Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21.098
Filtrar
Mais filtros

Coleção SES
Eixos temáticos
Intervalo de ano de publicação
1.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34115982

RESUMO

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Assuntos
Citomegalovirus/fisiologia , Macrófagos Alveolares/virologia , Animais , Apresentação de Antígeno , Efeito Espectador , Ciclo Celular , Linhagem Celular Transformada , Reprogramação Celular , Citomegalovirus/patogenicidade , Citomegalovirus/ultraestrutura , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Proteínas de Fluorescência Verde/metabolismo , Pulmão/patologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/ultraestrutura , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fenótipo , Células-Tronco/patologia , Replicação Viral/fisiologia , Via de Sinalização Wnt
2.
Nat Immunol ; 23(1): 50-61, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34853448

RESUMO

NP105-113-B*07:02-specific CD8+ T cell responses are considered among the most dominant in SARS-CoV-2-infected individuals. We found strong association of this response with mild disease. Analysis of NP105-113-B*07:02-specific T cell clones and single-cell sequencing were performed concurrently, with functional avidity and antiviral efficacy assessed using an in vitro SARS-CoV-2 infection system, and were correlated with T cell receptor usage, transcriptome signature and disease severity (acute n = 77, convalescent n = 52). We demonstrated a beneficial association of NP105-113-B*07:02-specific T cells in COVID-19 disease progression, linked with expansion of T cell precursors, high functional avidity and antiviral effector function. Broad immune memory pools were narrowed postinfection but NP105-113-B*07:02-specific T cells were maintained 6 months after infection with preserved antiviral efficacy to the SARS-CoV-2 Victoria strain, as well as Alpha, Beta, Gamma and Delta variants. Our data show that NP105-113-B*07:02-specific T cell responses associate with mild disease and high antiviral efficacy, pointing to inclusion for future vaccine design.


Assuntos
Antígeno HLA-B7/imunologia , Epitopos Imunodominantes/imunologia , Proteínas do Nucleocapsídeo/imunologia , SARS-CoV-2/imunologia , Linfócitos T Citotóxicos/imunologia , Idoso , Sequência de Aminoácidos , Anticorpos Antivirais/imunologia , Afinidade de Anticorpos/imunologia , COVID-19/imunologia , COVID-19/patologia , Linhagem Celular Transformada , Feminino , Perfilação da Expressão Gênica , Humanos , Memória Imunológica/imunologia , Masculino , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T/imunologia , Índice de Gravidade de Doença , Vaccinia virus/genética , Vaccinia virus/imunologia , Vaccinia virus/metabolismo
3.
Cell ; 175(6): 1607-1619.e15, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30500539

RESUMO

In the healthy adult liver, most hepatocytes proliferate minimally. However, upon physical or chemical injury to the liver, hepatocytes proliferate extensively in vivo under the direction of multiple extracellular cues, including Wnt and pro-inflammatory signals. Currently, liver organoids can be generated readily in vitro from bile-duct epithelial cells, but not hepatocytes. Here, we show that TNFα, an injury-induced inflammatory cytokine, promotes the expansion of hepatocytes in 3D culture and enables serial passaging and long-term culture for more than 6 months. Single-cell RNA sequencing reveals broad expression of hepatocyte markers. Strikingly, in vitro-expanded hepatocytes engrafted, and significantly repopulated, the injured livers of Fah-/- mice. We anticipate that tissue repair signals can be harnessed to promote the expansion of otherwise hard-to-culture cell-types, with broad implications.


