Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 12: 1343106, 2024.
Article in English | MEDLINE | ID: mdl-38371924

ABSTRACT

Tissue repair is significantly compromised in the aging human body resulting in critical disease conditions (such as myocardial infarction or Alzheimer's disease) and imposing a tremendous burden on global health. Reprogramming approaches (partial or direct reprogramming) are considered fruitful in addressing this unmet medical need. However, the efficacy, cellular maturity and specific targeting are still major challenges of direct reprogramming. Here we describe novel approaches in direct reprogramming that address these challenges. Extracellular signaling pathways (Receptor tyrosine kinases, RTK and Receptor Serine/Theronine Kinase, RSTK) and epigenetic marks remain central in rewiring the cellular program to determine the cell fate. We propose that modern protein design technologies (AI-designed minibinders regulating RTKs/RSTK, epigenetic enzymes, or pioneer factors) have potential to solve the aforementioned challenges. An efficient transdifferentiation/direct reprogramming may in the future provide molecular strategies to collectively reduce aging, fibrosis, and degenerative diseases.

2.
Elife ; 122023 07 10.
Article in English | MEDLINE | ID: mdl-37428012

ABSTRACT

Using embryonic stem cells (ESCs) in regenerative medicine or in disease modeling requires a complete understanding of these cells. Two main distinct developmental states of ESCs have been stabilized in vitro, a naïve pre-implantation stage and a primed post-implantation stage. Based on two recently published CRISPR-Cas9 knockout functional screens, we show here that the exit of the naïve state is impaired upon heme biosynthesis pathway blockade, linked in mESCs to the incapacity to activate MAPK- and TGFß-dependent signaling pathways after succinate accumulation. In addition, heme synthesis inhibition promotes the acquisition of 2 cell-like cells in a heme-independent manner caused by a mitochondrial succinate accumulation and leakage out of the cell. We further demonstrate that extracellular succinate acts as a paracrine/autocrine signal, able to trigger the 2C-like reprogramming through the activation of its plasma membrane receptor, SUCNR1. Overall, this study unveils a new mechanism underlying the maintenance of pluripotency under the control of heme synthesis.


Subject(s)
Embryonic Stem Cells , Succinic Acid , Cell Differentiation , Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells , Succinic Acid/metabolism , Animals , Mice
3.
Stem Cell Reports ; 18(1): 254-268, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36563686

ABSTRACT

Translational regulation is of paramount importance for proteome remodeling during stem cell differentiation at both the global and the transcript-specific levels. In this study, we characterized translational remodeling during hepatogenic differentiation of induced pluripotent stem cells (iPSCs) by polysome profiling. We demonstrate that protein synthesis increases during exit from pluripotency and is then globally repressed during later steps of hepatogenic maturation. This global downregulation of translation is accompanied by a decrease in the abundance of protein components of the translation machinery, which involves a global reduction in translational efficiency of terminal oligopyrimidine tract (TOP) mRNA encoding translation-related factors. Despite global translational repression during hepatogenic differentiation, key hepatogenic genes remain efficiently translated, and the translation of several transcripts involved in hepatospecific functions and metabolic maturation is even induced. We conclude that, during hepatogenic differentiation, a global decrease in protein synthesis is accompanied by a specific translational rewiring of hepatospecific transcripts.


Subject(s)
Carrier Proteins , Protein Biosynthesis , Down-Regulation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Differentiation/genetics , Carrier Proteins/genetics
4.
Metabolites ; 12(7)2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35888775

ABSTRACT

Pluripotent cells have been stabilized from pre- and post-implantation blastocysts, representing respectively naïve and primed stages of embryonic stem cells (ESCs) with distinct epigenetic, metabolic and transcriptomic features. Beside these two well characterized pluripotent stages, several intermediate states have been reported, as well as a small subpopulation of cells that have reacquired features of the 2C-embryo (2C-like cells) in naïve mouse ESC culture. Altogether, these represent a continuum of distinct pluripotency stages, characterized by metabolic transitions, for which we propose a new role for a long-known metabolite: succinate. Mostly seen as the metabolite of the TCA, succinate is also at the crossroad of several mitochondrial biochemical pathways. Its role also extends far beyond the mitochondrion, as it can be secreted, modify proteins by lysine succinylation and inhibit the activity of alpha-ketoglutarate-dependent dioxygenases, such as prolyl hydroxylase (PHDs) or histone and DNA demethylases. When released in the extracellular compartment, succinate can trigger several key transduction pathways after binding to SUCNR1, a G-Protein Coupled Receptor. In this review, we highlight the different intra- and extracellular roles that succinate might play in the fields of early pluripotency and embryo development.

6.
Int J Mol Sci ; 20(23)2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31766734

ABSTRACT

microRNAs are ~22bp nucleotide non-coding RNAs that play important roles in the post-transcriptional regulation of gene expression. Many studies have established that microRNAs are important for cell fate choices, including the naïve to primed pluripotency state transitions, and their intermediate state, the developmentally suspended diapause state in early development. However, the full extent of microRNAs associated with these stage transitions in human and mouse remain under-explored. By meta-analysis of microRNA-seq, RNA-seq, and metabolomics datasets from human and mouse, we found a set of microRNAs, and importantly, their experimentally validated target genes that show consistent changes in naïve to primed transitions (microRNA up, target genes down, or vice versa). The targets of these microRNAs regulate developmental pathways (e.g., the Hedgehog-pathway), primary cilium, and remodeling of metabolic processes (oxidative phosphorylation, fatty acid metabolism, and amino acid transport) during the transition. Importantly, we identified 115 microRNAs that significantly change in the same direction in naïve to primed transitions in both human and mouse, many of which are novel candidate regulators of pluripotency. Furthermore, we identified 38 microRNAs and 274 target genes that may be involved in diapause, where embryonic development is temporarily suspended prior to implantation to uterus. The upregulated target genes suggest that microRNAs activate stress response in the diapause stage. In conclusion, we provide a comprehensive resource of microRNAs and their target genes involved in naïve to primed transition and in the paused intermediate, the embryonic diapause stage.


Subject(s)
Databases, Genetic , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , MicroRNAs , Mouse Embryonic Stem Cells/metabolism , Animals , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , Mouse Embryonic Stem Cells/cytology
7.
Nat Commun ; 10(1): 4671, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604922

ABSTRACT

Mitochondrial trifunctional protein deficiency, due to mutations in hydratase subunit A (HADHA), results in sudden infant death syndrome with no cure. To reveal the disease etiology, we generated stem cell-derived cardiomyocytes from HADHA-deficient hiPSCs and accelerated their maturation via an engineered microRNA maturation cocktail that upregulated the epigenetic regulator, HOPX.  Here we report, matured HADHA mutant cardiomyocytes treated with an endogenous mixture of fatty acids manifest the disease phenotype: defective calcium dynamics and repolarization kinetics which results in a pro-arrhythmic state. Single cell RNA-seq reveals a cardiomyocyte developmental intermediate, based on metabolic gene expression. This intermediate gives rise to mature-like cardiomyocytes in control cells but, mutant cells transition to a pathological state with reduced fatty acid beta-oxidation, reduced mitochondrial proton gradient, disrupted cristae structure and defective cardiolipin remodeling. This study reveals that HADHA (tri-functional protein alpha), a monolysocardiolipin acyltransferase-like enzyme, is required for fatty acid beta-oxidation and cardiolipin remodeling, essential for functional mitochondria in human cardiomyocytes.


Subject(s)
Cardiolipins/metabolism , Fatty Acids/metabolism , Mitochondrial Trifunctional Protein, alpha Subunit/physiology , Myocytes, Cardiac/metabolism , Calcium/metabolism , Cell Line , Electrophysiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Human Embryonic Stem Cells , Humans , MicroRNAs/physiology , Mitochondria/physiology , Mitochondrial Trifunctional Protein/deficiency , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Oxidation-Reduction , Patch-Clamp Techniques , RNA-Seq , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology
8.
Sci Rep ; 9(1): 2195, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778087

ABSTRACT

Tissue resident adult stem cells are known to participate in tissue regeneration and repair that follows cell turnover, or injury. It has been well established that aging impedes the regeneration capabilities at the cellular level, but it is not clear if the different onset of stem cell aging between individuals can be predicted or prevented at an earlier stage. Here we studied the dental pulp stem cells (DPSCs), a population of adult stem cells that is known to participate in the repair of an injured tooth, and its properties can be affected by aging. The dental pulp from third molars of a diverse patient group were surgically extracted, generating cells that had a high percentage of mesenchymal stem cell markers CD29, CD44, CD146 and Stro1 and had the ability to differentiate into osteo/odontogenic and adipogenic lineages. Through RNA seq and qPCR analysis we identified homeobox protein, Barx1, as a marker for DPSCs. Furthermore, using high throughput transcriptomic and proteomic analysis we identified markers for DPSC populations with accelerated replicative senescence. In particular, we show that the transforming growth factor-beta (TGF-ß) pathway and the cytoskeletal proteins are upregulated in rapid aging DPSCs, indicating a loss of stem cell characteristics and spontaneous initiation of terminal differentiation. Importantly, using metabolic flux analysis, we identified a metabolic signature for the rapid aging DPSCs, prior to manifestation of senescence phenotypes. This metabolic signature therefore can be used to predict the onset of replicative senescence. Hence, the present study identifies Barx1 as a DPSCs marker and dissects the first predictive metabolic signature for DPSCs aging.


Subject(s)
Cellular Senescence , Dental Pulp/cytology , Energy Metabolism , Stem Cells/cytology , Stem Cells/metabolism , Adipogenesis , Biomarkers , Cell Differentiation , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Immunophenotyping , Odontogenesis , Osteogenesis , Proteomics , Signal Transduction , Transcriptome
9.
Cell Cycle ; 17(5): 535-549, 2018.
Article in English | MEDLINE | ID: mdl-29466914

ABSTRACT

To easily edit the genome of naïve human embryonic stem cells (hESC), we introduced a dual cassette encoding an inducible Cas9 into the AAVS1 site of naïve hESC (iCas9). The iCas9 line retained karyotypic stability, expression of pluripotency markers, differentiation potential, and stability in 5iLA and EPS pluripotency conditions. The iCas9 line induced efficient homology-directed repair (HDR) and non-homologous end joining (NHEJ) based mutations through CRISPR-Cas9 system. We utilized the iCas9 line to study the epigenetic regulator, PRC2 in early human pluripotency. The PRC2 requirement distinguishes between early pluripotency stages, however, what regulates PRC2 activity in these stages is not understood. We show reduced H3K27me3 and pluripotency markers in JARID2 2iL-I-F hESC mutants, indicating JARID2 requirement in maintenance of hESC 2iL-I-F state. These data suggest that JARID2 regulates PRC2 in 2iL-I-F state and the lack of PRC2 function in 5iLA state may be due to lack of sufficient JARID2 protein.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Polycomb Repressive Complex 2/metabolism , Blastocyst/cytology , Blastocyst/metabolism , Cell Self Renewal , DNA End-Joining Repair , Genetic Loci , Histones/metabolism , Human Embryonic Stem Cells , Humans , INDEL Mutation , Microscopy, Confocal , Phenotype , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/genetics , Presenilin-2/genetics , Presenilin-2/metabolism , Protein Domains
10.
Proc Natl Acad Sci U S A ; 114(38): 10125-10130, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28864533

ABSTRACT

The polycomb repressive complex 2 (PRC2) histone methyltransferase plays a central role in epigenetic regulation in development and in cancer, and hence to interrogate its role in a specific developmental transition, methods are needed for disrupting function of the complex with high temporal and spatial precision. The catalytic and substrate recognition functions of PRC2 are coupled by binding of the N-terminal helix of the Ezh2 methylase to an extended groove on the EED trimethyl lysine binding subunit. Disrupting PRC2 function can in principle be achieved by blocking this single interaction, but there are few approaches for blocking specific protein-protein interactions in living cells and organisms. Here, we describe the computational design of proteins that bind to the EZH2 interaction site on EED with subnanomolar affinity in vitro and form tight and specific complexes with EED in living cells. Induction of the EED binding proteins abolishes H3K27 methylation in human embryonic stem cells (hESCs) and at all but the earliest stage blocks self-renewal, pinpointing the first critical repressive H3K27me3 marks in development.


Subject(s)
Computer Simulation , Histones/metabolism , Human Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Histones/chemistry , Human Embryonic Stem Cells/cytology , Humans , Methylation , Polycomb Repressive Complex 2/chemistry
11.
Nat Cell Biol ; 17(12): 1523-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26571212

ABSTRACT

For nearly a century developmental biologists have recognized that cells from embryos can differ in their potential to differentiate into distinct cell types. Recently, it has been recognized that embryonic stem cells derived from both mice and humans exhibit two stable yet epigenetically distinct states of pluripotency: naive and primed. We now show that nicotinamide N-methyltransferase (NNMT) and the metabolic state regulate pluripotency in human embryonic stem cells (hESCs).  Specifically, in naive hESCs, NNMT and its enzymatic product 1-methylnicotinamide are highly upregulated, and NNMT is required for low S-adenosyl methionine (SAM) levels and the H3K27me3 repressive state. NNMT consumes SAM in naive cells, making it unavailable for histone methylation that represses Wnt and activates the HIF pathway in primed hESCs. These data support the hypothesis that the metabolome regulates the epigenetic landscape of the earliest steps in human development.


Subject(s)
Cell Differentiation , Epigenesis, Genetic/genetics , Human Embryonic Stem Cells/metabolism , Metabolome , Animals , Blotting, Western , Cells, Cultured , Embryonic Stem Cells/metabolism , Gas Chromatography-Mass Spectrometry , Gene Expression Profiling/methods , Gene Knockdown Techniques , Histones/metabolism , Humans , Lysine/metabolism , Mass Spectrometry , Metabolomics/methods , Methylation , Mice , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Nicotinamide N-Methyltransferase/genetics , Nicotinamide N-Methyltransferase/metabolism , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , S-Adenosylmethionine/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...