Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(8): 114629, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39146183

RESUMEN

In mice, the first liver-resident macrophages, known as Kupffer cells (KCs), are thought to derive from yolk sac (YS) hematopoietic progenitors that are specified prior to the emergence of the hematopoietic stem cell (HSC). To investigate human KC development, we recapitulated YS-like hematopoiesis from human pluripotent stem cells (hPSCs) and transplanted derivative macrophage progenitors into NSG mice previously humanized with hPSC-liver sinusoidal endothelial cells (LSECs). We demonstrate that hPSC-LSECs facilitate stable hPSC-YS-macrophage engraftment for at least 7 weeks. Single-cell RNA sequencing (scRNA-seq) of engrafted YS-macrophages revealed a homogeneous MARCO-expressing KC gene signature and low expression of monocyte-like macrophage genes. In contrast, human cord blood (CB)-derived macrophage progenitors generated grafts that contain multiple hematopoietic lineages in addition to KCs. Functional analyses showed that the engrafted KCs actively perform phagocytosis and erythrophagocytosis in vivo. Taken together, these findings demonstrate that it is possible to generate human KCs from hPSC-derived, YS-like progenitors.


Asunto(s)
Diferenciación Celular , Células Endoteliales , Macrófagos del Hígado , Hígado , Células Madre Pluripotentes , Humanos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Células Endoteliales/metabolismo , Células Endoteliales/citología , Animales , Hígado/citología , Hígado/metabolismo , Ratones , Fagocitosis , Hematopoyesis
2.
Cell Rep ; 43(8): 114647, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39153199

RESUMEN

Reproduction is paramount to animals. For it to be successful, a coordination of social behavior, physiology, and gamete production is necessary. How are social cues perceived and how do they affect physiology and gametogenesis? While females, ranging from insects to mammals, have provided multiple insights about this coordination, its existence remains largely unknown in males. Here, by using the Drosophila male as a model, we describe a phenomenon by which the availability of potential mating partners triggers an activation state on the stem cell populations of the testis, boosting spermatogenesis. We reveal its reliance on pheromonal communication, even in the absence of mating or other interactions with females. Finally, we identify the interorgan communication signaling network responsible-muscle-secreted tumor necrosis factor alpha (TNF-α)/Eiger and neuronally secreted octopamine trigger, respectively, the Jun N-terminal kinase (JNK) pathway and a change in calcium dynamics in the cyst stem cells. As a consequence, germ line stem cells increase their proliferation.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Espermatogénesis , Células Madre , Animales , Masculino , Espermatogénesis/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Madre/metabolismo , Femenino , Interacción Social , Testículo/metabolismo , Proliferación Celular , Conducta Sexual Animal/fisiología , Proteínas de la Membrana
3.
Cell Rep ; 43(8): 114558, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39088321

RESUMEN

Chromatin priming promotes cell-type-specific gene expression, lineage differentiation, and development. The mechanism of chromatin priming has not been fully understood. Here, we report that mouse hematopoietic stem and progenitor cells (HSPCs) lacking the Baf155 subunit of the BAF (BRG1/BRM-associated factor) chromatin remodeling complex produce a significantly reduced number of mature blood cells, leading to a failure of hematopoietic regeneration upon transplantation and 5-fluorouracil (5-FU) injury. Baf155-deficient HSPCs generate particularly fewer neutrophils, B cells, and CD8+ T cells at homeostasis, supporting a more immune-suppressive tumor microenvironment and enhanced tumor growth. Single-nucleus multiomics analysis reveals that Baf155-deficient HSPCs fail to establish accessible chromatin in selected regions that are enriched for putative enhancers and binding motifs of hematopoietic lineage transcription factors. Our study provides a fundamental mechanistic understanding of the role of Baf155 in hematopoietic lineage chromatin priming and the functional consequences of Baf155 deficiency in regeneration and tumor immunity.


