Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cell Rep Med ; 5(3): 101449, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508141

ABSTRACT

Tissue regeneration following an injury requires dynamic cell-state transitions that allow for establishing the cell identities required for the restoration of tissue homeostasis and function. Here, we present a biochemical intervention that induces an intermediate cell state mirroring a transition identified during normal differentiation of myoblasts and other multipotent and pluripotent cells to mature cells. When applied in somatic differentiated cells, the intervention, composed of one-carbon metabolites, reduces some dedifferentiation markers without losing the lineage identity, thus inducing limited reprogramming into a more flexible cell state. Moreover, the intervention enabled accelerated repair after muscle injury in young and aged mice. Overall, our study uncovers a conserved biochemical transitional phase that enhances cellular plasticity in vivo and hints at potential and scalable biochemical interventions of use in regenerative medicine and rejuvenation interventions that may be more tractable than genetic ones.


Subject(s)
Muscles , Myoblasts , Mice , Animals , Cell Differentiation , Myoblasts/metabolism
2.
Cell Rep ; 42(6): 112590, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37261952

ABSTRACT

Distinct metabolic conditions rewire circadian-clock-controlled signaling pathways leading to the de novo construction of signal transduction networks. However, it remains unclear whether metabolic hallmarks unique to pluripotent stem cells (PSCs) are connected to clock functions. Reprogramming somatic cells to a pluripotent state, here we highlighted non-canonical functions of the circadian repressor CRY1 specific to PSCs. Metabolic reprogramming, including AMPK inactivation and SREBP1 activation, was coupled with the accumulation of CRY1 in PSCs. Functional assays verified that CRY1 is required for the maintenance of self-renewal capacity, colony organization, and metabolic signatures. Genome-wide occupancy of CRY1 identified CRY1-regulatory genes enriched in development and differentiation in PSCs, albeit not somatic cells. Last, cells lacking CRY1 exhibit differential gene expression profiles during induced PSC (iPSC) reprogramming, resulting in impaired iPSC reprogramming efficiency. Collectively, these results suggest the functional implication of CRY1 in pluripotent reprogramming and ontogenesis, thereby dictating PSC identity.


Subject(s)
Circadian Clocks , Cryptochromes , Pluripotent Stem Cells , Cell Differentiation , Cellular Reprogramming , Circadian Clocks/genetics , Signal Transduction , Animals , Mice , Cryptochromes/metabolism
3.
Cell ; 186(4): 715-731.e19, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36754048

ABSTRACT

Transgenerational epigenetic inheritance in mammals remains a debated subject. Here, we demonstrate that DNA methylation of promoter-associated CpG islands (CGIs) can be transmitted from parents to their offspring in mice. We generated DNA methylation-edited mouse embryonic stem cells (ESCs), in which CGIs of two metabolism-related genes, the Ankyrin repeat domain 26 and the low-density lipoprotein receptor, were specifically methylated and silenced. DNA methylation-edited mice generated by microinjection of the methylated ESCs exhibited abnormal metabolic phenotypes. Acquired methylation of the targeted CGI and the phenotypic traits were maintained and transmitted across multiple generations. The heritable CGI methylation was subjected to reprogramming in parental PGCs and subsequently reestablished in the next generation at post-implantation stages. These observations provide a concrete step toward demonstrating transgenerational epigenetic inheritance in mammals, which may have implications in our understanding of evolutionary biology as well as the etiology, diagnosis, and prevention of non-genetically inherited human diseases.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Mice , Humans , Animals , CpG Islands , Inheritance Patterns , Mammals/genetics
5.
Nat Commun ; 13(1): 3646, 2022 06 25.
Article in English | MEDLINE | ID: mdl-35752626

ABSTRACT

The diverse functions of WASP, the deficiency of which causes Wiskott-Aldrich syndrome (WAS), remain poorly defined. We generated three isogenic WAS models using patient induced pluripotent stem cells and genome editing. These models recapitulated WAS phenotypes and revealed that WASP deficiency causes an upregulation of numerous RNA splicing factors and widespread altered splicing. Loss of WASP binding to splicing factor gene promoters frequently leads to aberrant epigenetic activation. WASP interacts with dozens of nuclear speckle constituents and constrains SRSF2 mobility. Using an optogenetic system, we showed that WASP forms phase-separated condensates that encompasses SRSF2, nascent RNA and active Pol II. The role of WASP in gene body condensates is corroborated by ChIPseq and RIPseq. Together our data reveal that WASP is a nexus regulator of RNA splicing that controls the transcription of splicing factors epigenetically and the dynamics of the splicing machinery through liquid-liquid phase separation.


Subject(s)
Wiskott-Aldrich Syndrome Protein , Wiskott-Aldrich Syndrome , Alternative Splicing , Cell Nucleus/metabolism , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism
6.
Cell Rep ; 39(4): 110730, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35476977

ABSTRACT

Mammals have limited regenerative capacity, whereas some vertebrates, like fish and salamanders, are able to regenerate their organs efficiently. The regeneration in these species depends on cell dedifferentiation followed by proliferation. We generate a mouse model that enables the inducible expression of the four Yamanaka factors (Oct-3/4, Sox2, Klf4, and c-Myc, or 4F) specifically in hepatocytes. Transient in vivo 4F expression induces partial reprogramming of adult hepatocytes to a progenitor state and concomitantly increases cell proliferation. This is indicated by reduced expression of differentiated hepatic-lineage markers, an increase in markers of proliferation and chromatin modifiers, global changes in DNA accessibility, and an acquisition of liver stem and progenitor cell markers. Functionally, short-term expression of 4F enhances liver regenerative capacity through topoisomerase2-mediated partial reprogramming. Our results reveal that liver-specific 4F expression in vivo induces cellular plasticity and counteracts liver failure, suggesting that partial reprogramming may represent an avenue for enhancing tissue regeneration.


Subject(s)
Cellular Reprogramming , Liver , Animals , Cell Dedifferentiation , Hepatocytes/metabolism , Liver/metabolism , Liver Regeneration , Mammals , Mice
7.
Front Cell Dev Biol ; 10: 786031, 2022.
Article in English | MEDLINE | ID: mdl-35309931

ABSTRACT

It is widely believed that cellular senescence plays a critical role in both aging and cancer, and that senescence is a fundamental, permanent growth arrest that somatic cells cannot avoid. Here we show that Myc plays an important role in self-renewal of esophageal epithelial cells, contributing to their resistance to cellular senescence. Myc is homogeneously expressed in basal cells of the esophageal epithelium and Myc positively regulates their self-renewal by maintaining their undifferentiated state. Indeed, Myc knockout induced a loss of the undifferentiated state of esophageal epithelial cells resulting in cellular senescence while forced MYC expression promoted oncogenic cell proliferation. A superoxide scavenger counteracted Myc knockout-induced senescence, therefore suggesting that a mitochondrial superoxide takes part in inducing senescence. Taken together, these analyses reveal extremely low levels of cellular senescence and senescence-associated phenotypes in the esophageal epithelium, as well as a critical role for Myc in self-renewal of basal cells in this organ. This provides new avenues for studying and understanding the links between stemness and resistance to cellular senescence.

8.
Nat Aging ; 2(3): 243-253, 2022 03.
Article in English | MEDLINE | ID: mdl-37118377

ABSTRACT

Partial reprogramming by expression of reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) for short periods of time restores a youthful epigenetic signature to aging cells and extends the life span of a premature aging mouse model. However, the effects of longer-term partial reprogramming in physiologically aging wild-type mice are unknown. Here, we performed various long-term partial reprogramming regimens, including different onset timings, during physiological aging. Long-term partial reprogramming lead to rejuvenating effects in different tissues, such as the kidney and skin, and at the organismal level; duration of the treatment determined the extent of the beneficial effects. The rejuvenating effects were associated with a reversion of the epigenetic clock and metabolic and transcriptomic changes, including reduced expression of genes involved in the inflammation, senescence and stress response pathways. Overall, our observations indicate that partial reprogramming protocols can be designed to be safe and effective in preventing age-related physiological changes. We further conclude that longer-term partial reprogramming regimens are more effective in delaying aging phenotypes than short-term reprogramming.


Subject(s)
Aging, Premature , Cellular Reprogramming , Animals , Mice , Cellular Reprogramming/genetics , Aging/genetics , Cellular Senescence , Aging, Premature/genetics , Disease Models, Animal
9.
Nat Commun ; 12(1): 3094, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035273

ABSTRACT

Short-term, systemic expression of the Yamanaka reprogramming factors (Oct-3/4, Sox2, Klf4 and c-Myc [OSKM]) has been shown to rejuvenate aging cells and promote tissue regeneration in vivo. However, the mechanisms by which OSKM promotes tissue regeneration are unknown. In this work, we focus on a specific tissue and demonstrate that local expression of OSKM, specifically in myofibers, induces the activation of muscle stem cells or satellite cells (SCs), which accelerates muscle regeneration in young mice. In contrast, expressing OSKM directly in SCs does not improve muscle regeneration. Mechanistically, expressing OSKM in myofibers regulates the expression of genes important for the SC microenvironment, including upregulation of p21, which in turn downregulates Wnt4. This is critical because Wnt4 is secreted by myofibers to maintain SC quiescence. Thus, short-term induction of the Yamanaka factors in myofibers may promote tissue regeneration by modifying the stem cell niche.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Myofibrils/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , Stem Cell Niche , Animals , Cells, Cultured , Female , Gene Expression , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice, Transgenic , Myofibrils/physiology , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , Satellite Cells, Skeletal Muscle/cytology , Wnt4 Protein/genetics
10.
Cell Res ; 31(2): 187-205, 2021 02.
Article in English | MEDLINE | ID: mdl-32737416

ABSTRACT

Accumulating evidence indicates an association between the circadian clock and the aging process. However, it remains elusive whether the deregulation of circadian clock proteins underlies stem cell aging and whether they are targetable for the alleviation of aging-associated syndromes. Here, we identified a transcription factor-independent role of CLOCK, a core component of the molecular circadian clock machinery, in counteracting human mesenchymal stem cell (hMSC) decay. CLOCK expression was decreased during hMSC aging. In addition, CLOCK deficiency accelerated hMSC senescence, whereas the overexpression of CLOCK, even as a transcriptionally inactive form, rejuvenated physiologically and pathologically aged hMSCs. Mechanistic studies revealed that CLOCK formed complexes with nuclear lamina proteins and KAP1, thus maintaining heterochromatin architecture and stabilizing repetitive genomic sequences. Finally, gene therapy with lentiviral vectors encoding CLOCK promoted cartilage regeneration and attenuated age-related articular degeneration in mice. These findings demonstrate a noncanonical role of CLOCK in stabilizing heterochromatin, promoting tissue regeneration, and mitigating aging-associated chronic diseases.


Subject(s)
CLOCK Proteins/metabolism , Cartilage, Articular/physiology , Cellular Senescence/genetics , Heterochromatin/metabolism , Mesenchymal Stem Cells/metabolism , Regeneration/genetics , Rejuvenation , Aging/metabolism , Animals , CLOCK Proteins/genetics , Circadian Clocks/genetics , Circadian Rhythm/genetics , Genetic Therapy/methods , Genetic Vectors/therapeutic use , HEK293 Cells , Humans , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Transfection
11.
Sci Rep ; 10(1): 3474, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32103065

ABSTRACT

Reliable approaches to identify stem cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly acquired by cancer cells has been challenging. We harnessed the power of reprogramming to examine GRP78, a chaperone protein generally restricted to the endoplasmic reticulum in normal tissues, but which is expressed on the cell surface of human embryonic stem cells and many cancer types. We have discovered that (1) cell surface GRP78 (sGRP78) is expressed on iPSCs and is important in reprogramming, (2) sGRP78 promotes cellular functions in both pluripotent and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a CD24-/CD44+ tumor initiating cell (TIC) population (4) sGRP78+ breast cancer cells are enriched for stemness genes and appear to be a subset of TICs (5) sGRP78+ breast cancer cells show an enhanced ability to seed metastatic organ sites in vivo. These collective findings show that GRP78 has important functions in regulating both pluripotency and oncogenesis, and suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo.


Subject(s)
Cell Differentiation , Cell Self Renewal , Heat-Shock Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Cellular Reprogramming , Endoplasmic Reticulum Chaperone BiP , Female , HEK293 Cells , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Knockout , Neoplastic Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
13.
Cell Res ; 29(10): 804-819, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31444470

ABSTRACT

In vivo genome editing represents a powerful strategy for both understanding basic biology and treating inherited diseases. However, it remains a challenge to develop universal and efficient in vivo genome-editing tools for tissues that comprise diverse cell types in either a dividing or non-dividing state. Here, we describe a versatile in vivo gene knock-in methodology that enables the targeting of a broad range of mutations and cell types through the insertion of a minigene at an intron of the target gene locus using an intracellularly linearized single homology arm donor. As a proof-of-concept, we focused on a mouse model of premature-aging caused by a dominant point mutation, which is difficult to repair using existing in vivo genome-editing tools. Systemic treatment using our new method ameliorated aging-associated phenotypes and extended animal lifespan, thus highlighting the potential of this methodology for a broad range of in vivo genome-editing applications.


Subject(s)
Gene Editing/methods , Animals , CRISPR-Cas Systems/genetics , DNA Repair , Dependovirus/genetics , GATA3 Transcription Factor/genetics , Gene Knock-In Techniques , Genetic Therapy/methods , Genetic Vectors/metabolism , Human Embryonic Stem Cells , Humans , Introns , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/cytology , Neurons/metabolism , RNA, Guide, Kinetoplastida/metabolism , Rats , Tubulin/genetics
14.
Protein Cell ; 10(7): 485-495, 2019 07.
Article in English | MEDLINE | ID: mdl-31041783

ABSTRACT

Identification of the precise molecular pathways involved in oncogene-induced transformation may help us gain a better understanding of tumor initiation and promotion. Here, we demonstrate that SOX2+ foregut epithelial cells are prone to oncogenic transformation upon mutagenic insults, such as KrasG12D and p53 deletion. GFP-based lineage-tracing experiments indicate that SOX2+ cells are the cells-of-origin of esophagus and stomach hyperplasia. Our observations indicate distinct roles for oncogenic KRAS mutation and P53 deletion. p53 homozygous deletion is required for the acquisition of an invasive potential, and KrasG12D expression, but not p53 deletion, suffices for tumor formation. Global gene expression analysis reveals secreting factors upregulated in the hyperplasia induced by oncogenic KRAS and highlights a crucial role for the CXCR2 pathway in driving hyperplasia. Collectively, the array of genetic models presented here demonstrate that stratified epithelial cells are susceptible to oncogenic insults, which may lead to a better understanding of tumor initiation and aid in the design of new cancer therapeutics.


Subject(s)
Esophageal Neoplasms/metabolism , Mutation , Receptors, Interleukin-8B/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cell Proliferation , Esophageal Neoplasms/pathology , Female , Male , Mice , Mice, Mutant Strains , Signal Transduction , Tumor Cells, Cultured
15.
Nature ; 561(7722): 243-247, 2018 09.
Article in English | MEDLINE | ID: mdl-30185909

ABSTRACT

Large cutaneous ulcers are, in severe cases, life threatening1,2. As the global population ages, non-healing ulcers are becoming increasingly common1,2. Treatment currently requires the transplantation of pre-existing epithelial components, such as skin grafts, or therapy using cultured cells2. Here we develop alternative supplies of epidermal coverage for the treatment of these kinds of wounds. We generated expandable epithelial tissues using in vivo reprogramming of wound-resident mesenchymal cells. Transduction of four transcription factors that specify the skin-cell lineage enabled efficient and rapid de novo epithelialization from the surface of cutaneous ulcers in mice. Our findings may provide a new therapeutic avenue for treating skin wounds and could be extended to other disease situations in which tissue homeostasis and repair are impaired.


Subject(s)
Cellular Reprogramming , Epithelial Cells/cytology , Skin Ulcer/pathology , Skin/cytology , Wounds and Injuries/pathology , Animals , Cell Lineage , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Regenerative Medicine , Skin/pathology , Skin Ulcer/therapy , Transcription Factors/genetics , Transcription Factors/metabolism , Wound Healing , Wounds and Injuries/therapy
16.
Science ; 356(6337): 503-508, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28473583

ABSTRACT

CpG islands (CGIs) are primarily promoter-associated genomic regions and are mostly unmethylated within highly methylated mammalian genomes. The mechanisms by which CGIs are protected from de novo methylation remain elusive. Here we show that insertion of CpG-free DNA into targeted CGIs induces de novo methylation of the entire CGI in human pluripotent stem cells (PSCs). The methylation status is stably maintained even after CpG-free DNA removal, extensive passaging, and differentiation. By targeting the DNA mismatch repair gene MLH1 CGI, we could generate a PSC model of a cancer-related epimutation. Furthermore, we successfully corrected aberrant imprinting in induced PSCs derived from an Angelman syndrome patient. Our results provide insights into how CpG-free DNA induces de novo CGI methylation and broaden the application of targeted epigenome editing for a better understanding of human development and disease.


Subject(s)
CpG Islands , DNA Methylation , Epigenesis, Genetic , Pluripotent Stem Cells/metabolism , DNA/metabolism , DNA Mismatch Repair/genetics , DNA Repair/genetics , Humans , MutL Protein Homolog 1/genetics , Mutagenesis, Insertional , Neurons/metabolism , Ubiquitin-Protein Ligases/genetics
17.
Cell ; 168(3): 473-486.e15, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28129541

ABSTRACT

Interspecies blastocyst complementation enables organ-specific enrichment of xenogenic pluripotent stem cell (PSC) derivatives. Here, we establish a versatile blastocyst complementation platform based on CRISPR-Cas9-mediated zygote genome editing and show enrichment of rat PSC-derivatives in several tissues of gene-edited organogenesis-disabled mice. Besides gaining insights into species evolution, embryogenesis, and human disease, interspecies blastocyst complementation might allow human organ generation in animals whose organ size, anatomy, and physiology are closer to humans. To date, however, whether human PSCs (hPSCs) can contribute to chimera formation in non-rodent species remains unknown. We systematically evaluate the chimeric competency of several types of hPSCs using a more diversified clade of mammals, the ungulates. We find that naïve hPSCs robustly engraft in both pig and cattle pre-implantation blastocysts but show limited contribution to post-implantation pig embryos. Instead, an intermediate hPSC type exhibits higher degree of chimerism and is able to generate differentiated progenies in post-implantation pig embryos.


Subject(s)
Chimerism , Gene Editing , Mammals/embryology , Animals , Blastocyst , CRISPR-Cas Systems , Cattle , Embryo, Mammalian/cytology , Female , Humans , Male , Mammals/classification , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pluripotent Stem Cells , Rats , Rats, Sprague-Dawley , Sus scrofa
18.
Cell ; 167(7): 1719-1733.e12, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984723

ABSTRACT

Aging is the major risk factor for many human diseases. In vitro studies have demonstrated that cellular reprogramming to pluripotency reverses cellular age, but alteration of the aging process through reprogramming has not been directly demonstrated in vivo. Here, we report that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in a mouse model of premature aging. Similarly, expression of OSKM in vivo improves recovery from metabolic disease and muscle injury in older wild-type mice. The amelioration of age-associated phenotypes by epigenetic remodeling during cellular reprogramming highlights the role of epigenetic dysregulation as a driver of mammalian aging. Establishing in vivo platforms to modulate age-associated epigenetic marks may provide further insights into the biology of aging.


Subject(s)
Aging/genetics , Cellular Reprogramming , Epigenesis, Genetic , Metabolic Diseases/genetics , Transcription Factors/metabolism , Aging, Premature/genetics , Aging, Premature/metabolism , Animals , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Lamin Type A/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Mice , Models, Animal , Pancreas/metabolism , Sarcopenia/metabolism
19.
Nature ; 540(7631): 144-149, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27851729

ABSTRACT

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Targeting/methods , Genome/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , Cell Division , Disease Models, Animal , Gene Knock-In Techniques , Genetic Therapy/methods , Neurons/cytology , Neurons/metabolism , Rats , Sequence Homology
20.
Nat Commun ; 7: 11056, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27025988

ABSTRACT

Meiosis is a unique process that allows the generation of reproductive cells. It remains largely unknown how meiosis is initiated in germ cells and why non-germline cells do not undergo meiosis. We previously demonstrated that knockdown of Max expression, a gene encoding a partner of MYC family proteins, strongly activates expression of germ cell-related genes in ESCs. Here we find that complete ablation of Max expression in ESCs results in profound cytological changes reminiscent of cells undergoing meiotic cell division. Furthermore, our analyses uncovers that Max expression is transiently attenuated in germ cells undergoing meiosis in vivo and its forced reduction induces meiosis-like cytological changes in cultured germline stem cells. Mechanistically, Max depletion alterations are, in part, due to impairment of the function of an atypical PRC1 complex (PRC1.6), in which MAX is one of the components. Our data highlight MAX as a new regulator of meiotic onset.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Germ Cells/metabolism , Meiosis/genetics , Mouse Embryonic Stem Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Ascorbic Acid/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Gametogenesis/drug effects , Gametogenesis/genetics , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Germ Cells/drug effects , Meiosis/drug effects , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Polycomb-Group Proteins/metabolism , Retinoids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...