Assuntos
Antígenos de Diferenciação/biossíntese , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Hepatócitos/metabolismo , Esferoides Celulares/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Linhagem Celular Transformada , Células Hep G2 , Hepatócitos/transplante , Células Endoteliais da Veia Umbilical Humana , Humanos , Fígado/lesões , Fígado/metabolismo , Camundongos Knockout , Esferoides Celulares/transplante , Fatores de Tempo
4.
Cell ; 172(4): 731-743.e12, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29425491

RESUMO

The noncanonical IKK family member TANK-binding kinase 1 (TBK1) is activated by pro-inflammatory cytokines, but its role in controlling metabolism remains unclear. Here, we report that the kinase uniquely controls energy metabolism. Tbk1 expression is increased in adipocytes of HFD-fed mice. Adipocyte-specific TBK1 knockout (ATKO) attenuates HFD-induced obesity by increasing energy expenditure; further studies show that TBK1 directly inhibits AMPK to repress respiration and increase energy storage. Conversely, activation of AMPK under catabolic conditions can increase TBK1 activity through phosphorylation, mediated by AMPK's downstream target ULK1. Surprisingly, ATKO also exaggerates adipose tissue inflammation and insulin resistance. TBK1 suppresses inflammation by phosphorylating and inducing the degradation of the IKK kinase NIK, thus attenuating NF-κB activity. Moreover, TBK1 mediates the negative impact of AMPK activity on NF-κB activation. These data implicate a unique role for TBK1 in mediating bidirectional crosstalk between energy sensing and inflammatory signaling pathways in both over- and undernutrition.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Metabolismo Energético , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Linhagem Celular Transformada , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Serina-Treonina Quinases/genética , Quinase Induzida por NF-kappaB
5.
Mol Cell ; 74(1): 101-117.e10, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30827740

RESUMO

During X-inactivation, Xist RNA spreads along an entire chromosome to establish silencing. However, the mechanism and functional RNA elements involved in spreading remain undefined. By performing a comprehensive endogenous Xist deletion screen, we identify Repeat B as crucial for spreading Xist and maintaining Polycomb repressive complexes 1 and 2 (PRC1/PRC2) along the inactive X (Xi). Unexpectedly, spreading of these three factors is inextricably linked. Deleting Repeat B or its direct binding partner, HNRNPK, compromises recruitment of PRC1 and PRC2. In turn, ablating PRC1 or PRC2 impairs Xist spreading. Therefore, Xist and Polycomb complexes require each other to propagate along the Xi, suggesting a positive feedback mechanism between RNA initiator and protein effectors. Perturbing Xist/Polycomb spreading causes failure of de novo Xi silencing, with partial compensatory downregulation of the active X, and also disrupts topological Xi reconfiguration. Thus, Repeat B is a multifunctional element that integrates interdependent Xist/Polycomb spreading, silencing, and changes in chromosome architecture.


Assuntos
Fibroblastos/metabolismo , Deleção de Genes , Inativação Gênica , Células-Tronco Embrionárias Murinas/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 2/genética , RNA Longo não Codificante/genética , Inativação do Cromossomo X , Cromossomo X/genética , Animais , Linhagem Celular Transformada , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Ribonucleoproteínas Nucleares Heterogêneas Grupo K , Masculino , Camundongos , Motivos de Nucleotídeos , Complexo Repressor Polycomb 1/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Ligação Proteica , RNA Longo não Codificante/metabolismo , Sequências Repetitivas de Ácido Nucleico , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Cromossomo X/metabolismo
6.
Genes Dev ; 33(11-12): 684-704, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31048545

RESUMO

DNA double-strand breaks (DSBs) at RNA polymerase II (RNAPII) transcribed genes lead to inhibition of transcription. The DNA-dependent protein kinase (DNA-PK) complex plays a pivotal role in transcription inhibition at DSBs by stimulating proteasome-dependent eviction of RNAPII at these lesions. How DNA-PK triggers RNAPII eviction to inhibit transcription at DSBs remains unclear. Here we show that the HECT E3 ubiquitin ligase WWP2 associates with components of the DNA-PK and RNAPII complexes and is recruited to DSBs at RNAPII transcribed genes. In response to DSBs, WWP2 targets the RNAPII subunit RPB1 for K48-linked ubiquitylation, thereby driving DNA-PK- and proteasome-dependent eviction of RNAPII. The lack of WWP2 or expression of nonubiquitylatable RPB1 abrogates the binding of nonhomologous end joining (NHEJ) factors, including DNA-PK and XRCC4/DNA ligase IV, and impairs DSB repair. These findings suggest that WWP2 operates in a DNA-PK-dependent shutoff circuitry for RNAPII clearance that promotes DSB repair by protecting the NHEJ machinery from collision with the transcription machinery.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Proteína Quinase Ativada por DNA/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Proteínas Nucleares/metabolismo , RNA Polimerase II/metabolismo , Transcrição Gênica , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitinação
7.
Mol Cell ; 71(4): 581-591.e5, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-30057196

RESUMO

Biological signals need to be robust and filter small fluctuations yet maintain sensitivity to signals across a wide range of magnitudes. Here, we studied how fluctuations in DNA damage signaling relate to maintenance of long-term cell-cycle arrest. Using live-cell imaging, we quantified division profiles of individual human cells in the course of 1 week after irradiation. We found a subset of cells that initially establish cell-cycle arrest and then sporadically escape and divide. Using fluorescent reporters and mathematical modeling, we determined that fluctuations in the oscillatory pattern of the tumor suppressor p53 trigger a sharp switch between p21 and CDK2, leading to escape from arrest. Transient perturbation of p53 stability mimicked the noise in individual cells and was sufficient to trigger escape from arrest. Our results show that the self-reinforcing circuitry that mediates cell-cycle transitions can translate small fluctuations in p53 signaling into large phenotypic changes.


Assuntos
Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células Epiteliais/metabolismo , Modelos Estatísticos , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Divisão Celular/efeitos da radiação , Linhagem Celular Transformada , Proliferação de Células/efeitos da radiação , Quinase 2 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Dano ao DNA , Células Epiteliais/citologia , Células Epiteliais/efeitos da radiação , Raios gama , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Estabilidade Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/efeitos da radiação , Imagem com Lapso de Tempo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Proteína Vermelha Fluorescente
8.
EMBO J ; 40(21): e104543, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34533226

RESUMO

The DNA polymerase zeta (Polζ) plays a critical role in bypassing DNA damage. REV3L, the catalytic subunit of Polζ, is also essential in mouse embryonic development and cell proliferation for reasons that remain incompletely understood. In this study, we reveal that REV3L protein interacts with heterochromatin components including repressive histone marks and localizes in pericentromeric regions through direct interaction with HP1 dimer. We demonstrate that Polζ/REV3L ensures progression of replication forks through difficult-to-replicate pericentromeric heterochromatin, thereby preventing spontaneous chromosome break formation. We also find that Rev3l-deficient cells are compromised in the repair of heterochromatin-associated double-stranded breaks, eliciting deletions in late-replicating regions. Lack of REV3L leads to further consequences that may be ascribed to heterochromatin replication and repair-associated functions of Polζ, with a disruption of the temporal replication program at specific loci. This is correlated with changes in epigenetic landscape and transcriptional control of developmentally regulated genes. These results reveal a new function of Polζ in preventing chromosome instability during replication of heterochromatic regions.


Assuntos
Replicação do DNA , Proteínas de Ligação a DNA/genética , DNA Polimerase Dirigida por DNA/genética , DNA/genética , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Heterocromatina/metabolismo , Animais , Linhagem Celular , Linhagem Celular Transformada , Proliferação de Células , Homólogo 5 da Proteína Cromobox/genética , Homólogo 5 da Proteína Cromobox/metabolismo , Instabilidade Cromossômica , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Embrião de Mamíferos , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células HeLa , Heterocromatina/química , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Transdução de Sinais
9.
Mol Cell ; 68(4): 659-672.e9, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29149594

RESUMO

Certain BH3-only proteins transiently bind and activate Bak and Bax, initiating their oligomerization and the permeabilization of the mitochondrial outer membrane, a pivotal step in the mitochondrial pathway to apoptosis. Here we describe the first crystal structures of an activator BH3 peptide bound to Bak and illustrate their use in the design of BH3 derivatives capable of inhibiting human Bak on mitochondria. These BH3 derivatives compete for the activation site at the canonical groove, are the first engineered inhibitors of Bak activation, and support the role of key conformational transitions associated with Bak activation.


Assuntos
Apoptose/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2 , Mitocôndrias , Peptídeos , Proteína Killer-Antagonista Homóloga a bcl-2 , Animais , Proteína 11 Semelhante a Bcl-2/química , Proteína 11 Semelhante a Bcl-2/farmacologia , Linhagem Celular Transformada , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Ligação Proteica , Relação Estrutura-Atividade , Proteína Killer-Antagonista Homóloga a bcl-2/química , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
10.
Mol Cell ; 68(1): 61-75.e5, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28943310

RESUMO

Double-strand breaks (DSBs) of DNA in eukaryotic cells are predominantly repaired by non-homologous end joining (NHEJ). The histone chaperone anti-silencing factor 1a (ASF1a) interacts with MDC1 and is recruited to sites of DSBs to facilitate the interaction of phospho-ATM with MDC1 and phosphorylation of MDC1, which are required for the recruitment of RNF8/RNF168 histone ubiquitin ligases. Thus, ASF1a deficiency reduces histone ubiquitination at DSBs, decreasing the recruitment of 53BP1, and decreases NHEJ, rendering cells more sensitive to DSBs. This role of ASF1a in DSB repair cannot be provided by the closely related ASF1b and does not require its histone chaperone activity. Homozygous deletion of ASF1A is seen in 10%-15% of certain cancers, suggesting that loss of NHEJ may be selected in some malignancies and that the deletion can be used as a molecular biomarker for cancers susceptible to radiotherapy or to DSB-inducing chemotherapy.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas de Ciclo Celular/genética , Reparo do DNA por Junção de Extremidades , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Nucleares/genética , Transativadores/genética , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Cromatina/química , Cromatina/metabolismo , DNA/genética , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Células HEK293 , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Fosforilação , Transdução de Sinais , Transativadores/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
11.
Proc Natl Acad Sci U S A ; 119(11): e2118930119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35254896

RESUMO

SignificanceHigh-risk (HR) human papillomaviruses (HPV) from the genus alpha cause anogenital and oropharyngeal cancers, whereas the contribution of HPV from the genus beta to the development of cutaneous squamous cell cancer is still under debate. HR-HPV genomes display potent immortalizing activity in human keratinocytes, the natural target cell for HPV. This paper shows that immortalization of keratinocytes by the beta-HPV49 genome requires the inactivation of the viral E8^E2 repressor protein and the presence of the E6 and E7 oncoproteins but also of the E1 and E2 replication proteins. This reveals important differences in the carcinogenic properties of HR-HPV and beta-HPV but also warrants further investigations on the distribution and mutation frequencies of beta-HPV in human cancers.


Assuntos
Betapapillomavirus/fisiologia , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Queratinócitos/virologia , Infecções por Papillomavirus/virologia , Replicação Viral , Linhagem Celular Transformada , Genoma Viral , Humanos , Queratinócitos/metabolismo , Proteínas Oncogênicas Virais/genética , RNA Viral
12.
EMBO J ; 39(11): e101573, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32323871

RESUMO

High expression of 2',5'-oligoadenylate synthetase 1 (OAS1), which adds AMP residues in 2',5' linkage to a variety of substrates, is observed in many cancers as a part of the interferon-related DNA damage resistance signature (IRDS). Poly(ADP-ribose) (PAR) is rapidly synthesized from NAD+ at sites of DNA damage to facilitate repair, but excessive PAR synthesis due to extensive DNA damage results in cell death by energy depletion and/or activation of PAR-dependent programmed cell death pathways. We find that OAS1 adds AMP residues in 2',5' linkage to PAR, inhibiting its synthesis in vitro and reducing its accumulation in cells. Increased OAS1 expression substantially improves cell viability following DNA-damaging treatments that stimulate PAR synthesis during DNA repair. We conclude that high expression of OAS1 in cancer cells promotes their ability to survive DNA damage by attenuating PAR synthesis and thus preventing cell death.


Assuntos
2',5'-Oligoadenilato Sintetase/biossíntese , Dano ao DNA , Regulação Enzimológica da Expressão Gênica , Poli ADP Ribosilação , 2',5'-Oligoadenilato Sintetase/genética , Monofosfato de Adenosina/genética , Monofosfato de Adenosina/metabolismo , Morte Celular , Linhagem Celular Transformada , Humanos
13.
Development ; 148(16)2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-34338291

RESUMO

Negative feedback loops represent a regulatory mechanism that guarantees that signaling thresholds are compatible with a physiological response. Previously, we established that Lrig1 acts through this mechanism to inhibit Ret activity. However, it is unclear whether other Lrig family members play similar roles. Here, we show that Lrig1 and Lrig3 are co-expressed in Ret-positive mouse dorsal root ganglion (DRG) neurons. Lrig3, like Lrig1, interacts with Ret and inhibits GDNF/Ret signaling. Treatment of DRG neurons with GDNF ligands induces a significant increase in the expression of Lrig1 and Lrig3. Our findings show that, whereas a single deletion of either Lrig1 or Lrig3 fails to promote Ret-mediated axonal growth, haploinsufficiency of Lrig1 in Lrig3 mutants significantly potentiates Ret signaling and axonal growth of DRG neurons in response to GDNF ligands. We observe that Lrig1 and Lrig3 act redundantly to ensure proper cutaneous innervation of nonpeptidergic axons and behavioral sensitivity to cold, which correlates with a significant increase in the expression of the cold-responsive channel TrpA1. Together, our findings provide insights into the in vivo functions through which Lrig genes control morphology, connectivity and function in sensory neurons.


Assuntos
Axônios/metabolismo , Epiderme/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais/genética , Animais , Animais Recém-Nascidos , Linhagem Celular Transformada , Gânglios Espinais/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Células HEK293 , Humanos , Ligantes , Masculino , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Neurônios Motores/metabolismo , Proteínas do Tecido Nervoso/genética , Crescimento Neuronal/genética , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transfecção
14.
PLoS Biol ; 19(2): e3001097, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33596193

RESUMO

The oncogenic human herpesviruses Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) are the causative agents of multiple malignancies. A hallmark of herpesviruses is their biphasic life cycle consisting of latent and lytic infection. In this study, we identified that cellular nonsense-mediated decay (NMD), an evolutionarily conserved RNA degradation pathway, critically regulates the latent-to-lytic switch of EBV and KSHV infection. The NMD machinery suppresses EBV and KSHV Rta transactivator expression and promotes maintenance of viral latency by targeting the viral polycistronic transactivator transcripts for degradation through the recognition of features in their 3' UTRs. Treatment with a small-molecule NMD inhibitor potently induced reactivation in a variety of EBV- and KSHV-infected cell types. In conclusion, our results identify NMD as an important host process that controls oncogenic herpesvirus reactivation, which may be targeted for the therapeutic induction of lytic reactivation and the eradication of tumor cells.


Assuntos
Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 8/fisiologia , Degradação do RNAm Mediada por Códon sem Sentido , Transativadores/fisiologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Infecções por Vírus Epstein-Barr/virologia , Regulação Viral da Expressão Gênica , Células HEK293 , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Humanos , RNA Viral , Sarcoma de Kaposi/virologia , Transativadores/genética , Latência Viral/genética
15.
Cell ; 139(4): 693-706, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19878981

RESUMO

Inflammation is linked clinically and epidemiologically to cancer, and NF-kappaB appears to play a causative role, but the mechanisms are poorly understood. We show that transient activation of Src oncoprotein can mediate an epigenetic switch from immortalized breast cells to a stably transformed line that forms self-renewing mammospheres that contain cancer stem cells. Src activation triggers an inflammatory response mediated by NF-kappaB that directly activates Lin28 transcription and rapidly reduces let-7 microRNA levels. Let-7 directly inhibits IL6 expression, resulting in higher levels of IL6 than achieved by NF-kappaB activation. IL6-mediated activation of the STAT3 transcription factor is necessary for transformation, and IL6 activates NF-kappaB, thereby completing a positive feedback loop. This regulatory circuit operates in other cancer cells lines, and its transcriptional signature is found in human cancer tissues. Thus, inflammation activates a positive feedback loop that maintains the epigenetic transformed state for many generations in the absence of the inducing signal.


Assuntos
Epigênese Genética , Inflamação/metabolismo , Interleucina-6/metabolismo , MicroRNAs/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Linhagem Celular , Linhagem Celular Transformada , Retroalimentação Fisiológica , Genes src , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo
16.
Mol Cell ; 63(4): 662-673, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27453047

RESUMO

DNA polymerase theta (Pol θ)-mediated end joining (TMEJ) has been implicated in the repair of chromosome breaks, but its cellular mechanism and role relative to canonical repair pathways are poorly understood. We show that it accounts for most repairs associated with microhomologies and is made efficient by coupling a microhomology search to removal of non-homologous tails and microhomology-primed synthesis across broken ends. In contrast to non-homologous end joining (NHEJ), TMEJ efficiently repairs end structures expected after aborted homology-directed repair (5' to 3' resected ends) or replication fork collapse. It typically does not compete with canonical repair pathways but, in NHEJ-deficient cells, is engaged more frequently and protects against translocation. Cell viability is also severely impaired upon combined deficiency in Pol θ and a factor that antagonizes end resection (Ku or 53BP1). TMEJ thus helps to sustain cell viability and genome stability by rescuing chromosome break repair when resection is misregulated or NHEJ is compromised.


Assuntos
Quebra Cromossômica , Reparo do DNA por Junção de Extremidades , DNA Polimerase Dirigida por DNA/metabolismo , Instabilidade Genômica , Animais , Sistemas CRISPR-Cas , Linhagem Celular Transformada , DNA Polimerase Dirigida por DNA/deficiência , DNA Polimerase Dirigida por DNA/genética , Genótipo , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Camundongos Knockout , Fenótipo , Fatores de Tempo , DNA Polimerase teta
17.
Mol Cell ; 64(4): 790-802, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27840028

RESUMO

Recent studies have revealed the importance of Ki-67 and the chromosome periphery in chromosome structure and segregation, but little is known about this elusive chromosome compartment. Here we used correlative light and serial block-face scanning electron microscopy, which we term 3D-CLEM, to model the entire mitotic chromosome complement at ultra-structural resolution. Prophase chromosomes exhibit a highly irregular surface appearance with a volume smaller than metaphase chromosomes. This may be because of the absence of the periphery, which associates with chromosomes only after nucleolar disassembly later in prophase. Indeed, the nucleolar volume almost entirely accounts for the extra volume found in metaphase chromosomes. Analysis of wild-type and Ki-67-depleted chromosomes reveals that the periphery comprises 30%-47% of the entire chromosome volume and more than 33% of the protein mass of isolated mitotic chromosomes determined by quantitative proteomics. Thus, chromatin makes up a surprisingly small percentage of the total mass of metaphase chromosomes.


Assuntos
Cromatina/ultraestrutura , Cromossomos/ultraestrutura , Metáfase , Microscopia Eletrônica de Varredura/métodos , Prófase , Linhagem Celular Transformada , Nucléolo Celular/química , Nucléolo Celular/ultraestrutura , Cromatina/química , Cromossomos/química , Expressão Gênica , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Epitélio Pigmentado da Retina/química , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/ultraestrutura
18.
Mol Cell ; 64(2): 388-404, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27768874

RESUMO

Common fragile sites (CFSs) are genomic regions that are unstable under conditions of replicative stress. Although the characteristics of CFSs that render them vulnerable to stress are associated mainly with replication, the cellular pathways that protect CFSs during replication remain unclear. Here, we identify and describe a role for FANCD2 as a trans-acting facilitator of CFS replication, in the absence of exogenous replicative stress. In the absence of FANCD2, replication forks stall within the AT-rich fragility core of CFS, leading to dormant origin activation. Furthermore, FANCD2 deficiency is associated with DNA:RNA hybrid formation at CFS-FRA16D, and inhibition of DNA:RNA hybrid formation suppresses replication perturbation. In addition, we also found that FANCD2 reduces the number of potential sites of replication initiation. Our data demonstrate that FANCD2 protein is required to ensure efficient CFS replication and provide mechanistic insight into how FANCD2 regulates CFS stability.


Assuntos
Sítios Frágeis do Cromossomo , Replicação do DNA , DNA/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , RNA/genética , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Linhagem Celular Transformada , DNA/metabolismo , Anemia de Fanconi , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Instabilidade Genômica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , RNA/metabolismo
19.
Mol Cell ; 62(4): 479-90, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27184077

RESUMO

Recurrent mutations in the splicing factor U2AF35 are found in several cancers and myelodysplastic syndrome (MDS). How oncogenic U2AF35 mutants promote transformation remains to be determined. Here we derive cell lines transformed by the oncogenic U2AF35(S34F) mutant and identify aberrantly processed pre-mRNAs by deep sequencing. We find that in U2AF35(S34F)-transformed cells the autophagy-related factor 7 (Atg7) pre-mRNA is abnormally processed, which unexpectedly is not due to altered splicing but rather selection of a distal cleavage and polyadenylation (CP) site. This longer Atg7 mRNA is translated inefficiently, leading to decreased ATG7 levels and an autophagy defect that predisposes cells to secondary mutations, resulting in transformation. MDS and acute myeloid leukemia patient samples harboring U2AF35(S34F) have a similar increased use of the ATG7 distal CP site, and previous studies have shown that mice with hematopoietic cells lacking Atg7 develop an MDS-like syndrome. Collectively, our results reveal a basis for U2AF35(S34F) oncogenic activity.


Assuntos
Proteína 7 Relacionada à Autofagia/genética , Transformação Celular Neoplásica/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas/genética , Processamento de Terminações 3' de RNA , Precursores de RNA/genética , RNA Mensageiro/genética , Fator de Processamento U2AF/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Autofagia , Proteína 7 Relacionada à Autofagia/metabolismo , Linhagem Celular Transformada , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mutação , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/patologia , Poliadenilação , Interferência de RNA , Precursores de RNA/metabolismo , RNA Mensageiro/metabolismo , Fator de Processamento U2AF/metabolismo , Fatores de Tempo , Transfecção , Carga Tumoral
20.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34088848

RESUMO

Homeothermic vertebrates produce heat in cold environments through thermogenesis, in which brown adipose tissue (BAT) increases mitochondrial oxidation along with uncoupling of the electron transport chain and activation of uncoupling protein 1 (UCP1). Although the transcription factors regulating the expression of UCP1 and nutrient oxidation genes have been extensively studied, only a few other proteins essential for BAT function have been identified. We describe the discovery of FAM195A, a BAT-enriched RNA binding protein, which is required for cold-dependent thermogenesis in mice. FAM195A knockout (KO) mice display whitening of BAT and an inability to thermoregulate. In BAT of FAM195A KO mice, enzymes involved in branched-chain amino acid (BCAA) metabolism are down-regulated, impairing their response to cold. Knockdown of FAM195A in brown adipocytes in vitro also impairs expression of leucine oxidation enzymes, revealing FAM195A to be a regulator of BCAA metabolism and a potential target for metabolic disorders.


Assuntos
Adipócitos Marrons , Tecido Adiposo Marrom , Temperatura Baixa , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Termogênese , Aminoácidos de Cadeia Ramificada/genética , Aminoácidos de Cadeia Ramificada/metabolismo , Animais , Linhagem Celular Transformada , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa