Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.834
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Mol Cell ; 81(17): 3589-3603.e13, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34324863

RESUMO

Transcription elongation has emerged as a regulatory hub in gene expression of metazoans. A major control point occurs during early elongation before RNA polymerase II (Pol II) is released into productive elongation. Prior research has linked BRD4 with transcription elongation. Here, we use rapid BET protein and BRD4-selective degradation along with quantitative genome-wide approaches to investigate direct functions of BRD4 in Pol II transcription regulation. Notably, as an immediate consequence of acute BRD4 loss, promoter-proximal pause release is impaired, and transcriptionally engaged Pol II past this checkpoint undergoes readthrough transcription. An integrated proteome-wide analysis uncovers elongation and 3'-RNA processing factors as core BRD4 interactors. BRD4 ablation disrupts the recruitment of general 3'-RNA processing factors at the 5'-control region, which correlates with RNA cleavage and termination defects. These studies, performed in human cells, reveal a BRD4-mediated checkpoint and begin to establish a molecular link between 5'-elongation control and 3'-RNA processing.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Nucleares/fisiologia , Elongação da Transcrição Genética/fisiologia , Fatores de Transcrição/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Expressão Gênica , Histonas/metabolismo , Humanos , Camundongos , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , RNA Polimerase II/metabolismo , RNA Polimerase II/fisiologia , Fatores de Transcrição/metabolismo , Terminação da Transcrição Genética/fisiologia , Transcrição Gênica/genética , Transcrição Gênica/fisiologia
2.
Mol Cell ; 79(2): 234-250.e9, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32579944

RESUMO

Somatic cell nuclear transfer (SCNT) can reprogram a somatic nucleus to a totipotent state. However, the re-organization of 3D chromatin structure in this process remains poorly understood. Using low-input Hi-C, we revealed that, during SCNT, the transferred nucleus first enters a mitotic-like state (premature chromatin condensation). Unlike fertilized embryos, SCNT embryos show stronger topologically associating domains (TADs) at the 1-cell stage. TADs become weaker at the 2-cell stage, followed by gradual consolidation. Compartments A/B are markedly weak in 1-cell SCNT embryos and become increasingly strengthened afterward. By the 8-cell stage, somatic chromatin architecture is largely reset to embryonic patterns. Unexpectedly, we found cohesin represses minor zygotic genome activation (ZGA) genes (2-cell-specific genes) in pluripotent and differentiated cells, and pre-depleting cohesin in donor cells facilitates minor ZGA and SCNT. These data reveal multi-step reprogramming of 3D chromatin architecture during SCNT and support dual roles of cohesin in TAD formation and minor ZGA repression.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Cromatina/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Técnicas de Transferência Nuclear , Zigoto/fisiologia , Animais , Linhagem Celular , Núcleo Celular , Montagem e Desmontagem da Cromatina , Biologia Computacional/métodos , Conjuntos de Dados como Assunto , Desenvolvimento Embrionário , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Coesinas
3.
Mol Cell ; 77(4): 825-839.e7, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31837995

RESUMO

In mammals, chromatin organization undergoes drastic reorganization during oocyte development. However, the dynamics of three-dimensional chromatin structure in this process is poorly characterized. Using low-input Hi-C (genome-wide chromatin conformation capture), we found that a unique chromatin organization gradually appears during mouse oocyte growth. Oocytes at late stages show self-interacting, cohesin-independent compartmental domains marked by H3K27me3, therefore termed Polycomb-associating domains (PADs). PADs and inter-PAD (iPAD) regions form compartment-like structures with strong inter-domain interactions among nearby PADs. PADs disassemble upon meiotic resumption from diplotene arrest but briefly reappear on the maternal genome after fertilization. Upon maternal depletion of Eed, PADs are largely intact in oocytes, but their reestablishment after fertilization is compromised. By contrast, depletion of Polycomb repressive complex 1 (PRC1) proteins attenuates PADs in oocytes, which is associated with substantial gene de-repression in PADs. These data reveal a critical role of Polycomb in regulating chromatin architecture during mammalian oocyte growth and early development.


Assuntos
Cromatina/química , Oócitos/crescimento & desenvolvimento , Oogênese/genética , Proteínas do Grupo Polycomb/fisiologia , Animais , Blastocisto/química , Proteínas de Ciclo Celular/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Embrião de Mamíferos/química , Inativação Gênica , Código das Histonas , Camundongos , Oócitos/química , Transcrição Gênica , Coesinas
4.
Mol Cell ; 80(3): 396-409.e6, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33108759

RESUMO

Cytokine activation of cells induces gene networks involved in inflammation and immunity. Transient gene activation can have a lasting effect even in the absence of ongoing transcription, known as long-term transcriptional memory. Here we explore the nature of the establishment and maintenance of interferon γ (IFNγ)-induced priming of human cells. We find that, although ongoing transcription and local chromatin signatures are short-lived, the IFNγ-primed state stably propagates through at least 14 cell division cycles. Single-cell analysis reveals that memory is manifested by an increased probability of primed cells to engage in target gene expression, correlating with the strength of initial gene activation. Further, we find that strongly memorized genes tend to reside in genomic clusters and that long-term memory of these genes is locally restricted by cohesin. We define the duration, stochastic nature, and molecular mechanisms of IFNγ-induced transcriptional memory, relevant to understanding enhanced innate immune signaling.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Interferon gama/metabolismo , Ativação Transcricional/genética , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular , Cromatina/genética , Proteínas Cromossômicas não Histona/fisiologia , Regulação da Expressão Gênica/imunologia , Células HeLa , Humanos , Inflamação , Interferon gama/fisiologia , Ligação Proteica/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/genética , Transcrição Gênica/genética , Ativação Transcricional/fisiologia , Coesinas
5.
Mol Cell ; 78(1): 127-140.e7, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32035037

RESUMO

As cells enter mitosis, the genome is restructured to facilitate chromosome segregation, accompanied by dramatic changes in gene expression. However, the mechanisms that underlie mitotic transcriptional regulation are unclear. In contrast to transcribed genes, centromere regions retain transcriptionally active RNA polymerase II (Pol II) in mitosis. Here, we demonstrate that chromatin-bound cohesin is necessary to retain elongating Pol II at centromeres. We find that WAPL-mediated removal of cohesin from chromosome arms during prophase is required for the dissociation of Pol II and nascent transcripts, and failure of this process dramatically alters mitotic gene expression. Removal of cohesin/Pol II from chromosome arms in prophase is important for accurate chromosome segregation and normal activation of gene expression in G1. We propose that prophase cohesin removal is a key step in reprogramming gene expression as cells transition from G2 through mitosis to G1.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Regulação da Expressão Gênica , Mitose/genética , Transcrição Gênica , Anáfase/genética , Animais , Aurora Quinase B/análise , Ciclo Celular , Proteínas de Ciclo Celular/análise , Linhagem Celular , Centrômero/enzimologia , Segregação de Cromossomos , Fase G1/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Humanos , Metáfase/genética , Prófase , RNA Polimerase II/metabolismo , Xenopus laevis , Coesinas
6.
Mol Cell ; 78(6): 1114-1132.e10, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32446320

RESUMO

Bromodomain-containing protein 4 (BRD4) is a cancer therapeutic target in ongoing clinical trials disrupting primarily BRD4-regulated transcription programs. The role of BRD4 in cancer has been attributed mainly to the abundant long isoform (BRD4-L). Here we show, by isoform-specific knockdown and endogenous protein detection, along with transgene expression, the less abundant BRD4 short isoform (BRD4-S) is oncogenic while BRD4-L is tumor-suppressive in breast cancer cell proliferation and migration, as well as mammary tumor formation and metastasis. Through integrated RNA-seq, genome-wide ChIP-seq, and CUT&RUN association profiling, we identify the Engrailed-1 (EN1) homeobox transcription factor as a key BRD4-S coregulator, particularly in triple-negative breast cancer. BRD4-S and EN1 comodulate the extracellular matrix (ECM)-associated matrisome network, including type II cystatin gene cluster, mucin 5, and cathepsin loci, via enhancer regulation of cancer-associated genes and pathways. Our work highlights the importance of targeted therapies for the oncogenic, but not tumor-suppressive, activity of BRD4.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Genes Homeobox , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Invasividade Neoplásica , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , Transcrição Gênica/genética , Neoplasias de Mama Triplo Negativas/genética
7.
Mol Cell ; 74(5): 1053-1068.e8, 2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31003867

RESUMO

Double-strand breaks (DSBs) initiate the homologous recombination that is crucial for meiotic chromosome pairing and segregation. Here, we unveil mouse ANKRD31 as a lynchpin governing multiple aspects of DSB formation. Spermatocytes lacking ANKRD31 have altered DSB locations and fail to target DSBs to the pseudoautosomal regions (PARs) of sex chromosomes. They also have delayed and/or fewer recombination sites but, paradoxically, more DSBs, suggesting DSB dysregulation. Unrepaired DSBs and pairing failures-stochastic on autosomes, nearly absolute on X and Y-cause meiotic arrest and sterility in males. Ankrd31-deficient females have reduced oocyte reserves. A crystal structure defines a pleckstrin homology (PH) domain in REC114 and its direct intermolecular contacts with ANKRD31. In vivo, ANKRD31 stabilizes REC114 association with the PAR and elsewhere. Our findings inform a model in which ANKRD31 is a scaffold anchoring REC114 and other factors to specific genomic locations, thereby regulating DSB formation.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Recombinação Homóloga/genética , Meiose/genética , Recombinases/química , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Pareamento Cromossômico , Segregação de Cromossomos/genética , Cromossomos , Cristalografia por Raios X , Quebras de DNA de Cadeia Dupla , Feminino , Masculino , Camundongos , Conformação Proteica , Recombinases/genética , Espermatócitos/química , Espermatócitos/metabolismo
8.
Annu Rev Cell Dev Biol ; 28: 59-87, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22578140

RESUMO

For unicellular organisms, the decision to enter the cell cycle can be viewed most fundamentally as a metabolic problem. A cell must assess its nutritional and metabolic status to ensure it can synthesize sufficient biomass to produce a new daughter cell. The cell must then direct the appropriate metabolic outputs to ensure completion of the division process. Herein, we discuss the changes in metabolism that accompany entry to, and exit from, the cell cycle for the unicellular eukaryote Saccharomyces cerevisiae. Studies of budding yeast under continuous, slow-growth conditions have provided insights into the essence of these metabolic changes at unprecedented temporal resolution. Some of these mechanisms by which cell growth and proliferation are coordinated with metabolism are likely to be conserved in multicellular organisms. An improved understanding of the metabolic basis of cell cycle control promises to reveal fundamental principles governing tumorigenesis, metazoan development, niche expansion, and many additional aspects of cell and organismal growth control.


Assuntos
Ciclo Celular , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Metabolismo Energético , Genes Fúngicos , Redes e Vias Metabólicas , Mitose , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/fisiologia , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia
9.
Mol Cell ; 72(4): 650-660.e8, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30392930

RESUMO

DNA replication is initiated by assembly of the kinase cell division cycle 7 (CDC7) with its regulatory activation subunit, activator of S-phase kinase (ASK), to activate DNA helicase. However, the mechanism underlying regulation of CDC7-ASK complex is unclear. Here, we show that ADP generated from CDC7-mediated MCM phosphorylation binds to an allosteric region of CDC7, disrupts CDC7-ASK interaction, and inhibits CDC7-ASK activity in a feedback way. EGFR- and ERK-activated casein kinase 2α (CK2α) phosphorylates nuclear phosphoglycerate kinase (PGK) 1 at S256, resulting in interaction of PGK1 with CDC7. CDC7-bound PGK1 converts ADP to ATP, thereby abrogating the inhibitory effect of ADP on CDC7-ASK activity, promoting the recruitment of DNA helicase to replication origins, DNA replication, cell proliferation, and brain tumorigenesis. These findings reveal an instrumental self-regulatory mechanism of CDC7-ASK activity by its kinase reaction product ADP and a nonglycolytic role for PGK1 in abrogating this negative feedback in promoting tumor development.


Assuntos
Difosfato de Adenosina/metabolismo , Caseína Quinase II/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Replicação do DNA , Fosfoglicerato Quinase/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Caseína Quinase II/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , DNA Helicases/genética , DNA Helicases/metabolismo , Feminino , Xenoenxertos , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfoglicerato Quinase/genética , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Origem de Replicação
10.
Nat Rev Mol Cell Biol ; 14(4): 197-210, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23847781

RESUMO

The protein kinase ataxia-telangiectasia mutated (ATM) is best known for its role as an apical activator of the DNA damage response in the face of DNA double-strand breaks (DSBs). Following induction of DSBs, ATM mobilizes one of the most extensive signalling networks that responds to specific stimuli and modifies directly or indirectly a broad range of targets. Although most ATM research has focused on this function, evidence suggests that ATM-mediated phosphorylation has a role in the response to other types of genotoxic stress. Moreover, it has become apparent that ATM is active in other cell signalling pathways involved in maintaining cellular homeostasis.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Dano ao DNA , Proteínas de Ligação a DNA/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Núcleo Celular/enzimologia , Homeostase , Humanos , Neurônios/enzimologia , Oxirredução , Mapas de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Vasos Retinianos/enzimologia , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
11.
Nat Rev Mol Cell Biol ; 14(9): 592-9, 2013 09.
Artigo em Inglês | MEDLINE | ID: mdl-23942451

RESUMO

Membrane trafficking and mitosis are two essential processes in eukaryotic cells. Surprisingly, many proteins best known for their role in membrane trafficking have additional 'moonlighting' functions in mitosis. Despite having proteins in common, there is insufficient evidence for a specific connection between these two processes. Instead, these phenomena demonstrate the adaptability of the membrane trafficking machinery that allows its repurposing for different cellular functions.


Assuntos
Membrana Celular/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Membrana/fisiologia , Mitose/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Centrossomo/metabolismo , Clatrina/metabolismo , Clatrina/fisiologia , Endocitose/fisiologia , Humanos , Cinetocoros/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/metabolismo , Modelos Biológicos , Ligação Proteica , Transporte Proteico/fisiologia
12.
PLoS Biol ; 19(1): e3001067, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33406066

RESUMO

To ensure genome stability, sexually reproducing organisms require that mating brings together exactly 2 haploid gametes and that meiosis occurs only in diploid zygotes. In the fission yeast Schizosaccharomyces pombe, fertilization triggers the Mei3-Pat1-Mei2 signaling cascade, which represses subsequent mating and initiates meiosis. Here, we establish a degron system to specifically degrade proteins postfusion and demonstrate that mating blocks not only safeguard zygote ploidy but also prevent lysis caused by aberrant fusion attempts. Using long-term imaging and flow-cytometry approaches, we identify previously unrecognized and independent roles for Mei3 and Mei2 in zygotes. We show that Mei3 promotes premeiotic S-phase independently of Mei2 and that cell cycle progression is both necessary and sufficient to reduce zygotic mating behaviors. Mei2 not only imposes the meiotic program and promotes the meiotic cycle, but also blocks mating behaviors independently of Mei3 and cell cycle progression. Thus, we find that fungi preserve zygote ploidy and survival by at least 2 mechanisms where the zygotic fate imposed by Mei2 and the cell cycle reentry triggered by Mei3 synergize to prevent zygotic mating.


Assuntos
Ciclo Celular/fisiologia , Fator de Acasalamento/fisiologia , Meiose/fisiologia , Zigoto/fisiologia , Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Proteínas Fúngicas/fisiologia , Genes Fúngicos/fisiologia , Fator de Acasalamento/genética , Fator de Acasalamento/metabolismo , Meiose/genética , Organismos Geneticamente Modificados , Ploidias , Proteínas de Ligação a RNA/fisiologia , Recombinação Genética/fisiologia , Schizosaccharomyces/fisiologia , Proteínas de Schizosaccharomyces pombe/fisiologia , Zigoto/crescimento & desenvolvimento , Zigoto/metabolismo
13.
Nat Rev Mol Cell Biol ; 13(12): 789-803, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23175282

RESUMO

Successful cell division requires the precise and timely coordination of chromosomal, cytoskeletal and membrane trafficking events. These processes are regulated by the competing actions of protein kinases and phosphatases. Aurora B is one of the most intensively studied kinases. In conjunction with inner centromere protein (INCENP), borealin (also known as Dasra) and survivin it forms the chromosomal passenger complex (CPC). This complex targets to different locations at differing times during mitosis, where it regulates key mitotic events: correction of chromosome-microtubule attachment errors; activation of the spindle assembly checkpoint; and construction and regulation of the contractile apparatus that drives cytokinesis. Our growing understanding of the CPC has seen it develop from a mere passenger riding on the chromosomes to one of the main controllers of mitosis.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Mitose , Animais , Aurora Quinase B , Aurora Quinases , Proteínas de Ciclo Celular/fisiologia , Centrômero/fisiologia , Drosophila melanogaster/fisiologia , Humanos , Camundongos , Microtúbulos/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Fuso Acromático/fisiologia
14.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34039707

RESUMO

Specified intestinal epithelial cells reprogram and contribute to the regeneration and renewal of the epithelium upon injury. Mutations that deregulate such renewal processes may contribute to tumorigenesis. Using intestinal organoids, we show that concomitant activation of Notch signaling and ablation of p53 induce a highly proliferative and regenerative cell state, which is associated with increased levels of Yap and the histone methyltransferase Mll1. The induced signaling system orchestrates high proliferation, self-renewal, and niche-factor-independent growth, and elevates the trimethylation of histone 3 at lysine 4 (H3K4me3). We demonstrate that Yap and Mll1 are also elevated in patient-derived colorectal cancer (CRC) organoids and control growth and viability. Our data suggest that Notch activation and p53 ablation induce a signaling circuitry involving Yap and the epigenetic regulator Mll1, which locks cells in a proliferative and regenerative state that renders them susceptible for tumorigenesis.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Histona-Lisina N-Metiltransferase/fisiologia , Proteína de Leucina Linfoide-Mieloide/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p53/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Humanos , Mutação , Organoides/metabolismo , Fatores de Transcrição/metabolismo
15.
PLoS Genet ; 17(6): e1009127, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34138874

RESUMO

During meiosis I, ring-shaped cohesin complexes play important roles in aiding the proper segregation of homologous chromosomes. RAD21L is a meiosis-specific vertebrate cohesin that is required for spermatogenesis in mice but is dispensable for oogenesis in young animals. The role of this cohesin in other vertebrate models has not been explored. Here, we tested if the zebrafish homolog Rad21l1 is required for meiotic chromosome dynamics during spermatogenesis and oogenesis. We found that Rad21l1 localizes to unsynapsed chromosome axes. It is also found between the axes of the mature tripartite synaptonemal complex (SC) in both sexes. We knocked out rad21l1 and found that nearly all rad21l1-/- mutants develop as fertile males, suggesting that the mutation causes a defect in juvenile oogenesis, since insufficient oocyte production triggers female to male sex reversal in zebrafish. Sex reversal was partially suppressed by mutation of the checkpoint gene tp53, suggesting that the rad21l1 mutation activates Tp53-mediated apoptosis or arrest in females. This response, however, is not linked to a defect in repairing Spo11-induced double-strand breaks since deletion of spo11 does not suppress the sex reversal phenotype. Compared to tp53 single mutant controls, rad21l1-/- tp53-/- double mutant females produce poor quality eggs that often die or develop into malformed embryos. Overall, these results indicate that the absence of rad21l1-/- females is due to a checkpoint-mediated response and highlight a role for a meiotic-specific cohesin subunit in oogenesis but not spermatogenesis.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Cromossômicas não Histona/fisiologia , Oogênese/fisiologia , Espermatogênese/fisiologia , Peixe-Zebra/genética , Animais , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Pareamento Cromossômico , Feminino , Genes p53 , Gônadas/anatomia & histologia , Masculino , Mutação , Peixe-Zebra/fisiologia , Coesinas
16.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33883277

