Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.199
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(11): 2855-2874.e19, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38657603

RESUMEN

Progress in understanding early human development has been impeded by the scarcity of reference datasets from natural embryos, particularly those with spatial information during crucial stages like gastrulation. We conducted high-resolution spatial transcriptomics profiling on 38,562 spots from 62 transverse sections of an intact Carnegie stage (CS) 8 human embryo. From this spatial transcriptomic dataset, we constructed a 3D model of the CS8 embryo, in which a range of cell subtypes are identified, based on gene expression patterns and positional register, along the anterior-posterior, medial-lateral, and dorsal-ventral axis in the embryo. We further characterized the lineage trajectories of embryonic and extra-embryonic tissues and associated regulons and the regionalization of signaling centers and signaling activities that underpin lineage progression and tissue patterning during gastrulation. Collectively, the findings of this study provide insights into gastrulation and post-gastrulation development of the human embryo.


Asunto(s)
Embrión de Mamíferos , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Imagenología Tridimensional , Humanos , Embrión de Mamíferos/metabolismo , Transcriptoma/genética , Gástrula/metabolismo , Gástrula/embriología , Transducción de Señal , Linaje de la Célula , Perfilación de la Expresión Génica , Tipificación del Cuerpo/genética
2.
Cell ; 186(10): 2092-2110.e23, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37172563

RESUMEN

The third and fourth weeks of gestation in primates are marked by several developmental milestones, including gastrulation and the formation of organ primordia. However, our understanding of this period is limited due to restricted access to in vivo embryos. To address this gap, we developed an embedded 3D culture system that allows for the extended ex utero culture of cynomolgus monkey embryos for up to 25 days post-fertilization. Morphological, histological, and single-cell RNA-sequencing analyses demonstrate that ex utero cultured monkey embryos largely recapitulated key events of in vivo development. With this platform, we were able to delineate lineage trajectories and genetic programs involved in neural induction, lateral plate mesoderm differentiation, yolk sac hematopoiesis, primitive gut, and primordial germ-cell-like cell development in monkeys. Our embedded 3D culture system provides a robust and reproducible platform for growing monkey embryos from blastocysts to early organogenesis and studying primate embryogenesis ex utero.


Asunto(s)
Embrión de Mamíferos , Desarrollo Embrionario , Animales , Macaca fascicularis , Blastocisto , Organogénesis , Primates
3.
Cell ; 186(18): 3776-3792.e16, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37478861

RESUMEN

In vitro stem cell models that replicate human gastrulation have been generated, but they lack the essential extraembryonic cells needed for embryonic development, morphogenesis, and patterning. Here, we describe a robust and efficient method that prompts human extended pluripotent stem cells to self-organize into embryo-like structures, termed peri-gastruloids, which encompass both embryonic (epiblast) and extraembryonic (hypoblast) tissues. Although peri-gastruloids are not viable due to the exclusion of trophoblasts, they recapitulate critical stages of human peri-gastrulation development, such as forming amniotic and yolk sac cavities, developing bilaminar and trilaminar embryonic discs, specifying primordial germ cells, initiating gastrulation, and undergoing early neurulation and organogenesis. Single-cell RNA-sequencing unveiled transcriptomic similarities between advanced human peri-gastruloids and primary peri-gastrulation cell types found in humans and non-human primates. This peri-gastruloid platform allows for further exploration beyond gastrulation and may potentially aid in the development of human fetal tissues for use in regenerative medicine.


Asunto(s)
Implantación del Embrión , Gastrulación , Células Madre Pluripotentes , Animales , Femenino , Humanos , Embarazo , Diferenciación Celular , Embrión de Mamíferos , Desarrollo Embrionario , Organogénesis , Células Madre Pluripotentes/metabolismo , Primates
4.
Annu Rev Biochem ; 87: 809-837, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29596003

RESUMEN

To maintain an asymmetric distribution of ions across membranes, protein pumps displace ions against their concentration gradient by using chemical energy. Here, we describe a functionally analogous but topologically opposite process that applies to the lipid transfer protein (LTP) oxysterol-binding protein (OSBP). This multidomain protein exchanges cholesterol for the phosphoinositide phosphatidylinositol 4-phosphate [PI(4)P] between two apposed membranes. Because of the subsequent hydrolysis of PI(4)P, this counterexchange is irreversible and contributes to the establishment of a cholesterol gradient along organelles of the secretory pathway. The facts that some natural anti-cancer molecules block OSBP and that many viruses hijack the OSBP cycle for the formation of intracellular replication organelles highlight the importance and potency of OSBP-mediated lipid exchange. The architecture of some LTPs is similar to that of OSBP, suggesting that the principles of the OSBP cycle-burning PI(4)P for the vectorial transfer of another lipid-might be general.


Asunto(s)
Colesterol/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Receptores de Esteroides/metabolismo , Transporte Biológico Activo , Proteínas Portadoras/metabolismo , Aparato de Golgi/metabolismo , Humanos , Ligandos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Modelos Moleculares , Oxiesteroles/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptores de Esteroides/química , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Replicación Viral/fisiología
5.
Immunity ; 54(7): 1433-1446.e5, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34062116

RESUMEN

The extra-embryonic yolk sac contains the first definitive multipotent hematopoietic cells, denominated erythromyeloid progenitors. They originate in situ prior to the emergence of hematopoietic stem cells and give rise to erythroid, monocytes, granulocytes, mast cells and macrophages, the latter in a Myb transcription factor-independent manner. We uncovered here the heterogeneity of yolk sac erythromyeloid progenitors, at the single cell level, and discriminated multipotent from committed progenitors, prior to fetal liver colonization. We identified two temporally distinct megakaryocyte differentiation pathways. The first occurs in the yolk sac, bypasses intermediate bipotent megakaryocyte-erythroid progenitors and, similar to the differentiation of macrophages, is Myb-independent. By contrast, the second originates later, from Myb-dependent bipotent progenitors expressing Csf2rb and colonize the fetal liver, where they give rise to megakaryocytes and to large numbers of erythrocytes. Understanding megakaryocyte development is crucial as they play key functions during vascular development, in particular in separating blood and lymphatic networks.


Asunto(s)
Diferenciación Celular/fisiología , Eritrocitos/citología , Megacariocitos/citología , Células Mieloides/citología , Células Madre/citología , Saco Vitelino/citología , Animales , Linaje de la Célula/fisiología , Células Cultivadas , Embrión de Mamíferos/citología , Femenino , Granulocitos/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Células Madre Multipotentes/citología , Embarazo
6.
EMBO J ; 43(5): 666-694, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38279026

RESUMEN

The efficacy of current antimitotic cancer drugs is limited by toxicity in highly proliferative healthy tissues. A cancer-specific dependency on the microtubule motor protein KIF18A therefore makes it an attractive therapeutic target. Not all cancers require KIF18A, however, and the determinants underlying this distinction remain unclear. Here, we show that KIF18A inhibition drives a modest and widespread increase in spindle assembly checkpoint (SAC) signaling from kinetochores which can result in lethal mitotic delays. Whether cells arrest in mitosis depends on the robustness of the metaphase-to-anaphase transition, and cells predisposed with weak basal anaphase-promoting complex/cyclosome (APC/C) activity and/or persistent SAC signaling through metaphase are uniquely sensitive to KIF18A inhibition. KIF18A-dependent cancer cells exhibit hallmarks of this SAC:APC/C imbalance, including a long metaphase-to-anaphase transition, and slow mitosis overall. Together, our data reveal vulnerabilities in the cell division apparatus of cancer cells that can be exploited for therapeutic benefit.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase , Neoplasias , Humanos , Ciclosoma-Complejo Promotor de la Anafase/genética , Dineínas , Cinesinas/genética , Cinetocoros , Mitosis , Neoplasias/genética
7.
Mol Cell ; 77(3): 618-632.e5, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31806350

RESUMEN

TMEM39A, encoding an ER-localized transmembrane protein, is a susceptibility locus for multiple autoimmune diseases. The molecular function of TMEM39A remains completely unknown. Here we demonstrated that TMEM39A, also called SUSR2, modulates autophagy activity by regulating the spatial distribution and levels of PtdIns(4)P. Depletion of SUSR2 elevates late endosomal/lysosomal PtdIns(4)P levels, facilitating recruitment of the HOPS complex to promote assembly of the SNARE complex for autophagosome maturation. SUSR2 knockdown also increases the degradative capability of lysosomes. Mechanistically, SUSR2 interacts with the ER-localized PtdIns(4)P phosphatase SAC1 and also the COPII SEC23/SEC24 subunits to promote the ER-to-Golgi transport of SAC1. Retention of SAC1 on the ER in SUSR2 knockdown cells increases the level of PtdIns(3)P produced by the VPS34 complex, promoting autophagosome formation. Our study reveals that TMEM39A/SUSR2 acts as an adaptor protein for efficient export of SAC1 from the ER and provides insights into the pathogenesis of diseases associated with TMEM39A mutations.


Asunto(s)
Autofagia/fisiología , Proteínas de la Membrana/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células COS , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Células HEK293 , Células HeLa , Humanos , Lisosomas/metabolismo , Proteínas de la Membrana/fisiología , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/fisiología , Transporte de Proteínas/fisiología
8.
Development ; 151(6)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38546043

RESUMEN

The timely degradation of proteins that regulate the cell cycle is essential for oocyte maturation. Oocytes are equipped to degrade proteins via the ubiquitin-proteasome system. In meiosis, anaphase promoting complex/cyclosome (APC/C), an E3 ubiquitin-ligase, is responsible for the degradation of proteins. Ubiquitin-conjugating enzyme E2 S (UBE2S), an E2 ubiquitin-conjugating enzyme, delivers ubiquitin to APC/C. APC/C has been extensively studied, but the functions of UBE2S in oocyte maturation and mouse fertility are not clear. In this study, we used Ube2s knockout mice to explore the role of UBE2S in mouse oocytes. Ube2s-deleted oocytes were characterized by meiosis I arrest with normal spindle assembly and spindle assembly checkpoint dynamics. However, the absence of UBE2S affected the activity of APC/C. Cyclin B1 and securin are two substrates of APC/C, and their levels were consistently high, resulting in the failure of homologous chromosome separation. Unexpectedly, the oocytes arrested in meiosis I could be fertilized and the embryos could become implanted normally, but died before embryonic day 10.5. In conclusion, our findings reveal an indispensable regulatory role of UBE2S in mouse oocyte meiosis and female fertility.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular , Meiosis , Animales , Femenino , Ratones , Ciclosoma-Complejo Promotor de la Anafase/genética , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Oocitos/metabolismo , Ubiquitinas/metabolismo
9.
Immunity ; 49(4): 640-653.e5, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332630

RESUMEN

Tissue-resident mast cells are associated with many inflammatory and physiological processes. Although mast cells arise from the yolk sac, the exact ontogeny of adult mast cells remains unclear. Here we have investigated the hematopoietic origin of mast cells using fate-mapping systems. We have shown that early erythro-myeloid progenitors (EMPs), late EMPs, and definitive hematopoietic stem cells (HSCs) each gave rise to mast cells in succession via an intermediate integrin ß7+ progenitor. From late embryogenesis to adult, early EMP-derived mast cells were largely replaced by late EMP-derived cells in most connective tissues except adipose and pleural cavity. Thus, mast cells with distinct origin displayed tissue-location preferences: early EMP-derived cells were limited to adipose and pleural cavity and late EMP-derived cells dominated most connective tissues, while HSC-derived cells were a main group in mucosa. Therefore, embryonic origin shapes the heterogeneity of adult mast cells, with diverse functions in immunity and development.


Asunto(s)
Células Eritroides/inmunología , Mastocitos/inmunología , Células Progenitoras Mieloides/inmunología , Animales , Linaje de la Célula/inmunología , Células Cultivadas , Tejido Conectivo/inmunología , Tejido Conectivo/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/inmunología , Células Eritroides/citología , Células Eritroides/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Cadenas beta de Integrinas/inmunología , Cadenas beta de Integrinas/metabolismo , Mastocitos/citología , Mastocitos/metabolismo , Ratones Transgénicos , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo
10.
Immunity ; 48(6): 1160-1171.e5, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29858009

RESUMEN

Hematopoiesis occurs in distinct waves. "Definitive" hematopoietic stem cells (HSCs) with the potential for all blood lineages emerge in the aorta-gonado-mesonephros, while "primitive" progenitors, whose potential is thought to be limited to erythrocytes, megakaryocytes, and macrophages, arise earlier in the yolk sac (YS). Here, we questioned whether other YS lineages exist that have not been identified, partially owing to limitations of current lineage tracing models. We established the use of Cdh5-CreERT2 for hematopoietic fate mapping, which revealed the YS origin of mast cells (MCs). YS-derived MCs were replaced by definitive MCs, which maintained themselves independently from the bone marrow in the adult. Replacement occurred with tissue-specific kinetics. MCs in the embryonic skin, but not other organs, remained largely YS derived prenatally and were phenotypically and transcriptomically distinct from definite adult MCs. We conclude that within myeloid lineages, dual hematopoietic origin is shared between macrophages and MCs.


Asunto(s)
Linaje de la Célula/inmunología , Hematopoyesis/fisiología , Mastocitos/citología , Animales , Hemangioblastos/citología , Células Madre Hematopoyéticas/citología , Macrófagos/citología , Macrófagos/inmunología , Mastocitos/inmunología , Ratones , Piel/citología , Piel/inmunología , Saco Vitelino/citología , Saco Vitelino/embriología
11.
Proc Natl Acad Sci U S A ; 121(2): e2316583121, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38170753

RESUMEN

The kinetochore scaffold 1 (KNL1) protein recruits spindle assembly checkpoint (SAC) proteins to ensure accurate chromosome segregation during mitosis. Despite such a conserved function among eukaryotic organisms, its molecular architectures have rapidly evolved so that the functional mode of plant KNL1 is largely unknown. To understand how SAC signaling is regulated at kinetochores, we characterized the function of the KNL1 gene in Arabidopsis thaliana. The KNL1 protein was detected at kinetochores throughout the mitotic cell cycle, and null knl1 mutants were viable and fertile but exhibited severe vegetative and reproductive defects. The mutant cells showed serious impairments of chromosome congression and segregation, that resulted in the formation of micronuclei. In the absence of KNL1, core SAC proteins were no longer detected at the kinetochores, and the SAC was not activated by unattached or misaligned chromosomes. Arabidopsis KNL1 interacted with SAC essential proteins BUB3.3 and BMF3 through specific regions that were not found in known KNL1 proteins of other species, and recruited them independently to kinetochores. Furthermore, we demonstrated that upon ectopic expression, the KNL1 homolog from the dicot tomato was able to functionally substitute KNL1 in A. thaliana, while others from the monocot rice or moss associated with kinetochores but were not functional, as reflected by sequence variations of the kinetochore proteins in different plant lineages. Our results brought insights into understanding the rapid evolution and lineage-specific connection between KNL1 and the SAC signaling molecules.


Asunto(s)
Arabidopsis , Arabidopsis/genética , Arabidopsis/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Mitosis , Cinetocoros/metabolismo , Huso Acromático/genética , Huso Acromático/metabolismo , Segregación Cromosómica
12.
Immunol Rev ; 315(1): 71-78, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36705244

RESUMEN

The Innate Lymphoid Cell (ILC) family is a relatively recently described immune cell family involved in innate immune responses and tissue homeostasis. Lymphoid Tissue Inducer (LTi) cells are part of the type 3 (ILC3) family. The ILC3 family is the main ILC population within the embryo, in which the LTi cells are critically associated with embryonic lymph node formation. Recent studies have shown more insights in ILC origin and residency from local embryonic and tissue resident precursors. Embryonic LTi cells originating from a different hemogenic endothelial source were shown to be replaced by HSC derived progenitors in adult. This review will discuss the layered origin of the ILC3 family with an emphasis on the LTi cell lineage.


Asunto(s)
Inmunidad Innata , Linfocitos , Humanos , Linfocitos T Colaboradores-Inductores , Tejido Linfoide , Linaje de la Célula
13.
Trends Biochem Sci ; 47(4): 352-366, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35184951

RESUMEN

Benzimidazole 1 (BUB1) and budding uninhibited by benzimidazole 1-related 1 (BUBR1) are multidomain paralogs with key roles in chromosome alignment during mitosis and the spindle assembly checkpoint (SAC), an evolutionarily conserved signaling pathway that monitors errors in chromosome segregation during cell division in eukaryotes. Although BUB1 and BUBR1 share a similar domain organization and short linear interaction motifs (SLiMs), they control distinct aspects of chromosome congression and the SAC. Here we discuss the roles of BUB1 and BUBR1 SLiMs in mitosis and complement this with additional insights gleamed from studying their evolution. We show that BUB1 and BUBR1 SLiMs form highly specific interactions that are carefully orchestrated in space and time and contend that they define BUB1 and BUBR1 as organizing hubs that drive SAC signaling and ensure genome stability.


Asunto(s)
Mitosis , Proteínas Serina-Treonina Quinasas , Proteínas de Ciclo Celular/metabolismo , Segregación Cromosómica , Cinetocoros/metabolismo , Transducción de Señal , Huso Acromático/metabolismo
14.
Traffic ; 25(1): e12923, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37926951

RESUMEN

Phosphoinositides are lipid signaling molecules acting at the interface of membranes and the cytosol to regulate membrane trafficking, lipid transport and responses to extracellular stimuli. Peroxisomes are multicopy organelles that are highly responsive to changes in metabolic and environmental conditions. In yeast, peroxisomes are tethered to the cell cortex at defined focal structures containing the peroxisome inheritance protein, Inp1p. We investigated the potential impact of changes in cortical phosphoinositide levels on the peroxisome compartment of the yeast cell. Here we show that the phosphoinositide, phosphatidylinositol-4-phosphate (PI4P), found at the junction of the cortical endoplasmic reticulum and plasma membrane (cER-PM) acts to regulate the cell's peroxisome population. In cells lacking a cER-PM tether or the enzymatic activity of the lipid phosphatase Sac1p, cortical PI4P is elevated, peroxisome numbers and motility are increased, and peroxisomes are no longer firmly tethered to Inp1p-containing foci. Reattachment of the cER to the PM through an artificial ER-PM "staple" in cells lacking the cER-PM tether does not restore peroxisome populations to the wild-type condition, demonstrating that integrity of PI4P signaling at the cell cortex is required for peroxisome homeostasis.


Asunto(s)
Peroxisomas , Fosfatidilinositoles , Fosfatidilinositoles/metabolismo , Peroxisomas/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de la Membrana/metabolismo , Regulación de la Población , Retículo Endoplásmico/metabolismo , Membrana Celular/metabolismo
15.
Development ; 150(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36515557

RESUMEN

The spindle assembly checkpoint (SAC) is a surveillance system that preserves genome integrity by delaying anaphase onset until all chromosomes are correctly attached to spindle microtubules. Recruitment of SAC proteins to unattached kinetochores generates an inhibitory signal that prolongs mitotic duration. Chordate embryos are atypical in that spindle defects do not delay mitotic progression during early development, implying that either the SAC is inactive or the cell-cycle target machinery is unresponsive. Here, we show that in embryos of the chordate Phallusia mammillata, the SAC delays mitotic progression from the 8th cleavage divisions. Unattached kinetochores are not recognized by the SAC machinery until the 7th cell cycle, when the SAC is acquired. After acquisition, SAC strength, which manifests as the degree of mitotic lengthening induced by spindle perturbations, is specific to different cell types and is modulated by cell size, showing similarity to SAC control in early Caenorhabditis elegans embryos. We conclude that SAC acquisition is a process that is likely specific to chordate embryos, while modulation of SAC efficiency in SAC proficient stages depends on cell fate and cell size, which is similar to non-chordate embryos.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular , Huso Acromático , Animales , Huso Acromático/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Caenorhabditis elegans/metabolismo , Tamaño de la Célula , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
16.
J Biol Chem ; 300(1): 105497, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38016514

RESUMEN

For many decades, our understanding of G protein-coupled receptor (GPCR) activity and cyclic AMP (cAMP) signaling was limited exclusively to the plasma membrane. However, a growing body of evidence has challenged this view by introducing the concept of endocytosis-dependent GPCR signaling. This emerging paradigm emphasizes not only the sustained production of cAMP but also its precise subcellular localization, thus transforming our understanding of the spatiotemporal organization of this process. Starting from this alternative point of view, our recent work sheds light on the role of an endocytosis-dependent calcium release from the endoplasmic reticulum in the control of nuclear cAMP levels. This is achieved through the activation of local soluble adenylyl cyclase, which in turn regulates the activation of local protein kinase A (PKA) and downstream transcriptional events. In this review, we explore the dynamic evolution of research on cyclic AMP signaling, including the findings that led us to formulate the novel three-wave hypothesis. We delve into how we abandoned the paradigm of cAMP generation limited to the plasma membrane and the changing perspectives on the rate-limiting step in nuclear PKA activation.


Asunto(s)
Membrana Celular , AMP Cíclico , Transducción de Señal , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Núcleo Celular/metabolismo
17.
Chromosoma ; 133(1): 77-92, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37256347

RESUMEN

Chromosome gains or losses often lead to copy number variations (CNV) and loss of heterozygosity (LOH). Both quantities are low in hematologic "liquid" cancers versus solid tumors in data of The Cancer Genome Atlas (TCGA) that also shows the fraction of a genome affected by LOH is ~ one-half of that with CNV. Suspension cultures of p53-null THP-1 leukemia-derived cells conform to these trends, despite novel evidence here of genetic heterogeneity and transiently elevated CNV after perturbation. Single-cell DNAseq indeed reveals at least 8 distinct THP-1 aneuploid clones with further intra-clonal variation, suggesting ongoing genetic evolution. Importantly, acute inhibition of the mitotic spindle assembly checkpoint (SAC) produces CNV levels that are typical of high-CNV solid tumors, with subsequent cell death and down-selection to novel CNV. Pan-cancer analyses show p53 inactivation associates with aneuploidy, but leukemias exhibit a weaker trend even though p53 inactivation correlates with poor survival. Overexpression of p53 in THP-1 does not rescue established aneuploidy or LOH but slightly increases cell death under oxidative or confinement stress, and triggers p21, a key p53 target, but without affecting net growth. Our results suggest that factors other than p53 exert stronger pressures against aneuploidy in liquid cancers, and identifying such CNV suppressors could be useful across liquid and solid tumor types.


Asunto(s)
Leucemia , Neoplasias , Humanos , Puntos de Control de la Fase M del Ciclo Celular , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Variaciones en el Número de Copia de ADN , Heterogeneidad Genética , Aneuploidia , Neoplasias/genética , Neoplasias/metabolismo , Leucemia/genética , Leucemia/metabolismo , Huso Acromático/metabolismo
18.
Trends Genet ; 38(11): 1099-1100, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35792016

RESUMEN

A recent study by Hu et al. describes N6-methyladenosine (m6A)-selective allyl chemical labeling and sequencing (m6A-SAC-seq), which allows for quantitative, stoichiometric, and positional analyses of m6A at single-nucleotide resolution across the whole transcriptome level. Information on the m6A stoichiometry will provide additional layers of gene regulatory pathways mediated by m6A modification during diverse molecular, cellular, and physiological events.


Asunto(s)
Adenosina , Transcriptoma , Adenosina/genética , Adenosina/metabolismo , Nucleótidos , Transcriptoma/genética
19.
Development ; 149(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35502779

RESUMEN

The mechanisms underlying bone development, repair and regeneration are reliant on the interplay and communication between osteoclasts and other surrounding cells. Osteoclasts are multinucleated monocyte lineage cells with resorptive abilities, forming the bone marrow cavity during development. This marrow cavity, essential to hematopoiesis and osteoclast-osteoblast interactions, provides a setting to investigate the origin of osteoclasts and their multi-faceted roles. This Review examines recent developments in the embryonic understanding of osteoclast origin, as well as interactions within the immune environment to regulate normal and pathological bone development, homeostasis and repair.


Asunto(s)
Resorción Ósea , Osteoclastos , Desarrollo Óseo , Resorción Ósea/patología , Diferenciación Celular/fisiología , Homeostasis , Humanos , Osteoclastos/patología
20.
EMBO Rep ; 24(3): e56007, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36588479

RESUMEN

Legionella pneumophila replicates in macrophages and amoeba within a unique compartment, the Legionella-containing vacuole (LCV). Hallmarks of LCV formation are the phosphoinositide lipid conversion from PtdIns(3)P to PtdIns(4)P, fusion with ER-derived vesicles and a tight association with the ER. Proteomics of purified LCVs indicate the presence of membrane contact sites (MCS) proteins possibly implicated in lipid exchange. Using dually fluorescence-labeled Dictyostelium discoideum amoeba, we reveal that VAMP-associated protein (Vap) and the PtdIns(4)P 4-phosphatase Sac1 localize to the ER, and Vap also localizes to the LCV membrane. Furthermore, Vap as well as Sac1 promote intracellular replication of L. pneumophila and LCV remodeling. Oxysterol binding proteins (OSBPs) preferentially localize to the ER (OSBP8) or the LCV membrane (OSBP11), respectively, and restrict (OSBP8) or promote (OSBP11) bacterial replication and LCV expansion. The sterol probes GFP-D4H* and filipin indicate that sterols are rapidly depleted from LCVs, while PtdIns(4)P accumulates. In addition to Sac1, the PtdIns(4)P-subverting L. pneumophila effector proteins LepB and SidC also support LCV remodeling. Taken together, the Legionella- and host cell-driven PtdIns(4)P gradient at LCV-ER MCSs promotes Vap-, OSBP- and Sac1-dependent pathogen vacuole maturation.


Asunto(s)
Dictyostelium , Legionella pneumophila , Legionella , Vacuolas/metabolismo , Legionella/metabolismo , Dictyostelium/microbiología , Fosfatidilinositoles/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Bacterianas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA