Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 344
Filtrar
1.
Cell Rep ; 43(6): 114372, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38878289

RESUMO

Emerging evidence highlights the regulatory role of paired-like (PRD-like) homeobox transcription factors (TFs) in embryonic genome activation (EGA). However, the majority of PRD-like genes are lost in rodents, thus prompting an investigation into PRD-like TFs in other mammals. Here, we showed that PRD-like TFs were transiently expressed during EGA in human, monkey, and porcine fertilized embryos, yet they exhibited inadequate expression in their cloned embryos. This study, using pig as the research model, identified LEUTX as a key PRD-like activator of porcine EGA through genomic profiling and found that LEUTX overexpression restored EGA failure and improved preimplantation development and cloning efficiency in porcine cloned embryos. Mechanistically, LEUTX opened EGA-related genomic regions and established histone acetylation via recruiting acetyltransferases p300 and KAT2A. These findings reveal the regulatory mechanism of LEUTX to govern EGA in pigs, which may provide valuable insights into the study of early embryo development for other non-rodent mammals.


Assuntos
Genoma , Técnicas de Transferência Nuclear , Animais , Suínos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Desenvolvimento Embrionário/genética , Embrião de Mamíferos/metabolismo , Humanos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Acetilação , Clonagem de Organismos/métodos , Histonas/metabolismo , Blastocisto/metabolismo
2.
Cell Rep ; 43(6): 114309, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38848215

RESUMO

Glioblastomas are the most common malignant brain tumors in adults; they are highly aggressive and heterogeneous and show a high degree of plasticity. Here, we show that methyltransferase-like 7B (METTL7B) is an essential regulator of lineage specification in glioblastoma, with an impact on both tumor size and invasiveness. Single-cell transcriptomic analysis of these tumors and of cerebral organoids derived from expanded potential stem cells overexpressing METTL7B reveal a regulatory role for the gene in the neural stem cell-to-astrocyte differentiation trajectory. Mechanistically, METTL7B downregulates the expression of key neuronal differentiation players, including SALL2, via post-translational modifications of histone marks.


Assuntos
Diferenciação Celular , Linhagem da Célula , Glioblastoma , Metiltransferases , Glioblastoma/patologia , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Metiltransferases/metabolismo , Metiltransferases/genética , Linhagem da Célula/genética , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Linhagem Celular Tumoral , Astrócitos/metabolismo , Astrócitos/patologia , Organoides/metabolismo , Organoides/patologia
4.
J Med Ethics ; 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38834240

RESUMO

There are increasing numbers of clinical trials assessing high-risk, irreversible treatments. Trial participants should only expect knowledge gain to society, no personal therapeutic benefit. However, participation may lead to long-term harms and prevent future therapeutic options. While some discussion has occurred around post-trial access to treatments for participants who received therapeutic benefit, there are no post-trial support requirements for those suffering long-term consequences from trial participation. Participants may be left with significant medical, psychological, social, technical or financial needs. All trials will end at some point, regardless of their success. Subsequently, they should be designed to take into account the post-trial period including the impact on the ongoing health of a participant and their post-trial needs.

5.
Cell Rep ; 43(7): 114247, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38907996

RESUMO

Human induced pluripotent stem cell (hiPSC)-derived intestinal organoids are valuable tools for researching developmental biology and personalized therapies, but their closed topology and relative immature state limit applications. Here, we use organ-on-chip technology to develop a hiPSC-derived intestinal barrier with apical and basolateral access in a more physiological in vitro microenvironment. To replicate growth factor gradients along the crypt-villus axis, we locally expose the cells to expansion and differentiation media. In these conditions, intestinal epithelial cells self-organize into villus-like folds with physiological barrier integrity, and myofibroblasts and neurons emerge and form a subepithelial tissue in the bottom channel. The growth factor gradients efficiently balance dividing and mature cell types and induce an intestinal epithelial composition, including absorptive and secretory lineages, resembling the composition of the human small intestine. This well-characterized hiPSC-derived intestine-on-chip system can facilitate personalized studies on physiological processes and therapy development in the human small intestine.