RESUMO

The position of recombination events established along chromosomes in early prophase I and the chromosome remodeling that takes place in late prophase I are intrinsically linked steps of meiosis that need to be tightly regulated to ensure accurate chromosome segregation and haploid gamete formation. Here, we show that RAD-51 foci, which form at the sites of programmed meiotic DNA double-strand breaks (DSBs), exhibit a biased distribution toward off-centered positions along the chromosomes in wild-type Caenorhabditis elegans, and we identify two meiotic roles for chromatin-associated protein HIM-17 that ensure normal chromosome remodeling in late prophase I. During early prophase I, HIM-17 regulates the distribution of DSB-dependent RAD-51 foci and crossovers on chromosomes, which is critical for the formation of distinct chromosome subdomains (short and long arms of the bivalents) later during chromosome remodeling. During late prophase I, HIM-17 promotes the normal expression and localization of protein phosphatases GSP-1/2 to the surface of the bivalent chromosomes and may promote GSP-1 phosphorylation, thereby antagonizing Aurora B kinase AIR-2 loading on the long arms and preventing premature loss of sister chromatid cohesion. We propose that HIM-17 plays distinct roles at different stages during meiotic progression that converge to promote normal chromosome remodeling and accurate chromosome segregation.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Meiose/fisiologia , Recombinação Genética/fisiologia , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiologia , Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Segregação de Cromossomos/genética , Cromossomos/metabolismo , Troca Genética/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Rad51 Recombinase/metabolismo , Recombinação Genética/genética
17.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34504016

RESUMO

Expression and function of odorant receptors (ORs), which account for more than 50% of G protein-coupled receptors, are being increasingly reported in nonolfactory sites. However, ORs that can be targeted by drugs to treat diseases remain poorly identified. Tumor-derived lactate plays a crucial role in multiple signaling pathways leading to generation of tumor-associated macrophages (TAMs). In this study, we hypothesized that the macrophage OR Olfr78 functions as a lactate sensor and shapes the macrophage-tumor axis. Using Olfr78+/+ and Olfr78-/- bone marrow-derived macrophages with or without exogenous Olfr78 expression, we demonstrated that Olfr78 sensed tumor-derived lactate, which was the main factor in tumor-conditioned media responsible for generation of protumoral M2-TAMs. Olfr78 functioned together with Gpr132 to mediate lactate-induced generation of protumoral M2-TAMs. In addition, syngeneic Olfr78-deficient mice exhibited reduced tumor progression and metastasis together with an increased anti- versus protumoral immune cell population. We propose that the Olfr78-lactate interaction is a therapeutic target to reduce and prevent tumor progression and metastasis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Odorantes/metabolismo , Macrófagos Associados a Tumor/metabolismo , Animais , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Ácido Láctico/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Receptores Acoplados a Proteínas G/fisiologia , Receptores Odorantes/fisiologia , Transdução de Sinais , Microambiente Tumoral , Macrófagos Associados a Tumor/fisiologia
18.
PLoS Biol ; 18(11): e3000917, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33180788

RESUMO

The transition from mitosis into the first gap phase of the cell cycle in budding yeast is controlled by the Mitotic Exit Network (MEN). The network interprets spatiotemporal cues about the progression of mitosis and ensures that release of Cdc14 phosphatase occurs only after completion of key mitotic events. The MEN has been studied intensively; however, a unified understanding of how localisation and protein activity function together as a system is lacking. In this paper, we present a compartmental, logical model of the MEN that is capable of representing spatial aspects of regulation in parallel to control of enzymatic activity. We show that our model is capable of correctly predicting the phenotype of the majority of mutants we tested, including mutants that cause proteins to mislocalise. We use a continuous time implementation of the model to demonstrate that Cdc14 Early Anaphase Release (FEAR) ensures robust timing of anaphase, and we verify our findings in living cells. Furthermore, we show that our model can represent measured cell-cell variation in Spindle Position Checkpoint (SPoC) mutants. This work suggests a general approach to incorporate spatial effects into logical models. We anticipate that the model itself will be an important resource to experimental researchers, providing a rigorous platform to test hypotheses about regulation of mitotic exit.


Assuntos
Ciclo Celular/genética , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Divisão do Núcleo Celular/fisiologia , Pontos de Checagem da Fase M do Ciclo Celular/genética , Mitose/fisiologia , Fosforilação , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/fisiologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomycetales/genética , Saccharomycetales/metabolismo , Fuso Acromático/fisiologia
19.
PLoS Biol ; 18(1): e3000591, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31929526

RESUMO

A major challenge for cancer immunotherapy is sustaining T-cell activation and recruitment in immunosuppressive solid tumors. Here, we report that the levels of the Hippo pathway effector Yes-associated protein (Yap) are sharply induced upon the activation of cluster of differentiation 4 (CD4)-positive and cluster of differentiation 8 (CD8)-positive T cells and that Yap functions as an immunosuppressive factor and inhibitor of effector differentiation. Loss of Yap in T cells results in enhanced T-cell activation, differentiation, and function, which translates in vivo to an improved ability for T cells to infiltrate and repress tumors. Gene expression analyses of tumor-infiltrating T cells following Yap deletion implicates Yap as a mediator of global T-cell responses in the tumor microenvironment and as a negative regulator of T-cell tumor infiltration and patient survival in diverse human cancers. Collectively, our results indicate that Yap plays critical roles in T-cell biology and suggest that Yap inhibition improves T-cell responses in cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ciclo Celular/fisiologia , Quimiotaxia de Leucócito/genética , Linfócitos T/fisiologia , Microambiente Tumoral/imunologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Células Cultivadas , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Imunoterapia Adotiva , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Microambiente Tumoral/genética , Proteínas de Sinalização YAP
20.
PLoS Biol ; 18(12): e3000941, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33264286

RESUMO

Adverse cardiac remodeling after myocardial infarction (MI) causes structural and functional changes in the heart leading to heart failure. The initial post-MI pro-inflammatory response followed by reparative or anti-inflammatory response is essential for minimizing the myocardial damage, healing, and scar formation. Bone marrow-derived macrophages (BMDMs) are recruited to the injured myocardium and are essential for cardiac repair as they can adopt both pro-inflammatory or reparative phenotypes to modulate inflammatory and reparative responses, respectively. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are the key mediators of the Hippo signaling pathway and are essential for cardiac regeneration and repair. However, their functions in macrophage polarization and post-MI inflammation, remodeling, and healing are not well established. Here, we demonstrate that expression of YAP and TAZ is increased in macrophages undergoing pro-inflammatory or reparative phenotype changes. Genetic deletion of YAP/TAZ leads to impaired pro-inflammatory and enhanced reparative response. Consistently, YAP activation enhanced pro-inflammatory and impaired reparative response. We show that YAP/TAZ promote pro-inflammatory response by increasing interleukin 6 (IL6) expression and impede reparative response by decreasing Arginase-I (Arg1) expression through interaction with the histone deacetylase 3 (HDAC3)-nuclear receptor corepressor 1 (NCoR1) repressor complex. These changes in macrophages polarization due to YAP/TAZ deletion results in reduced fibrosis, hypertrophy, and increased angiogenesis, leading to improved cardiac function after MI. Also, YAP activation augmented MI-induced cardiac fibrosis and remodeling. In summary, we identify YAP/TAZ as important regulators of macrophage-mediated pro-inflammatory or reparative responses post-MI.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Macrófagos/metabolismo , Transativadores/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Variação Biológica da População/genética , Variação Biológica da População/fisiologia , Proteínas de Ciclo Celular/fisiologia , Feminino , Inflamação/metabolismo , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Fenótipo , Fosfoproteínas/metabolismo , Transdução de Sinais , Transativadores/fisiologia , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa