Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.180
Filter
1.
Sci Rep ; 14(1): 15732, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977826

ABSTRACT

YAP plays a vital role in controlling growth and differentiation in various cell lineages. Although the expression of YAP in mice testicular and spermatogenic cells suggests its role in mammalian spermatogenesis, the role of YAP in the development of human male germ cells has not yet been determined. Using an in vitro model and a gene editing approach, we generated human spermatogonia stem cell-like cells (hSSLCs) from human embryonic stem cells (hESCs) and investigated the role of YAP in human spermatogenesis. The results showed that reducing YAP expression during the early stage of spermatogenic differentiation increased the number of PLZF+ hSSLCs and haploid spermatid-like cells. We also demonstrated that the up-regulation of YAP is essential for maintaining spermatogenic cell survival during the later stages of spermatogenic differentiation. The expression of YAP that deviates from this pattern results in a lower number of hSSLCs and an increased level of spermatogenic cell death. Taken together, our result demonstrates that the dynamic expression pattern of YAP is essential for human spermatogenesis. Modulating the level of YAP during human spermatogenesis could improve the production yield of male germ cells derived from hESCs, which could provide the optimization method for in vitro gametogenesis and gain insight into the application in the treatment of male infertility.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Human Embryonic Stem Cells , Spermatogenesis , Transcription Factors , YAP-Signaling Proteins , Male , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Spermatogonia/metabolism , Spermatogonia/cytology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Promyelocytic Leukemia Zinc Finger Protein/genetics
2.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38994775

ABSTRACT

Super-enhancers (SEs) are expansive regions of genomic DNA that regulate the expression of genes involved in cell identity and cell fate. We recently identified developmental stage- and cell type-specific modules within the murine Vsx2 SE. Here, we show that the human VSX2 SE modules have similar developmental stage- and cell type-specific activity in reporter gene assays. By inserting the human sequence of one VSX2 SE module into a mouse with microphthalmia, eye size was rescued. To understand the function of these SE modules during human retinal development, we deleted individual modules in human embryonic stem cells and generated retinal organoids. Deleting one module results in small organoids, recapitulating the small-eyed phenotype of mice with microphthalmia, while deletion of the other module led to disruptions in bipolar neuron development. This prototypical SE serves as a model for understanding developmental stage- and cell type-specific effects of neurogenic transcription factors with complex expression patterns. Moreover, by elucidating the gene regulatory mechanisms, we can begin to examine how dysregulation of these mechanisms contributes to phenotypic diversity and disease.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins , Retina , Transcription Factors , Animals , Humans , Mice , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Microphthalmos/genetics , Microphthalmos/pathology , Neurogenesis/genetics , Organoids/metabolism , Retina/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics
3.
Stem Cell Res Ther ; 15(1): 180, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38902824

ABSTRACT

BACKGROUND: Telomeres consist of repetitive DNA sequences at the chromosome ends to protect chromosomal stability, and primarily maintained by telomerase or occasionally by alternative telomere lengthening of telomeres (ALT) through recombination-based mechanisms. Additional mechanisms that may regulate telomere maintenance remain to be explored. Simultaneous measurement of telomere length and transcriptome in the same human embryonic stem cell (hESC) revealed that mRNA expression levels of UBQLN1 exhibit linear relationship with telomere length. METHODS: In this study, we first generated UBQLN1-deficient hESCs and compared with the wild-type (WT) hESCs the telomere length and molecular change at RNA and protein level by RNA-seq and proteomics. Then we identified the potential interacting proteins with UBQLN1 using immunoprecipitation-mass spectrometry (IP-MS). Furthermore, the potential mechanisms underlying the shortened telomeres in UBQLN1-deficient hESCs were analyzed. RESULTS: We show that Ubiquilin1 (UBQLN1) is critical for telomere maintenance in human embryonic stem cells (hESCs) via promoting mitochondrial function. UBQLN1 deficiency leads to oxidative stress, loss of proteostasis, mitochondria dysfunction, DNA damage, and telomere attrition. Reducing oxidative damage and promoting mitochondria function by culture under hypoxia condition or supplementation with N-acetylcysteine partly attenuate the telomere attrition induced by UBQLN1 deficiency. Moreover, UBQLN1 deficiency/telomere shortening downregulates genes for neuro-ectoderm lineage differentiation. CONCLUSIONS: Altogether, UBQLN1 functions to scavenge ubiquitinated proteins, preventing their overloading mitochondria and elevated mitophagy. UBQLN1 maintains mitochondria and telomeres by regulating proteostasis and plays critical role in neuro-ectoderm differentiation.


Subject(s)
Autophagy-Related Proteins , Human Embryonic Stem Cells , Mitochondria , Proteostasis , Telomere Homeostasis , Telomere , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Mitochondria/metabolism , Telomere/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Oxidative Stress , DNA Damage
4.
Stem Cell Reports ; 19(7): 993-1009, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38942028

ABSTRACT

Understanding the regulation of human embryonic stem cells (hESCs) pluripotency is critical to advance the field of developmental biology and regenerative medicine. Despite the recent progress, molecular events regulating hESC pluripotency, especially the transition between naive and primed states, still remain unclear. Here we show that naive hESCs display lower levels of O-linked N-acetylglucosamine (O-GlcNAcylation) than primed hESCs. O-GlcNAcase (OGA), the key enzyme catalyzing the removal of O-GlcNAc from proteins, is highly expressed in naive hESCs and is important for naive pluripotency. Depletion of OGA accelerates naive-to-primed pluripotency transition. OGA is transcriptionally regulated by EP300 and acts as a transcription regulator of genes important for maintaining naive pluripotency. Moreover, we profile protein O-GlcNAcylation of the two pluripotency states by quantitative proteomics. Together, this study identifies OGA as an important factor of naive pluripotency in hESCs and suggests that O-GlcNAcylation has a broad effect on hESCs homeostasis.


Subject(s)
Human Embryonic Stem Cells , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation , Acetylglucosamine/metabolism , beta-N-Acetylhexosaminidases/metabolism , Cell Line , Glycosylation
5.
Stem Cell Reports ; 19(7): 973-992, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38942030

ABSTRACT

Genetic differences between pluripotent stem cell lines cause variable activity of extracellular signaling pathways, limiting reproducibility of directed differentiation protocols. Here we used human embryonic stem cells (hESCs) to interrogate how exogenous factors modulate endogenous signaling events during specification of foregut endoderm lineages. We find that transforming growth factor ß1 (TGF-ß1) activates a putative human OTX2/LHX1 gene regulatory network which promotes anterior fate by antagonizing endogenous Wnt signaling. In contrast to Porcupine inhibition, TGF-ß1 effects cannot be reversed by exogenous Wnt ligands, suggesting that induction of SHISA proteins and intracellular accumulation of Fzd receptors render TGF-ß1-treated cells refractory to Wnt signaling. Subsequently, TGF-ß1-mediated inhibition of BMP and Wnt signaling suppresses liver fate and promotes pancreas fate. Furthermore, combined TGF-ß1 treatment and Wnt inhibition during pancreatic specification reproducibly and robustly enhance INSULIN+ cell yield across hESC lines. This modification of widely used differentiation protocols will enhance pancreatic ß cell yield for cell-based therapeutic applications.


Subject(s)
Bone Morphogenetic Proteins , Cell Differentiation , Endoderm , Human Embryonic Stem Cells , Wnt Signaling Pathway , Humans , Endoderm/cytology , Endoderm/metabolism , Cell Differentiation/drug effects , Wnt Signaling Pathway/drug effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Bone Morphogenetic Proteins/metabolism , Cell Lineage/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Cell Line , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
6.
Stem Cell Reports ; 19(6): 830-838, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38759646

ABSTRACT

The differentiation of human pluripotent stem cells into ventral mesencephalic dopaminergic (DA) fate is relevant for the treatment of Parkinson's disease. Shortcuts to obtaining DA cells through direct reprogramming often include forced expression of the transcription factor LMX1A. Although reprogramming with LMX1A can generate tyrosine hydroxylase (TH)-positive cells, their regional identity remains elusive. Using an in vitro model of early human neural tube patterning, we report that forced LMX1A expression induced a ventral-to-dorsal fate shift along the entire neuroaxis with the emergence of roof plate fates despite the presence of ventralizing molecules. The LMX1A-expressing progenitors gave rise to grafts containing roof plate-derived choroid plexus cysts as well as ectopically induced TH-positive neurons of a forebrain identity. Early activation of LMX1A prior to floor plate specification was necessary for the dorsalizing effect. Our work suggests using caution in employing LMX1A for the induction of DA fate, as this factor may generate roof plate rather than midbrain fates.


Subject(s)
Cell Differentiation , Dopaminergic Neurons , Human Embryonic Stem Cells , LIM-Homeodomain Proteins , Mesencephalon , Transcription Factors , Humans , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/cytology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Mesencephalon/cytology , Mesencephalon/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Body Patterning/genetics , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/genetics , Animals , Gene Expression Regulation, Developmental
7.
STAR Protoc ; 5(2): 103089, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38795356

ABSTRACT

Generating stable human embryonic stem cells (hESCs) with targeted genetic mutations allows for the interrogation of protein function in numerous cellular contexts while maintaining a relatively high degree of isogenicity. We describe a step-by-step protocol for generating knockout hESC lines with mutations in genes involved in synaptic transmission using CRISPR-Cas9. We describe steps for gRNA design, cloning, stem cell transfection, and clone isolation. We then detail procedures for gene knockout validation and differentiation of stem cells into functional induced neurons.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Human Embryonic Stem Cells , Neurons , Humans , CRISPR-Cas Systems/genetics , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Gene Editing/methods , Cell Differentiation/genetics , Gene Knockout Techniques/methods , RNA, Guide, CRISPR-Cas Systems/genetics , Synapses/metabolism , Synapses/genetics
8.
Stem Cell Reports ; 19(5): 729-743, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38701777

ABSTRACT

Embryonic stem cells (ESCs) are defined as stem cells with self-renewing and differentiation capabilities. These unique properties are tightly regulated and controlled by complex genetic and molecular mechanisms, whose understanding is essential for both basic and translational research. A large number of studies have mostly focused on understanding the molecular mechanisms governing pluripotency and differentiation of ESCs, while the regulation of proliferation has received comparably less attention. Here, we investigate the role of ZZZ3 (zinc finger ZZ-type containing 3) in human ESCs homeostasis. We found that knockdown of ZZZ3 negatively impacts ribosome biogenesis, translation, and mTOR signaling, leading to a significant reduction in cell proliferation. This process occurs without affecting pluripotency, suggesting that ZZZ3-depleted ESCs enter a "dormant-like" state and that proliferation and pluripotency can be uncoupled also in human ESCs.


Subject(s)
Cell Proliferation , Homeostasis , Human Embryonic Stem Cells , Ribosomes , Signal Transduction , TOR Serine-Threonine Kinases , Humans , TOR Serine-Threonine Kinases/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Ribosomes/metabolism , Cell Differentiation/genetics , Protein Biosynthesis
9.
Stem Cell Res ; 77: 103427, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38696852

ABSTRACT

The DNAJC19 gene, a member of DNAJ heat shock protein (Hsp40) family, is localized within the inner mitochondrial membrane (IMM) and plays a crucial role in regulating the function and localization of mitochondrial Hsp70 (MtHsp70). Mutations in the DNAJC19 gene cause Dilated Cardiomyopathy with Ataxia Syndrome (DCMA). The precise mechanisms underlying the DCMA phenotype caused by DNAJC19 mutations remain poorly understood, and effective treatment modalities were lacking unitl recently. By using CRISPR-Cas9 gene editing technology, this study generated a DNAJC19-knockout (DNAJC19-KO) human embryonic stem cell line (hESC), which will be a useful tool in studying the pathogenesis of DCMA.


Subject(s)
CRISPR-Cas Systems , HSP40 Heat-Shock Proteins , Human Embryonic Stem Cells , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Gene Knockout Techniques , Cell Line , Homozygote
10.
Stem Cell Res ; 77: 103436, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733811

ABSTRACT

Y chromosome deletion and karyotype abnormalities are commonly associated with congenital non-obstructive azoospermia, impairing spermatogenesis. Specifically, the deletion of the Y chromosome Azoospermia factor a (AZFa) has been identified in infertile males with severely impaired spermatogenesis. AZFa, encompassing megabase-scale of the Y chromosome region, poses challenges in modeling AZFa deletion-related male infertility using gene editing tools. Here, we successfully created an AZFa-deleted human embryonic stem cell line utilizing the CRISPR/Cas9 gene editing tool. Our analysis indicates the AZFa-deleted stem cell line holds promise for differentiation into ectoderm, mesoderm, and endoderm, highlighting its potential for further comprehensive study.


Subject(s)
Human Embryonic Stem Cells , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Male , Cell Line , Chromosomes, Human, Y/genetics , Cell Differentiation , CRISPR-Cas Systems , Chromosome Deletion , Gene Editing
11.
PLoS One ; 19(5): e0298274, 2024.
Article in English | MEDLINE | ID: mdl-38753762

ABSTRACT

The membrane peroxisomal proteins PEX11, play a crucial role in peroxisome proliferation by regulating elongation, membrane constriction, and fission of pre-existing peroxisomes. In this study, we evaluated the function of PEX11B gene in neural differentiation of human embryonic stem cell (hESC) by inducing shRNAi-mediated knockdown of PEX11B expression. Our results demonstrate that loss of PEX11B expression led to a significant decrease in the expression of peroxisomal-related genes including ACOX1, PMP70, PEX1, and PEX7, as well as neural tube-like structures and neuronal markers. Inhibition of SIRT1 using pharmacological agents counteracted the effects of PEX11B knockdown, resulting in a relative increase in PEX11B expression and an increase in differentiated neural tube-like structures. However, the neuroprotective effects of SIRT1 were eliminated by PPAR inhibition, indicating that PPARÉ£ may mediate the interaction between PEX11B and SIRT1. Our findings suggest that both SIRT1 and PPARÉ£ have neuroprotective effects, and also this study provides the first indication for a potential interaction between PEX11B, SIRT1, and PPARÉ£ during hESC neural differentiation.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells , Membrane Proteins , PPAR gamma , Sirtuin 1 , Humans , Sirtuin 1/metabolism , Sirtuin 1/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Cell Differentiation/drug effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Neurons/metabolism , Neurons/cytology , Neurons/drug effects , Cell Line , Peroxisomes/metabolism
12.
Cell Commun Signal ; 22(1): 300, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816763

ABSTRACT

Optimizing the efficiency of definitive endoderm (DE) differentiation is necessary for the generation of diverse organ-like structures. In this study, we used the small molecule inhibitor saracatinib (SAR) to enhance DE differentiation of human embryonic stem cells and induced pluripotent stem cells. SAR significantly improved DE differentiation efficiency at low concentrations. The interaction between SAR and Focal Adhesion Kinase (FAK) was explored through RNA-seq and molecular docking simulations, which further supported the inhibition of DE differentiation by p-FAK overexpression in SAR-treated cells. In addition, we found that SAR inhibited the nuclear translocation of Yes-associated protein (YAP), a downstream effector of FAK, which promoted DE differentiation. Moreover, the addition of SAR enabled a significant reduction in activin A (AA) from 50 to 10 ng/mL without compromising DE differentiation efficiency. For induction of the pancreatic lineage, 10 ng/ml AA combined with SAR at the DE differentiation stage yielded a comparative number of PDX1+/NKX6.1+ pancreatic progenitor cells to those obtained by 50 ng/ml AA treatment. Our study highlights SAR as a potential modulator that facilitates the cost-effective generation of DE cells and provides insight into the orchestration of cell fate determination.


Subject(s)
Benzodioxoles , Cell Differentiation , Endoderm , Quinazolines , Signal Transduction , Humans , Cell Differentiation/drug effects , Endoderm/drug effects , Endoderm/cytology , Endoderm/metabolism , Benzodioxoles/pharmacology , Signal Transduction/drug effects , Quinazolines/pharmacology , Transcription Factors/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Activins/metabolism , Molecular Docking Simulation
13.
Nat Cell Biol ; 26(6): 903-916, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38702503

ABSTRACT

Dynamic changes in mechanical microenvironments, such as cell crowding, regulate lineage fates as well as cell proliferation. Although regulatory mechanisms for contact inhibition of proliferation have been extensively studied, it remains unclear how cell crowding induces lineage specification. Here we found that a well-known oncogene, ETS variant transcription factor 4 (ETV4), serves as a molecular transducer that links mechanical microenvironments and gene expression. In a growing epithelium of human embryonic stem cells, cell crowding dynamics is translated into ETV4 expression, serving as a pre-pattern for future lineage fates. A switch-like ETV4 inactivation by cell crowding derepresses the potential for neuroectoderm differentiation in human embryonic stem cell epithelia. Mechanistically, cell crowding inactivates the integrin-actomyosin pathway and blocks the endocytosis of fibroblast growth factor receptors (FGFRs). The disrupted FGFR endocytosis induces a marked decrease in ETV4 protein stability through ERK inactivation. Mathematical modelling demonstrates that the dynamics of cell density in a growing human embryonic stem cell epithelium precisely determines the spatiotemporal ETV4 expression pattern and, consequently, the timing and geometry of lineage development. Our findings suggest that cell crowding dynamics in a stem cell epithelium drives spatiotemporal lineage specification using ETV4 as a key mechanical transducer.


Subject(s)
Cell Differentiation , Cell Lineage , Human Embryonic Stem Cells , Proto-Oncogene Proteins c-ets , Transcription Factors , Humans , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Endocytosis , Cell Proliferation , Integrins/metabolism , Integrins/genetics , Signal Transduction , Mechanotransduction, Cellular
14.
Genome Biol ; 25(1): 122, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38741214

ABSTRACT

BACKGROUND: Pluripotent states of embryonic stem cells (ESCs) with distinct transcriptional profiles affect ESC differentiative capacity and therapeutic potential. Although single-cell RNA sequencing has revealed additional subpopulations and specific features of naive and primed human pluripotent stem cells (hPSCs), the underlying mechanisms that regulate their specific transcription and that control their pluripotent states remain elusive. RESULTS: By single-cell analysis of high-resolution, three-dimensional (3D) genomic structure, we herein demonstrate that remodeling of genomic structure is highly associated with the pluripotent states of human ESCs (hESCs). The naive pluripotent state is featured with specialized 3D genomic structures and clear chromatin compartmentalization that is distinct from the primed state. The naive pluripotent state is achieved by remodeling the active euchromatin compartment and reducing chromatin interactions at the nuclear center. This unique genomic organization is linked to enhanced chromatin accessibility on enhancers and elevated expression levels of naive pluripotent genes localized to this region. In contradistinction, the primed state exhibits intermingled genomic organization. Moreover, active euchromatin and primed pluripotent genes are distributed at the nuclear periphery, while repressive heterochromatin is densely concentrated at the nuclear center, reducing chromatin accessibility and the transcription of naive genes. CONCLUSIONS: Our data provide insights into the chromatin structure of ESCs in their naive and primed states, and we identify specific patterns of modifications in transcription and chromatin structure that might explain the genes that are differentially expressed between naive and primed hESCs. Thus, the inversion or relocation of heterochromatin to euchromatin via compartmentalization is related to the regulation of chromatin accessibility, thereby defining pluripotent states and cellular identity.


Subject(s)
Pluripotent Stem Cells , Single-Cell Analysis , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Genome, Human , Euchromatin/genetics , Euchromatin/metabolism , Chromatin/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Heterochromatin/metabolism , Embryonic Stem Cells/metabolism , Chromatin Assembly and Disassembly
15.
Circulation ; 149(25): 1960-1979, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38752370

ABSTRACT

BACKGROUND: Cardiomyocyte differentiation involves a stepwise clearance of repressors and fate-restricting regulators through the modulation of BMP (bone morphogenic protein)/Wnt-signaling pathways. However, the mechanisms and how regulatory roadblocks are removed with specific developmental signaling pathways remain unclear. METHODS: We conducted a genome-wide CRISPR screen to uncover essential regulators of cardiomyocyte specification in human embryonic stem cells using a myosin heavy chain 6 (MYH6)-GFP (green fluorescence protein) reporter system. After an independent secondary single guide ribonucleic acid validation of 25 candidates, we identified NF2 (neurofibromin 2), a moesin-ezrin-radixin like (MERLIN) tumor suppressor, as an upstream driver of early cardiomyocyte lineage specification. Independent monoclonal NF2 knockouts were generated using CRISPR-Cas9, and cell states were inferred through bulk RNA sequencing and protein expression analysis across differentiation time points. Terminal lineage differentiation was assessed by using an in vitro 2-dimensional-micropatterned gastruloid model, trilineage differentiation, and cardiomyocyte differentiation. Protein interaction and post-translation modification of NF2 with its interacting partners were assessed using site-directed mutagenesis, coimmunoprecipitation, and proximity ligation assays. RESULTS: Transcriptional regulation and trajectory inference from NF2-null cells reveal the loss of cardiomyocyte identity and the acquisition of nonmesodermal identity. Sustained elevation of early mesoderm lineage repressor SOX2 and upregulation of late anticardiac regulators CDX2 and MSX1 in NF2 knockout cells reflect a necessary role for NF2 in removing regulatory roadblocks. Furthermore, we found that NF2 and AMOT (angiomotin) cooperatively bind to YAP (yes-associated protein) during mesendoderm formation, thereby preventing YAP activation, independent of canonical MST (mammalian sterile 20-like serine-threonine protein kinase)-LATS (large tumor suppressor serine-threonine protein kinase) signaling. Mechanistically, cardiomyocyte lineage identity was rescued by wild-type and NF2 serine-518 phosphomutants, but not NF2 FERM (ezrin-radixin-meosin homology protein) domain blue-box mutants, demonstrating that the critical FERM domain-dependent formation of the AMOT-NF2-YAP scaffold complex at the adherens junction is required for early cardiomyocyte lineage differentiation. CONCLUSIONS: These results provide mechanistic insight into the essential role of NF2 during early epithelial-mesenchymal transition by sequestering the repressive effect of YAP and relieving regulatory roadblocks en route to cardiomyocytes.


Subject(s)
Cell Differentiation , Cell Lineage , Myocytes, Cardiac , Neurofibromin 2 , Humans , Myocytes, Cardiac/metabolism , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , CRISPR-Cas Systems , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology
16.
Stem Cell Res ; 78: 103445, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38820864

ABSTRACT

Forkhead box protein J1 (FOXJ1), a member of the forkhead family, is an important transcription factor regulating multiciliated cell differentiation and motile ciliogenic program. Here, we established a FOXJ1- EGFP knock-in human embryonic stem cell (hESC) line by inserting a P2A-EGFP gene cassette of FOXJ1 using CRISPR/Cas9 system. The reporter cell line retained a normal karyotype, expressed comparable pluripotent marker genes, and maintained differentiation potential. This reporter cell line enables live identification of multiciliated cells during the general lung differentiation and will be a valuable tool for studying the multiciliated cell differentiation, ciliogenesis and mechanism of related pulmonary diseases.


Subject(s)
CRISPR-Cas Systems , Forkhead Transcription Factors , Human Embryonic Stem Cells , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Cell Line , Cell Differentiation , Gene Knock-In Techniques/methods , Gene Targeting/methods , Genes, Reporter
17.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38739758

ABSTRACT

The complicated process of neuronal development is initiated early in life, with the genetic mechanisms governing this process yet to be fully elucidated. Single-cell RNA sequencing (scRNA-seq) is a potent instrument for pinpointing biomarkers that exhibit differential expression across various cell types and developmental stages. By employing scRNA-seq on human embryonic stem cells, we aim to identify differentially expressed genes (DEGs) crucial for early-stage neuronal development. Our focus extends beyond simply identifying DEGs. We strive to investigate the functional roles of these genes through enrichment analysis and construct gene regulatory networks to understand their interactions. Ultimately, this comprehensive approach aspires to illuminate the molecular mechanisms and transcriptional dynamics governing early human brain development. By uncovering potential links between these DEGs and intelligence, mental disorders, and neurodevelopmental disorders, we hope to shed light on human neurological health and disease. In this study, we have used scRNA-seq to identify DEGs involved in early-stage neuronal development in hESCs. The scRNA-seq data, collected on days 26 (D26) and 54 (D54), of the in vitro differentiation of hESCs to neurons were analyzed. Our analysis identified 539 DEGs between D26 and D54. Functional enrichment of those DEG biomarkers indicated that the up-regulated DEGs participated in neurogenesis, while the down-regulated DEGs were linked to synapse regulation. The Reactome pathway analysis revealed that down-regulated DEGs were involved in the interactions between proteins located in synapse pathways. We also discovered interactions between DEGs and miRNA, transcriptional factors (TFs) and DEGs, and between TF and miRNA. Our study identified 20 significant transcription factors, shedding light on early brain development genetics. The identified DEGs and gene regulatory networks are valuable resources for future research into human brain development and neurodevelopmental disorders.


Subject(s)
Biomarkers , Brain , Gene Regulatory Networks , Human Embryonic Stem Cells , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Brain/metabolism , Brain/embryology , Brain/cytology , Biomarkers/metabolism , Neurons/metabolism , Neurons/cytology , Cell Differentiation/genetics , RNA-Seq , Neurogenesis/genetics , Gene Expression Regulation, Developmental , Gene Expression Profiling , Sequence Analysis, RNA/methods , Single-Cell Gene Expression Analysis
18.
Stem Cell Res Ther ; 15(1): 133, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704588

ABSTRACT

BACKGROUND: Human hematopoietic organoids have a wide application value for modeling human bone marrow diseases, such as acute hematopoietic radiation injury. However, the manufacturing of human hematopoietic organoids is an unaddressed challenge because of the complexity of hematopoietic tissues. METHODS: To manufacture hematopoietic organoids, we obtained CD34+ hematopoietic stem and progenitor cells (HSPCs) from human embryonic stem cells (hESCs) using stepwise induction and immunomagnetic bead-sorting. We then mixed these CD34+ HSPCs with niche-related cells in Gelatin-methacryloyl (GelMA) to form a three-dimensional (3D) hematopoietic organoid. Additionally, we investigated the effects of radiation damage and response to granulocyte colony-stimulating factor (G-CSF) in hematopoietic organoids. RESULTS: The GelMA hydrogel maintained the undifferentiated state of hESCs-derived HSPCs by reducing intracellular reactive oxygen species (ROS) levels. The established hematopoietic organoids in GelMA with niche-related cells were composed of HSPCs and multilineage blood cells and demonstrated the adherence of hematopoietic cells to niche cells. Notably, these hematopoietic organoids exhibited radiation-induced hematopoietic cell injury effect, including increased intracellular ROS levels, γ-H2AX positive cell percentages, and hematopoietic cell apoptosis percentages. Moreover, G-CSF supplementation in the culture medium significantly improved the survival of HSPCs and enhanced myeloid cell regeneration in these hematopoietic organoids after radiation. CONCLUSIONS: These findings substantiate the successful manufacture of a preliminary 3D hematopoietic organoid from hESCs-derived HSPCs, which was utilized for modeling hematopoietic radiation injury and assessing the radiation-mitigating effects of G-CSF in vitro. Our study provides opportunities to further aid in the standard and scalable production of hematopoietic organoids for disease modeling and drug testing.


Subject(s)
Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cells , Organoids , Humans , Organoids/metabolism , Organoids/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/cytology , Granulocyte Colony-Stimulating Factor/pharmacology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Regeneration/drug effects , Cell Differentiation/drug effects , Antigens, CD34/metabolism
19.
J Extracell Vesicles ; 13(5): e12445, 2024 May.
Article in English | MEDLINE | ID: mdl-38711334

ABSTRACT

Small extracellular vesicles (sEV) derived from various cell sources have been demonstrated to enhance cardiac function in preclinical models of myocardial infarction (MI). The aim of this study was to compare different sources of sEV for cardiac repair and determine the most effective one, which nowadays remains limited. We comprehensively assessed the efficacy of sEV obtained from human primary bone marrow mesenchymal stromal cells (BM-MSC), human immortalized MSC (hTERT-MSC), human embryonic stem cells (ESC), ESC-derived cardiac progenitor cells (CPC), human ESC-derived cardiomyocytes (CM), and human primary ventricular cardiac fibroblasts (VCF), in in vitro models of cardiac repair. ESC-derived sEV (ESC-sEV) exhibited the best pro-angiogenic and anti-fibrotic effects in vitro. Then, we evaluated the functionality of the sEV with the most promising performances in vitro, in a murine model of MI-reperfusion injury (IRI) and analysed their RNA and protein compositions. In vivo, ESC-sEV provided the most favourable outcome after MI by reducing adverse cardiac remodelling through down-regulating fibrosis and increasing angiogenesis. Furthermore, transcriptomic, and proteomic characterizations of sEV derived from hTERT-MSC, ESC, and CPC revealed factors in ESC-sEV that potentially drove the observed functions. In conclusion, ESC-sEV holds great promise as a cell-free treatment for promoting cardiac repair following MI.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Myocardial Infarction , Myocytes, Cardiac , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Humans , Animals , Mice , Myocardial Infarction/therapy , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Male , Myocardial Reperfusion Injury/therapy , Myocardial Reperfusion Injury/metabolism , Disease Models, Animal , Neovascularization, Physiologic , Cells, Cultured
20.
Stem Cell Res ; 77: 103438, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38776701

ABSTRACT

Here we present the generation of a human embryonic stem cell line with the potential to escape immune rejection upon transplantation to an alternate species, in this case sus scrofa. For in vivo detection the cells were modified by CRISPR-Cas9 to express human secreted alkaline phosphatase. To avoid immune recognition and subsequent rejection by host, genes encoding hB2M and hCIITA were knocked out and the porcine gene for CD47 was introduced. Upon editing and subsequent culture, cells maintained molecular and phenotypic pluripotent charactaristics and a normal karyotype supporting viability and functionality of the engineered cell line.


Subject(s)
CRISPR-Cas Systems , Human Embryonic Stem Cells , Animals , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Swine , Cell Line
SELECTION OF CITATIONS
SEARCH DETAIL
...