Asunto(s)
Diferenciación Celular , Cromatina , Hematopoyesis , Células Madre Hematopoyéticas , Animales , Cromatina/metabolismo , Ratones , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Ratones Endogámicos C57BL , Regeneración , Fluorouracilo/farmacología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
4.
Cell Rep ; 43(8): 114582, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39096488

RESUMEN

Male infertility is a recognized side effect of chemoradiotherapy. Extant spermatogonial stem cells (SSCs) may act as originators for any subsequent recovery. However, which type of SSCs, the mechanism by which they survive and resist toxicity, and how they act to restart spermatogenesis remain largely unknown. Here, we identify a small population of Set domain-containing protein 4 (Setd4)-expressing SSCs that occur in a relatively dormant state in the mouse seminiferous tubule. Extant beyond high-dose chemoradiotherapy, these cells then activate to recover spermatogenesis. Recovery fails when Setd4+ SSCs are deleted. Confirmed to be of fetal origin, these Setd4+ SSCs are shown to facilitate early testicular development and also contribute to steady-state spermatogenesis in adulthood. Upon activation, chromatin remodeling increases their genome-wide accessibility, enabling Notch1 and Aurora activation with corresponding silencing of p21 and p53. Here, Setd4+ SSCs are presented as the originators of both testicular development and spermatogenesis recovery in chemoradiotherapy-induced infertility.


Asunto(s)
Infertilidad Masculina , Espermatogénesis , Masculino , Animales , Espermatogénesis/efectos de los fármacos , Espermatogénesis/efectos de la radiación , Infertilidad Masculina/terapia , Ratones , Quimioradioterapia/efectos adversos , Quimioradioterapia/métodos , Células Madre/metabolismo , Células Madre/efectos de la radiación , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/metabolismo , Receptor Notch1/metabolismo , Receptor Notch1/genética
5.
Cell Rep ; 43(8): 114569, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39088319

RESUMEN

Wound healing in response to acute injury is mediated by the coordinated and transient activation of parenchymal, stromal, and immune cells that resolves to homeostasis. Environmental, genetic, and epigenetic factors associated with inflammation and aging can lead to persistent activation of the microenvironment and fibrosis. Here, we identify opposing roles of interleukin-4 (IL-4) cytokine signaling in interstitial macrophages and type II alveolar epithelial cells (ATIIs). We show that IL4Ra signaling in macrophages promotes regeneration of the alveolar epithelium after bleomycin-induced lung injury. Using organoids and mouse models, we show that IL-4 directly acts on a subset of ATIIs to induce the expression of the transcription factor SOX9 and reprograms them toward a progenitor-like state with both airway and alveolar lineage potential. In the contexts of aging and bleomycin-induced lung injury, this leads to aberrant epithelial cell differentiation and bronchiolization, consistent with cellular and histological changes observed in interstitial lung disease.


Asunto(s)
Bleomicina , Linaje de la Célula , Interleucina-4 , Pulmón , Factor de Transcripción SOX9 , Animales , Interleucina-4/metabolismo , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Ratones , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Células Madre Adultas/metabolismo , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Envejecimiento/metabolismo , Diferenciación Celular , Transducción de Señal , Humanos , Macrófagos/metabolismo
6.
Cell Rep ; 43(8): 114542, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39046877

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is widely used to enhance myeloid recovery after chemotherapy and to mobilize hematopoietic stem cells (HSCs) for transplantation. Unfortunately, through the course of chemotherapy, cancer patients can acquire leukemogenic mutations that cause therapy-related myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). This raises the question of whether therapeutic G-CSF might potentiate therapy-related MDS/AML by disproportionately stimulating mutant HSCs and other myeloid progenitors. A common mutation in therapy-related MDS/AML involves chromosome 7 deletions that inactivate many tumor suppressor genes, including KMT2C. Here, we show that Kmt2c deletions hypersensitize murine HSCs and myeloid progenitors to G-CSF, as evidenced by increased HSC mobilization and enhanced granulocyte production from granulocyte-monocyte progenitors (GMPs). Furthermore, Kmt2c attenuates the G-CSF response independently from its SET methyltransferase function. Altogether, the data raise concerns that monosomy 7 can hypersensitize progenitors to G-CSF, such that clinical use of G-CSF may amplify the risk of therapy-related MDS/AML.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos , Granulocitos , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas , Factor Estimulante de Colonias de Granulocitos/metabolismo , Animales , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Granulocitos/metabolismo , Granulocitos/efectos de los fármacos , Ratones Endogámicos C57BL , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Humanos , Metiltransferasas/metabolismo , Metiltransferasas/genética
7.
Cell Rep ; 43(7): 114475, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38996072

RESUMEN

Endomucin (EMCN) currently represents the only hematopoietic stem cell (HSC) marker expressed by both murine and human HSCs. Here, we report that EMCN+ long-term repopulating HSCs (LT-HSCs; CD150+CD48-LSK) have a higher long-term multi-lineage repopulating capacity compared to EMCN- LT-HSCs. Cell cycle analyses and transcriptional profiling demonstrated that EMCN+ LT-HSCs were more quiescent compared to EMCN- LT-HSCs. Emcn-/- and Emcn+/+ mice displayed comparable steady-state hematopoiesis, as well as frequencies, transcriptional programs, and long-term multi-lineage repopulating capacity of their LT-HSCs. Complementary functional analyses further revealed increased cell cycle entry upon treatment with 5-fluorouracil and reduced granulocyte colony-stimulating factor (GCSF) mobilization of Emcn-/- LT-HSCs, demonstrating that EMCN expression by LT-HSCs associates with quiescence in response to hematopoietic stress and is indispensable for effective LT-HSC mobilization. Transplantation of wild-type bone marrow cells into Emcn-/- or Emcn+/+ recipients demonstrated that EMCN is essential for endothelial cell-dependent maintenance/self-renewal of the LT-HSC pool and sustained blood cell production post-transplant.


Asunto(s)
Linaje de la Célula , Hematopoyesis , Células Madre Hematopoyéticas , Animales , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C57BL , Movimiento Celular , Fluorouracilo/farmacología , Humanos , Factor Estimulante de Colonias de Granulocitos/metabolismo , Ciclo Celular , Células Endoteliales/metabolismo
8.
Cell Rep ; 43(7): 114247, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38907996

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Células Epiteliales , Células Madre Pluripotentes Inducidas , Intestino Delgado , Neuronas , Organoides , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Humanos , Intestino Delgado/citología , Intestino Delgado/metabolismo , Neuronas/metabolismo , Neuronas/citología , Células Epiteliales/metabolismo , Células Epiteliales/citología , Organoides/metabolismo , Organoides/citología , Dispositivos Laboratorio en un Chip , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citología
9.
Cell Rep ; 43(6): 114309, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38848215

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Glioblastoma , Metiltransferasas , Glioblastoma/patología , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Metiltransferasas/metabolismo , Metiltransferasas/genética , Linaje de la Célula/genética , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Línea Celular Tumoral , Astrocitos/metabolismo , Astrocitos/patología , Organoides/metabolismo , Organoides/patología
10.
Cell Rep ; 43(7): 114388, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38935497

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Daño del ADN , Células Madre Hematopoyéticas , Megacariocitos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Animales , Ratones , Megacariocitos/metabolismo , Megacariocitos/citología , Trombopoyesis , Puntos de Control de la Fase G2 del Ciclo Celular , Ratones Endogámicos C57BL , Humanos
11.
Cell Rep ; 43(6): 114340, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38865239

RESUMEN

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.


Asunto(s)
Blastocisto , Factor Nuclear 3-beta del Hepatocito , Células Madre Pluripotentes , Glándulas Salivales , Animales , Glándulas Salivales/citología , Glándulas Salivales/metabolismo , Blastocisto/metabolismo , Blastocisto/citología , Ratones , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Linaje de la Célula , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética
12.
Cell Rep ; 43(6): 114372, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38878289

RESUMEN

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.


Asunto(s)
Genoma , Técnicas de Transferencia Nuclear , Animales , Porcinos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Desarrollo Embrionario/genética , Embrión de Mamíferos/metabolismo , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Acetilación , Clonación de Organismos/métodos , Histonas/metabolismo , Blastocisto/metabolismo
13.
Cell Rep ; 43(5): 114177, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38691453

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Complejo Mediador , Desarrollo de Músculos , Regeneración , Células Madre , Animales , Ratones , Desarrollo de Músculos/genética , Células Madre/metabolismo , Células Madre/citología , Complejo Mediador/metabolismo , Complejo Mediador/genética , Músculo Esquelético/metabolismo , Transcripción Genética , Ratones Endogámicos C57BL , Transactivadores/metabolismo , Transactivadores/genética
14.
Cell Rep ; 43(5): 114170, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38700983

RESUMEN

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.


Asunto(s)
Reprogramación Celular , Ensamble y Desensamble de Cromatina , Histonas , Humanos , Reprogramación Celular/genética , Histonas/metabolismo , Análisis de la Célula Individual , Activación Transcripcional , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Cromatina/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteína Homeótica Nanog/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología
15.
Cell Rep ; 43(5): 114227, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38735044

RESUMEN

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.


Asunto(s)
Cromatina , Células Madre Hematopoyéticas , Proteínas de Homeodominio , Proteínas Represoras , Humanos , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Cromatina/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Animales , Ratones , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos/genética
16.
Cell Rep ; 43(5): 114219, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38748874

RESUMEN

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.


Asunto(s)
Encéfalo , Diferenciación Celular , Organoides , Humanos , Organoides/metabolismo , Encéfalo/metabolismo , Encéfalo/embriología , Animales , Ratones , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Proteoma/metabolismo , Transducción de Señal , Transcriptoma/genética , Proteómica/métodos , Neurogénesis , Proteínas Proto-Oncogénicas c-akt/metabolismo
17.
Cell Rep ; 43(5): 114232, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38761378

RESUMEN

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.


Asunto(s)
Trofoblastos , Humanos , Trofoblastos/metabolismo , Trofoblastos/citología , Metilación de ADN , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Modelos Biológicos , Implantación del Embrión , Diferenciación Celular , Epigénesis Genética , Transcriptoma/genética , Proteómica/métodos
18.
Cell Rep ; 43(6): 114270, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38787726

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/terapia , Humanos , Animales , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Fetales/metabolismo
19.
Cell Rep ; 43(4): 114031, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38583153

RESUMEN

Outer radial glia (oRG) emerge as cortical progenitor cells that support the development of an enlarged outer subventricular zone (oSVZ) and the expansion of the neocortex. The in vitro generation of oRG is essential to investigate the underlying mechanisms of human neocortical development and expansion. By activating the STAT3 signaling pathway using leukemia inhibitory factor (LIF), which is not expressed in guided cortical organoids, we define a cortical organoid differentiation method from human pluripotent stem cells (hPSCs) that recapitulates the expansion of a progenitor pool into the oSVZ. The oSVZ comprises progenitor cells expressing specific oRG markers such as GFAP, LIFR, and HOPX, closely matching human fetal oRG. Finally, incorporating neural crest-derived LIF-producing cortical pericytes into cortical organoids recapitulates the effects of LIF treatment. These data indicate that increasing the cellular complexity of the organoid microenvironment promotes the emergence of oRG and supports a platform to study oRG in hPSC-derived brain organoids routinely.


Asunto(s)
Diferenciación Celular , Ventrículos Laterales , Factor Inhibidor de Leucemia , Organoides , Células Madre Pluripotentes , Humanos , Organoides/metabolismo , Organoides/citología , Factor Inhibidor de Leucemia/metabolismo , Factor Inhibidor de Leucemia/farmacología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo , Factor de Transcripción STAT3/metabolismo , Neuroglía/metabolismo , Neuroglía/citología , Transducción de Señal
20.
Cell Rep ; 43(5): 114136, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38643480

RESUMEN

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.


Asunto(s)
Factor de Transcripción CDX2 , Linaje de la Célula , Cromatina , Regulación del Desarrollo de la Expresión Génica , Factor de Transcripción AP-2 , Animales , Cromatina/metabolismo , Ratones , Linaje de la Célula/genética , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción CDX2/metabolismo , Factor de Transcripción CDX2/genética , Proteína Homeótica Nanog/metabolismo , Proteína Homeótica Nanog/genética , Blastocisto/metabolismo , Blastocisto/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Femenino , Histonas/metabolismo , Diferenciación Celular/genética , Ectodermo/metabolismo , Ectodermo/citología , Desarrollo Embrionario/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA