the entirety of S phase. Replication asynchrony was carried through differentiation to neuronal precursor cells in a manner consistent with gene expression. This study establishes asynchronous DNA replication as a hallmark of large imprinted gene clusters.


Subject(s)
DNA Replication Timing , Genomic Imprinting , Humans , DNA Methylation/genetics , Cell Differentiation/genetics , DNA Replication/genetics , Human Embryonic Stem Cells/metabolism , Multigene Family , Prader-Willi Syndrome/genetics , Alleles
8.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39250534

ABSTRACT

During the first week of development, human embryos form a blastocyst composed of an inner cell mass and trophectoderm (TE) cells, the latter of which are progenitors of placental trophoblast. Here, we investigated the expression of transcripts in the human TE from early to late blastocyst stages. We identified enrichment of the transcription factors GATA2, GATA3, TFAP2C and KLF5 and characterised their protein expression dynamics across TE development. By inducible overexpression and mRNA transfection, we determined that these factors, together with MYC, are sufficient to establish induced trophoblast stem cells (iTSCs) from primed human embryonic stem cells. These iTSCs self-renew and recapitulate morphological characteristics, gene expression profiles, and directed differentiation potential, similar to existing human TSCs. Systematic omission of each, or combinations of factors, revealed the crucial importance of GATA2 and GATA3 for iTSC transdifferentiation. Altogether, these findings provide insights into the transcription factor network that may be operational in the human TE and broaden the methods for establishing cellular models of early human placental progenitor cells, which may be useful in the future to model placental-associated diseases.


Subject(s)
Cell Transdifferentiation , Transcription Factors , Trophoblasts , Humans , Trophoblasts/cytology , Trophoblasts/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , GATA3 Transcription Factor/metabolism , GATA3 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , GATA2 Transcription Factor/genetics , Female , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , Blastocyst/metabolism , Blastocyst/cytology , Pregnancy , Cell Differentiation
9.
Stem Cell Res Ther ; 15(1): 274, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218930

ABSTRACT

BACKGROUND: Understanding the lineage differentiation of human prostate not only is crucial for basic research on human developmental biology but also significantly contributes to the management of prostate-related disorders. Current knowledge mainly relies on studies on rodent models, lacking human-derived alternatives despite clinical samples may provide a snapshot at certain stage. Human embryonic stem cells can generate all the embryonic lineages including the prostate, and indeed a few studies demonstrate such possibility based on co-culture or co-transplantation with urogenital mesenchyme into mouse renal capsule. METHODS: To establish a stepwise protocol to obtain prostatic organoids in vitro from human embryonic stem cells, we apply chemicals and growth factors by mimicking the regulation network of transcription factors and signal transduction pathways, and construct cell lines carrying an inducible NKX3-1 expressing cassette, together with three-dimensional culture system. Unpaired t test was applied for statistical analyses. RESULTS: We first successfully generate the definitive endoderm, hindgut, and urogenital sinus cells. The embryonic stem cell-derived urogenital sinus cells express prostatic key transcription factors AR and FOXA1, but fail to express NKX3-1. Therefore, we construct NKX3-1-inducible cell line by homologous recombination, which is eventually able to yield AR, FOXA1, and NKX3-1 triple-positive urogenital prostatic lineage cells through stepwise differentiation. Finally, combined with 3D culture we successfully derive prostate-like organoids with certain structures and prostatic cell populations. CONCLUSIONS: This study reveals the crucial role of NKX3-1 in prostatic differentiation and offers the inducible NKX3-1 cell line, as well as provides a stepwise differentiation protocol to generate human prostate-like organoids, which should facilitate the studies on prostate development and disease pathogenesis.


Subject(s)
Cell Differentiation , Cell Lineage , Homeodomain Proteins , Human Embryonic Stem Cells , Prostate , Transcription Factors , Humans , Prostate/cytology , Prostate/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Male , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Organoids/metabolism , Organoids/cytology , Mice , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Animals , Cell Line
10.
Cells ; 13(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273019

ABSTRACT

Epidermal transplantation is a common and widely used surgical technique in clinical medicine. Derivatives of embryonic stem cells have the potential to serve as a source of transplantable cells. However, allograft rejection is one of the main challenges. To investigate the immunogenicity of keratinocytes derived from human embryonic stem cells (ESKCs), we conducted a series of in vivo and in vitro experiments. The results showed that ESKCs have low HLA molecule expression, limited antigen presentation capabilities, and a weak ability to stimulate the proliferation and secretion of inflammatory factors in allogeneic PBMCs in vitro. In humanized immune mouse models, ESKCs elicited weak transplant rejection responses in the host. Overall, we found that ESKCs have low immunogenicity and may have potential applications in the field of regenerative medicine.