6.
Cell Rep ; 43(6): 114340, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38865239

RESUMO

Whole salivary gland generation and transplantation offer potential therapies for salivary gland dysfunction. However, the specific lineage required to engineer complete salivary glands has remained elusive. In this study, we identify the Foxa2 lineage as a critical lineage for salivary gland development through conditional blastocyst complementation (CBC). Foxa2 lineage marking begins at the boundary between the endodermal and ectodermal regions of the oral epithelium before the formation of the primordial salivary gland, thereby labeling the entire gland. Ablation of Fgfr2 within the Foxa2 lineage in mice leads to salivary gland agenesis. We reversed this phenotype by injecting donor pluripotent stem cells into the mouse blastocysts, resulting in mice that survived to adulthood with salivary glands of normal size, comparable to those of their littermate controls. These findings demonstrate that CBC-based salivary gland regeneration serves as a foundational experimental approach for future advanced cell-based therapies.


Assuntos
Blastocisto , Fator 3-beta Nuclear de Hepatócito , Células-Tronco Pluripotentes , Glândulas Salivares , Animais , Glândulas Salivares/citologia , Glândulas Salivares/metabolismo , Blastocisto/metabolismo , Blastocisto/citologia , Camundongos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Linhagem da Célula , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética
7.
Cell Rep ; 43(7): 114388, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38935497

RESUMO

In contrast to most hematopoietic lineages, megakaryocytes (MKs) can derive rapidly and directly from hematopoietic stem cells (HSCs). The underlying mechanism is unclear, however. Here, we show that DNA damage induces MK markers in HSCs and that G2 arrest, an integral part of the DNA damage response, suffices for MK priming followed by irreversible MK differentiation in HSCs, but not in progenitors. We also show that replication stress causes DNA damage in HSCs and is at least in part due to uracil misincorporation in vitro and in vivo. Consistent with this notion, thymidine attenuated DNA damage, improved HSC maintenance, and reduced the generation of CD41+ MK-committed HSCs. Replication stress and concomitant MK differentiation is therefore one of the barriers to HSC maintenance. DNA damage-induced MK priming may allow rapid generation of a lineage essential to immediate organismal survival, while also removing damaged cells from the HSC pool.

8.
Cytotherapy ; 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38775774

RESUMO

In recent years, Malaysia has seen a surge in stem cell therapy for various medical conditions. However, the regulation of stem cell research and therapy in Malaysia faces several challenges such as the emergence of unregulated clinics and a lack of specific legislation. Some urgent measures, including enactment of specific laws, strengthened monitoring, as well as increased public awareness and education, are crucial. Therefore, stem cell therapy regulation requires concerted efforts by the policymakers, regulator bodies and healthcare professionals. This commentary discusses the current guidelines and challenges in Malaysian stem cell therapy regulation and proposes some future recommendations that could pave the way for responsible progress of stem cell research and therapy globally.

9.
Cell Rep ; 43(5): 114219, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38748874

RESUMO

Defining the molecular networks orchestrating human brain formation is crucial for understanding neurodevelopment and neurological disorders. Challenges in acquiring early brain tissue have incentivized the use of three-dimensional human pluripotent stem cell (hPSC)-derived neural organoids to recapitulate neurodevelopment. To elucidate the molecular programs that drive this highly dynamic process, here, we generate a comprehensive trans-omic map of the phosphoproteome, proteome, and transcriptome of the exit of pluripotency and neural differentiation toward human cerebral organoids (hCOs). These data reveal key phospho-signaling events and their convergence on transcriptional factors to regulate hCO formation. Comparative analysis with developing human and mouse embryos demonstrates the fidelity of our hCOs in modeling embryonic brain development. Finally, we demonstrate that biochemical modulation of AKT signaling can control hCO differentiation. Together, our data provide a comprehensive resource to study molecular controls in human embryonic brain development and provide a guide for the future development of hCO differentiation protocols.


Assuntos
Encéfalo , Diferenciação Celular , Organoides , Humanos , Organoides/metabolismo , Encéfalo/metabolismo , Encéfalo/embriologia , Animais , Camundongos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Proteoma/metabolismo , Transdução de Sinais , Transcriptoma/genética , Proteômica/métodos , Neurogênese , Proteínas Proto-Oncogênicas c-akt/metabolismo
10.
Cell Rep ; 43(5): 114232, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38761378

RESUMO

The advent of novel 2D and 3D models for human development, including trophoblast stem cells and blastoids, has expanded opportunities for investigating early developmental events, gradually illuminating the enigmatic realm of human development. While these innovations have ushered in new prospects, it has become essential to establish well-defined benchmarks for the cell sources of these models. We aimed to propose a comprehensive characterization of pluripotent and trophoblastic stem cell models by employing a combination of transcriptomic, proteomic, epigenetic, and metabolic approaches. Our findings reveal that extended pluripotent stem cells share many characteristics with primed pluripotent stem cells, with the exception of metabolic activity. Furthermore, our research demonstrates that DNA hypomethylation and high metabolic activity define trophoblast stem cells. These results underscore the necessity of considering multiple hallmarks of pluripotency rather than relying on a single criterion. Multiplying hallmarks alleviate stage-matching bias.


Assuntos
Trofoblastos , Humanos , Trofoblastos/metabolismo , Trofoblastos/citologia , Metilação de DNA , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Modelos Biológicos , Implantação do Embrião , Diferenciação Celular , Epigênese Genética , Transcriptoma/genética , Proteômica/métodos
11.
Cell Rep ; 43(5): 114177, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38691453

RESUMO

Muscle stem cells (MuSCs) contribute to a robust muscle regeneration process after injury, which is highly orchestrated by the sequential expression of multiple key transcription factors. However, it remains unclear how key transcription factors and cofactors such as the Mediator complex cooperate to regulate myogenesis. Here, we show that the Mediator Med23 is critically important for MuSC-mediated muscle regeneration. Med23 is increasingly expressed in activated/proliferating MuSCs on isolated myofibers or in response to muscle injury. Med23 deficiency reduced MuSC proliferation and enhanced its precocious differentiation, ultimately compromising muscle regeneration. Integrative analysis revealed that Med23 oppositely impacts Ternary complex factor (TCF)-targeted MuSC proliferation genes and myocardin-related transcription factor (MRTF)-targeted myogenic differentiation genes. Consistently, Med23 deficiency decreases the ETS-like transcription factor 1 (Elk1)/serum response factor (SRF) binding at proliferation gene promoters but promotes MRTF-A/SRF binding at myogenic gene promoters. Overall, our study reveals the important transcriptional control mechanism of Med23 in balancing MuSC proliferation and differentiation in muscle regeneration.


Assuntos
Diferenciação Celular , Proliferação de Células , Complexo Mediador , Desenvolvimento Muscular , Regeneração , Células-Tronco , Animais , Camundongos , Desenvolvimento Muscular/genética , Células-Tronco/metabolismo , Células-Tronco/citologia , Complexo Mediador/metabolismo , Complexo Mediador/genética , Músculo Esquelético/metabolismo , Transcrição Gênica , Camundongos Endogâmicos C57BL , Transativadores/metabolismo , Transativadores/genética
12.
Cell Rep ; 43(5): 114227, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38735044

RESUMO

CUX1 is a homeodomain-containing transcription factor that is essential for the development and differentiation of multiple tissues. CUX1 is recurrently mutated or deleted in cancer, particularly in myeloid malignancies. However, the mechanism by which CUX1 regulates gene expression and differentiation remains poorly understood, creating a barrier to understanding the tumor-suppressive functions of CUX1. Here, we demonstrate that CUX1 directs the BAF chromatin remodeling complex to DNA to increase chromatin accessibility in hematopoietic cells. CUX1 preferentially regulates lineage-specific enhancers, and CUX1 target genes are predictive of cell fate in vivo. These data indicate that CUX1 regulates hematopoietic lineage commitment and homeostasis via pioneer factor activity, and CUX1 deficiency disrupts these processes in stem and progenitor cells, facilitating transformation.


Assuntos
Cromatina , Células-Tronco Hematopoéticas , Proteínas de Homeodomínio , Proteínas Repressoras , Humanos , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Cromatina/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Animais , Camundongos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos/genética
13.
Cell Rep ; 43(5): 114170, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38700983

RESUMO

During cell fate transitions, cells remodel their transcriptome, chromatin, and epigenome; however, it has been difficult to determine the temporal dynamics and cause-effect relationship between these changes at the single-cell level. Here, we employ the heterokaryon-mediated reprogramming system as a single-cell model to dissect key temporal events during early stages of pluripotency conversion using super-resolution imaging. We reveal that, following heterokaryon formation, the somatic nucleus undergoes global chromatin decompaction and removal of repressive histone modifications H3K9me3 and H3K27me3 without acquisition of active modifications H3K4me3 and H3K9ac. The pluripotency gene OCT4 (POU5F1) shows nascent and mature RNA transcription within the first 24 h after cell fusion without requiring an initial open chromatin configuration at its locus. NANOG, conversely, has significant nascent RNA transcription only at 48 h after cell fusion but, strikingly, exhibits genomic reopening early on. These findings suggest that the temporal relationship between chromatin compaction and gene activation during cellular reprogramming is gene context dependent.


Assuntos
Reprogramação Celular , Montagem e Desmontagem da Cromatina , Histonas , Humanos , Reprogramação Celular/genética , Histonas/metabolismo , Análise de Célula Única , Ativação Transcricional , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Cromatina/metabolismo , Proteína Homeobox Nanog/metabolismo , Proteína Homeobox Nanog/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia
14.
Cell Rep ; 43(6): 114270, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38787726

RESUMO

Stem cells play pivotal roles in maintaining intestinal homeostasis, orchestrating regeneration, and in key steps of colorectal cancer (CRC) initiation and progression. Intriguingly, adult stem cells are reduced during many of these processes. On the contrary, primitive fetal programs, commonly detected in development, emerge during tissue repair, CRC metastasis, and therapy resistance. Recent findings indicate a dynamic continuum between adult and fetal stem cell programs. We discuss critical mechanisms facilitating the plasticity between stem cell states and highlight the heterogeneity observed upon the appearance of fetal-like states. We focus on therapeutic opportunities that arise by targeting fetal-like CRC cells and how those concepts can be translated into the clinic.


Assuntos
Neoplasias Colorretais , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/terapia , Humanos , Animais , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Fetais/metabolismo
15.
Cell Rep ; 43(4): 114092, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38607913

RESUMO

Macrophages conduct critical roles in heart repair, but the niche required to nurture and anchor them is poorly studied. Here, we investigated the macrophage niche in the regenerating heart. We analyzed cell-cell interactions through published single-cell RNA sequencing datasets and identified a strong interaction between fibroblast/epicardial (Fb/Epi) cells and macrophages. We further visualized the association of macrophages with Fb/Epi cells and the blockage of macrophage response without Fb/Epi cells in the regenerating zebrafish heart. Moreover, we found that ptx3a+ epicardial cells associate with reparative macrophages, and their depletion resulted in fewer reparative macrophages. Further, we identified csf1a expression in ptx3a+ cells and determined that pharmacological inhibition of the csf1a pathway or csf1a knockout blocked the reparative macrophage response. Moreover, we found that genetic overexpression of csf1a enhanced the reparative macrophage response with or without heart injury. Altogether, our studies illuminate a cardiac Fb/Epi niche, which mediates a beneficial macrophage response after heart injury.


Assuntos
Fibroblastos , Coração , Macrófagos , Regeneração , Peixe-Zebra , Animais , Proteína C-Reativa/metabolismo , Proteína C-Reativa/genética , Fibroblastos/metabolismo , Coração/fisiologia , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Macrófagos/metabolismo , Pericárdio/metabolismo , Pericárdio/citologia , Regeneração/fisiologia , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
16.
Cell Rep ; 43(5): 114136, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38643480

RESUMO

Embryos, originating from fertilized eggs, undergo continuous cell division and differentiation, accompanied by dramatic changes in transcription, translation, and metabolism. Chromatin regulators, including transcription factors (TFs), play indispensable roles in regulating these processes. Recently, the trophoblast regulator TFAP2C was identified as crucial in initiating early cell fate decisions. However, Tfap2c transcripts persist in both the inner cell mass and trophectoderm of blastocysts, prompting inquiry into Tfap2c's function in post-lineage establishment. In this study, we delineate the dynamics of TFAP2C during the mouse peri-implantation stage and elucidate its collaboration with the key lineage regulators CDX2 and NANOG. Importantly, we propose that de novo formation of H3K9me3 in the extraembryonic ectoderm during implantation antagonizes TFAP2C binding to crucial developmental genes, thereby maintaining its lineage identity. Together, these results highlight the plasticity of the chromatin environment in designating the genomic binding of highly adaptable lineage-specific TFs and regulating embryonic cell fates.


Assuntos
Fator de Transcrição CDX2 , Linhagem da Célula , Cromatina , Regulação da Expressão Gênica no Desenvolvimento , Fator de Transcrição AP-2 , Animais , Cromatina/metabolismo , Camundongos , Linhagem da Célula/genética , Fator de Transcrição AP-2/metabolismo , Fator de Transcrição AP-2/genética , Fator de Transcrição CDX2/metabolismo , Fator de Transcrição CDX2/genética , Proteína Homeobox Nanog/metabolismo , Proteína Homeobox Nanog/genética , Blastocisto/metabolismo , Blastocisto/citologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Feminino , Histonas/metabolismo , Diferenciação Celular/genética , Ectoderma/metabolismo , Ectoderma/citologia , Desenvolvimento Embrionário/genética
17.
Front Public Health ; 12: 1364809, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38628851

RESUMO

Introduction: Although stem cell research and therapeutic applications hold great promise for medical advancements, and have rapidly progressed globally, there remains a lack of genuine public awareness of the status of this subject in Saudi Arabia. Successful integration of stem cell therapy into healthcare relies on public awareness, understanding, and trust. Therefore, we aimed in this cross-sectional study to assess the public's knowledge, awareness, trust, support, participation, and confidence in stem cell treatments and centers involved in it. Materials and methods: A voluntary questionnaire of 20 questions was distributed randomly via social media outlets. Results: Three thousand five hundred eighty four individuals participated in the survey, with approximately half of them falling within the age range of 35-50 years (46.71%). Majority of the participants, 90.71%, would like to know more about stem cell therapy and more than half of the participants (56.94%) were unfamiliar with the idea, and a comparable proportion (50.41%) expressed concerns about the safety of stem cell therapy. A lower level of awareness, indicated by a score of 5, was evenly distributed across all age groups and genders. However, regardless of gender, older participants-especially those 50 years of age or older-tended to report higher levels of confidence, trust, and support than participants in other age groups. Moreover, trust, support, participation, and confidence score for those attained high school or less was statistically significantly lower than those attained master's or PhD degree. Of the participants, 33.57% had either received stem cell therapy themselves or known someone who had; about 24.07% of them reported that it was a cosmetic type of treatment. Conclusion: The study emphasizes the persistent need for awareness and educational initiatives to minimize the lack of public awareness and understanding of approved stem cell treatments in Saudi Arabia. It advocates for increased education, transparency, and communication to bridge knowledge gaps and enhance public trust to ensure the understanding of successful treatment.


Assuntos
Pesquisa com Células-Tronco , Confiança , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Estudos Transversais , Arábia Saudita , Conhecimentos, Atitudes e Prática em Saúde , Hospitais
18.
Cell Rep ; 43(4): 114052, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38573860

RESUMO

Skeletal muscles exert remarkable regenerative or adaptive capacities in response to injuries or mechanical loads. However, the cellular networks underlying muscle adaptation are poorly understood compared to those underlying muscle regeneration. We employed single-cell RNA sequencing to investigate the gene expression patterns and cellular networks activated in overloaded muscles and compared these results with those observed in regenerating muscles. The cellular composition of the 4-day overloaded muscle, when macrophage infiltration peaked, closely resembled that of the 10-day regenerating muscle. In addition to the mesenchymal progenitor-muscle satellite cell (MuSC) axis, interactome analyses or targeted depletion experiments revealed communications between mesenchymal progenitors-macrophages and macrophages-MuSCs. Furthermore, granulin, a macrophage-derived factor, inhibited MuSC differentiation, and Granulin-knockout mice exhibited blunted muscle hypertrophy due to the premature differentiation of overloaded MuSCs. These findings reveal the critical role of granulin through the relayed communications of mesenchymal progenitors, macrophages, and MuSCs in facilitating efficient muscle hypertrophy.


Assuntos
Diferenciação Celular , Hipertrofia , Macrófagos , Células-Tronco Mesenquimais , Camundongos Knockout , Células Satélites de Músculo Esquelético , Animais , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Granulinas , Comunicação Celular , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Masculino , Regeneração
19.
Cell Rep ; 43(4): 114024, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38581679

RESUMO

Mouse embryonic stem cells (mESCs) in the primed pluripotency state, which resembles the post-implantation epiblast, can be de-differentiated in culture to a naive state that resembles the pre-implantation inner cell mass. We report that primed-to-naive mESC transition entails a significant slowdown of DNA replication forks and the compensatory activation of dormant origins. Using isolation of proteins on nascent DNA coupled to mass spectrometry, we identify key changes in replisome composition that are responsible for these effects. Naive mESC forks are enriched in MRE11 nuclease and other DNA repair proteins. MRE11 is recruited to newly synthesized DNA in response to transcription-replication conflicts, and its inhibition or genetic downregulation in naive mESCs is sufficient to restore the fork rate of primed cells. Transcriptomic analyses indicate that MRE11 exonuclease activity is required for the complete primed-to-naive mESC transition, demonstrating a direct link between DNA replication dynamics and the mESC de-differentiation process.


Assuntos
Replicação do DNA , Proteína Homóloga a MRE11 , Animais , Camundongos , Proteína Homóloga a MRE11/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Desdiferenciação Celular , Proteínas de Ligação a DNA/metabolismo
20.
Cell Rep ; 43(4): 114113, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38625792

RESUMO

The continuous regeneration of spermatogonial stem cells (SSCs) underpins spermatogenesis and lifelong male fertility, but the developmental origins of the SSC pool remain unclear. Here, we document that hnRNPU is essential for establishing the SSC pool. In male mice, conditional loss of hnRNPU in prospermatogonia (ProSG) arrests spermatogenesis and results in sterility. hnRNPU-deficient ProSG fails to differentiate and migrate to the basement membrane to establish SSC pool in infancy. Moreover, hnRNPU deletion leads to the accumulation of ProSG and disrupts the process of T1-ProSG to T2-ProSG transition. Single-cell transcriptional analyses reveal that germ cells are in a mitotically quiescent state and lose their unique identity upon hnRNPU depletion. We further show that hnRNPU could bind to Vrk1, Slx4, and Dazl transcripts that have been identified to suffer aberrant alternative splicing in hnRNPU-deficient testes. These observations offer important insights into SSC pool establishment and may have translational implications for male fertility.


Assuntos
Espermatogênese , Espermatogônias , Animais , Masculino , Camundongos , Células-Tronco Germinativas Adultas/metabolismo , Processamento Alternativo/genética , Diferenciação Celular , Espermatogênese/genética , Espermatogônias/metabolismo , Espermatogônias/citologia , Células-Tronco/metabolismo , Células-Tronco/citologia , Testículo/metabolismo , Testículo/citologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...