Subject(s)
Human Embryonic Stem Cells , Keratinocytes , Humans , Keratinocytes/immunology , Keratinocytes/metabolism , Keratinocytes/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/immunology , Human Embryonic Stem Cells/metabolism , Animals , Mice , Cell Proliferation , Graft Rejection/immunology , HLA Antigens/immunology , HLA Antigens/metabolism
11.
Sci Rep ; 14(1): 20565, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39232000

ABSTRACT

Studies on MECP2 function and its implications in Rett Syndrome (RTT) have traditionally centered on neurons. Here, using human embryonic stem cell (hESC) lines, we modeled MECP2 loss-of-function to explore its effects on astrocyte (AST) development and dysfunction in the brain. Ultrastructural analysis of RTT hESC-derived cerebral organoids revealed significantly smaller mitochondria compared to controls (CTRs), particularly pronounced in glia versus neurons. Employing a multiomics approach, we observed increased gene expression and accessibility of a subset of nuclear-encoded mitochondrial genes upon mutation of MECP2 in ASTs compared to neurons. Analysis of hESC-derived ASTs showed reduced mitochondrial respiration and altered key proteins in the tricarboxylic acid cycle and electron transport chain in RTT versus CTRs. Additionally, RTT ASTs exhibited increased cytosolic amino acids under basal conditions, which were depleted upon increased energy demands. Notably, mitochondria isolated from RTT ASTs exhibited increased reactive oxygen species and influenced neuronal activity when transferred to cortical neurons. These findings underscore MECP2 mutation's differential impact on mitochondrial and metabolic pathways in ASTs versus neurons, suggesting that dysfunctional AST mitochondria may contribute to RTT pathophysiology by affecting neuronal health.


Subject(s)
Astrocytes , Methyl-CpG-Binding Protein 2 , Mitochondria , Mutation , Neurons , Reactive Oxygen Species , Rett Syndrome , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Mitochondria/metabolism , Astrocytes/metabolism , Reactive Oxygen Species/metabolism , Humans , Neurons/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Rett Syndrome/pathology , Human Embryonic Stem Cells/metabolism , Cell Line
12.
Stem Cell Res Ther ; 15(1): 298, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267174

ABSTRACT

BACKGROUND: Cardiovascular progenitor cells (CPCs) derived from human embryonic stem cells (hESCs) are considered valuable cell sources for investigating cardiovascular physiology in vitro. Meeting the diverse needs of this application requires the large-scale production of CPCs in an in vitro environment. This study aimed to use an effective culture system utilizing signaling factors for the large-scale expansion of hESC-derived CPCs with the potential to differentiate into functional cardiac lineage cells. METHODS AND RESULTS: Initially, CPCs were generated from hESCs using a 4-day differentiation protocol with a combination of four small molecules (CHIR99021, IWP2, SB-431542, and purmorphamine). These CPCs were then expanded and maintained in a medium containing three factors (bFGF, CHIR, and A83-01), resulting in a > 6,000-fold increase after 8 passages. These CPCs were successfully cryopreserved for an extended period in late passages. The expanded CPCs maintained their gene and protein expression signatures as well as their differentiation capacity through eight passages. Additionally, these CPCs could differentiate into four types of cardiac lineage cells: cardiomyocytes, endothelial cells, smooth muscle cells, and fibroblasts, demonstrating appropriate functionality. Furthermore, the coculture of these CPC-derived cardiovascular lineage cells in rat tail collagen resulted in cardiac microtissue formation, highlighting the potential of this 3D platform for studying cardiovascular physiology in vitro. CONCLUSION: In conclusion, expandable hESC-derived CPCs demonstrated the ability to self-renewal and differentiation into functional cardiovascular lineage cells consistently across passages, which may apply as potential cell sources for in vitro cardiovascular studies.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Animals , Rats , Cell Lineage , Cells, Cultured
13.
Cancer Immunol Immunother ; 73(11): 231, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261363

ABSTRACT

CD98, also known as SLC3A2, is a multifunctional cell surface molecule consisting of amino acid transporters. CD98 is ubiquitously expressed in many types of tissues, but expressed at higher levels in cancerous tissues than in normal tissues. CD98 is also upregulated in most hepatocellular carcinoma (HCC) patients; however, the function of CD98 in HCC cells has been little studied. In this study, we generated a panel of monoclonal antibodies (MAbs) against surface proteins on human embryonic stem cells (hESCs). NPB15, one of the MAbs, bound to hESCs and various cancer cells, including HCC cells and non-small cell lung carcinoma (NSCLC) cells, but not to peripheral blood mononuclear cells (PBMCs) and primary hepatocytes. Immunoprecipitation and mass spectrometry identified the target antigen of NPB15 as CD98. CD98 depletion decreased cell proliferation, clonogenic survival, and migration and induced apoptosis in HCC cells. In addition, CD98 depletion decreased the expression of cancer stem cell (CSC) markers in HCC cells. In tumorsphere cultures, the expression of CD98 interacting with NPB15 was significantly increased, as were known CSC markers. After cell sorting by NPB15, cells with high expression of CD98 (CD98-high) showed higher clonogenic survival than cells with low expression of CD98 (CD98-low) in HCC cells, suggesting CD98 as a potential CSC marker on HCC cells. The chimeric version of NPB15 was able to induce antibody-dependent cellular cytotoxicity (ADCC) against HCC cells in vitro. NPB15 injection showed antitumor activity in an HCC xenograft mouse model. The results suggest that NPB15 may be developed as a therapeutic antibody for HCC patients.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Hepatocellular , Fusion Regulatory Protein-1 , Liver Neoplasms , Xenograft Model Antitumor Assays , Humans , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Animals , Mice , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/immunology , Fusion Regulatory Protein-1/metabolism , Fusion Regulatory Protein-1/immunology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/immunology , Cell Proliferation , Cell Line, Tumor , Apoptosis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Fusion Regulatory Protein 1, Heavy Chain
14.
Sci Rep ; 14(1): 19522, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174599

ABSTRACT

Individuals with the Autism Susceptibility Candidate 2 (AUTS2) gene disruptions exhibit symptoms such as intellectual disability, microcephaly, growth retardation, and distinct skeletal and facial differences. The role of AUTS2 in neurodevelopment has been investigated using animal and embryonic stem cell models. However, the precise molecular mechanisms of how AUTS2 influences neurodevelopment, particularly in humans, are not thoroughly understood. Our study employed a 3D human cerebral organoid culture system, in combination with genetic, genomic, cellular, and molecular approaches, to investigate how AUTS2 impacts neurodevelopment through cellular signaling pathways. We used CRISPR/Cas9 technology to create AUTS2-deficient human embryonic stem cells and then generated cerebral organoids with these cells. Our transcriptomic analyses revealed that the absence of AUTS2 in cerebral organoids reduces the populations of cells committed to the neuronal lineage, resulting in an overabundance of cells with a transcription profile resembling that of choroid plexus (ChP) cells. Intriguingly, we found that AUTS2 negatively regulates the WNT/ß-catenin signaling pathway, evidenced by its overactivation in AUTS2-deficient cerebral organoids and in luciferase reporter cells lacking AUTS2. Importantly, treating the AUTS2-deficient cerebral organoids with a WNT inhibitor reversed the overexpression of ChP genes and increased the downregulated neuronal gene expression. This study offers new insights into the role of AUTS2 in neurodevelopment and suggests potential targeted therapies for neurodevelopmental disorders.


Subject(s)
Cell Differentiation , Cytoskeletal Proteins , Neurons , Organoids , Transcription Factors , Wnt Signaling Pathway , Humans , Organoids/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , CRISPR-Cas Systems , beta Catenin/metabolism , beta Catenin/genetics , Human Embryonic Stem Cells/metabolism , Brain/metabolism , Brain/pathology
15.
Nat Commun ; 15(1): 7186, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169036

ABSTRACT

Nucleoli are fundamentally essential sites for ribosome biogenesis in cells and formed by liquid-liquid phase separation (LLPS) for a multilayer condensate structure. How the nucleoli integrity is maintained remains poorly understood. Here, we reveal that METTL3/METTL14, the typical methyltransferase complex catalyzing N6-methyladnosine (m6A) on mRNAs maintain nucleoli integrity in human embryonic stem cells (hESCs). METTL3/METTL14 deficiency impairs nucleoli and leads to the complete loss of self-renewal in hESCs. We further show that SUV39H1/H2 protein, the methyltransferases catalyzing H3K9me3 were dramatically elevated in METTL3/METTL14 deficient cells, which causes an accumulation and infiltration of H3K9me3 across the whole nucleolus and impairs the LLPS. Mechanistically, METTL3/METTL14 complex serves as an essential adapter for CRL4 E3 ubiquitin ligase targeting SUV39H1/H2 for polyubiquitination and proteasomal degradation and therefore prevents H3K9me3 accumulation in nucleoli. Together, these findings uncover a previously unknown role of METTL3/METTL14 to maintain nucleoli integrity by facilitating SUV39H1/H2 degradation in human cells.


Subject(s)
Cell Nucleolus , Methyltransferases , Repressor Proteins , Humans , Methyltransferases/metabolism , Methyltransferases/genetics , Cell Nucleolus/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Histones/metabolism , Ubiquitination , Human Embryonic Stem Cells/metabolism , Proteolysis , HEK293 Cells , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Histone-Lysine N-Methyltransferase
16.
STAR Protoc ; 5(3): 103263, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39128010

ABSTRACT

Thyroid carcinoma represents the first malignancy among the endocrine organs. Investigating the cellular hierarchy and the mechanisms underlying the initiation of thyroid carcinoma is crucial in thyroid cancer research. Here, we present a protocol for deriving thyroid cell lineage from human embryonic stem cells. We also describe steps for engineering thyroid progenitor cells utilizing CRISPR-Cas9 technology, which can be used to perform in vivo studies, thus facilitating the development of representative thyroid tumorigenesis models. For complete details on the use and execution of this protocol, please refer to Veschi et al.1.


Subject(s)
CRISPR-Cas Systems , Cell Lineage , Gene Editing , Thyroid Gland , Thyroid Neoplasms , Humans , CRISPR-Cas Systems/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Gene Editing/methods , Thyroid Gland/pathology , Thyroid Gland/cytology , Thyroid Gland/metabolism , Cell Lineage/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Disease Progression
17.
Mol Metab ; 89: 102017, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39182839

ABSTRACT

OBJECTIVE: Human embryonic stem cell (hESC; SC)-derived pancreatic ß cells can be used to study diabetes pathologies and develop cell replacement therapies. Although current differentiation protocols yield SCß cells with varying degrees of maturation, these cells still differ from deceased donor human ß cells in several respects. We sought to develop a reporter cell line that could be used to dynamically track SCß cell functional maturation. METHODS: To monitor SCß cell maturation in vitro, we created an IAPP-2A-mScar and INSULIN-2A-EGFP dual fluorescent reporter (INS2A-EGFP/+;IAPP2A-mScarlet/+) hESC line using CRISPR/Cas9. Pluripotent SC were then differentiated using a 7-stage protocol to islet-like cells. Immunohistochemistry, flow cytometry, qPCR, GSIS and electrophysiology were used to characterise resulting cell populations. RESULTS: We observed robust expression of EGFP and mScarlet fluorescent proteins in insulin- and IAPP-expressing cells without any compromise to their differentiation. We show that the proportion of insulin-producing cells expressing IAPP increases over a 4-week maturation period, and that a subset of insulin-expressing cells remain IAPP-free. Compared to this IAPP-free population, we show these insulin- and IAPP-expressing cells are less polyhormonal, more glucose-sensitive, and exhibit decreased action potential firing in low (2.8 mM) glucose. CONCLUSIONS: The INS2A-EGFP/+;IAPP2A-mScarlet/+ hESC line provides a useful tool for tracking populations of maturing hESC-derived ß cells in vitro. This tool has already been shared with 3 groups and is freely available to all.


Subject(s)
Cell Differentiation , Insulin-Secreting Cells , Insulin , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Genes, Reporter , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/genetics , Cell Line , CRISPR-Cas Systems
18.
Nucleic Acids Res ; 52(18): 10879-10896, 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39166492

ABSTRACT

N6-methyladenonsine (m6A) is ubiquitously distributed in mammalian mRNA. However, the precise involvement of m6A in early development has yet to be fully elucidated. Here, we report that deletion of the m6A demethylase ALKBH5 in human embryonic stem cells (hESCs) severely impairs definitive endoderm (DE) differentiation. ALKBH5-/- hESCs fail to undergo the primitive streak (PS) intermediate transition that precedes endoderm specification. Mechanistically, we show that ALKBH5 deficiency induces m6A hypermethylation around the 3' untranslated region (3'UTR) of GATA6 transcripts and destabilizes GATA6 mRNA in a YTHDF2-dependent manner. Moreover, GATA6 binds to the promoters of critical regulatory genes involved in Wnt/ß-catenin signaling transduction, including the canonical Wnt antagonist DKK1 and DKK4, which are unexpectedly repressed upon the dysregulation of GATA6 mRNA metabolism. Remarkably, DKK1 and DKK4 both exhibit a pleiotropic effect in modulating the Wnt/ß-catenin cascade and guard the endogenous signaling activation underlying DE formation as potential downstream targets of the ALKBH5-GATA6 regulation. Here, we unravel a role of ALKBH5 in human endoderm formation in vitro by modulating the canonical Wnt signaling logic through the previously unrecognized functions of DKK1/4, thus capturing a more comprehensive role of m6A in early human embryogenesis.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Cell Differentiation , Endoderm , Intercellular Signaling Peptides and Proteins , Wnt Signaling Pathway , Humans , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , Endoderm/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Cell Differentiation/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , 3' Untranslated Regions , GATA6 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , beta Catenin/metabolism , beta Catenin/genetics , Gene Expression Regulation, Developmental , Cell Line , RNA, Messenger/metabolism , RNA, Messenger/genetics , Promoter Regions, Genetic
19.
Cells ; 13(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39195283

ABSTRACT

Human pluripotent stem cells (hPSCs) are pivotal in regenerative medicine, yet their in vitro expansion often leads to genetic abnormalities, raising concerns about their safety in clinical applications. This study analyzed ten human embryonic stem cell lines across multiple passages to elucidate the dynamics of chromosomal abnormalities and single-nucleotide variants (SNVs) in 380 cancer-related genes. Prolonged in vitro culture resulted in 80% of the lines acquiring gains of chromosome 20q or 1q, both known for conferring an in vitro growth advantage. 70% of lines also acquired other copy number variants (CNVs) outside the recurrent set. Additionally, we detected 122 SNVs in 88 genes, with all lines acquiring at least one de novo SNV during culture. Our findings showed higher loads of both CNVs and SNVs at later passages, which were due to the cumulative acquisition of mutations over a longer time in culture, and not to an increased rate of mutagenesis over time. Importantly, we observed that SNVs and rare CNVs followed the acquisition of chromosomal gains in 1q and 20q, while most of the low-passage and genetically balanced samples were devoid of cancer-associated mutations. This suggests that recurrent chromosomal abnormalities are potential drivers for the acquisition of other mutations.


Subject(s)
Chromosome Aberrations , DNA Copy Number Variations , Mutation , Neoplasms , Pluripotent Stem Cells , Humans , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Pluripotent Stem Cells/metabolism , DNA Copy Number Variations/genetics , Polymorphism, Single Nucleotide/genetics , Cell Line , Human Embryonic Stem Cells/metabolism , Cell Culture Techniques/methods
20.
Genome Biol ; 25(1): 211, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118163

ABSTRACT

BACKGROUND: The Pharyngeal Endoderm (PE) is an extremely relevant developmental tissue, serving as the progenitor for the esophagus, parathyroids, thyroids, lungs, and thymus. While several studies have highlighted the importance of PE cells, a detailed transcriptional and epigenetic characterization of this important developmental stage is still missing, especially in humans, due to technical and ethical constraints pertaining to its early formation. RESULTS: Here we fill this knowledge gap by developing an in vitro protocol for the derivation of PE-like cells from human Embryonic Stem Cells (hESCs) and by providing an integrated multi-omics characterization. Our PE-like cells robustly express PE markers and are transcriptionally homogenous and similar to in vivo mouse PE cells. In addition, we define their epigenetic landscape and dynamic changes in response to Retinoic Acid by combining ATAC-Seq and ChIP-Seq of histone modifications. The integration of multiple high-throughput datasets leads to the identification of new putative regulatory regions and to the inference of a Retinoic Acid-centered transcription factor network orchestrating the development of PE-like cells. CONCLUSIONS: By combining hESCs differentiation with computational genomics, our work reveals the epigenetic dynamics that occur during human PE differentiation, providing a solid resource and foundation for research focused on the development of PE derivatives and the modeling of their developmental defects in genetic syndromes.


Subject(s)
Cell Differentiation , Endoderm , Epigenesis, Genetic , Human Embryonic Stem Cells , Humans , Endoderm/cytology , Endoderm/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Pharynx/cytology , Pharynx/metabolism , Tretinoin/pharmacology , Tretinoin/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Transcription Factors/genetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL