Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Curr Opin Biotechnol ; 85: 103060, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38194753

RESUMO

Cellular dynamics, the transition of a cell from one state to another, is central to understanding developmental processes and disease progression. Single-cell transcriptomics has been pushing the frontiers of cellular dynamics studies into a genome-wide and single-cell level. While most single-cell RNA sequencing approaches are disruptive and only provide a snapshot of cell states, the dynamics of a cell could be reconstructed by either exploiting temporal information hiding in the transcriptomics data or integrating additional information. In this review, we describe these approaches, highlighting their underlying principles, key assumptions, and the rationality to interpret the results as models. We also discuss the recently emerging nondisruptive live-cell transcriptomics methods, which are highly complementary to the computational models for their assumption-free nature.


Assuntos
Análise de Célula Única , Transcriptoma , Transcriptoma/genética , Análise de Célula Única/métodos , Perfilação da Expressão Gênica
2.
Nat Commun ; 13(1): 7227, 2022 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-36433946

RESUMO

Gut-draining mesenteric lymph nodes (LN) provide the framework to shape intestinal adaptive immune responses. Based on the transcriptional signatures established by our previous work, the composition and immunomodulatory function of LN stromal cells (SC) vary according to location. Here, we describe the single-cell composition and development of the SC compartment within mesenteric LNs derived from postnatal to aged mice. We identify CD34+ SC and fibroblastic reticular stromal cell (FRC) progenitors as putative progenitors, both supplying the typical rapid postnatal mesenteric LN expansion. We further establish the location-specific chromatin accessibility and DNA methylation landscape of non-endothelial SCs and identify a microbiota-independent core epigenomic signature, showing characteristic differences between SCs from mesenteric and skin-draining peripheral LNs. The epigenomic landscape of SCs points to dynamic expression of Irf3 along the differentiation trajectories of FRCs. Accordingly, a mesenchymal stem cell line acquires a Cxcl9+ FRC molecular phenotype upon lentiviral overexpression of Irf3, and the relevance of Irf3 for SC biology is further underscored by the diminished proportion of Ccl19+ and Cxcl9+ FRCs in LNs of Irf3-/- mice. Together, our data constitute a comprehensive transcriptional and epigenomic map of mesenteric LNSC development in early life and dissect location-specific, microbiota-independent properties of non-endothelial SCs.


Assuntos
Linfonodos , Células Estromais , Camundongos , Animais , Camundongos Endogâmicos C57BL , Células Estromais/metabolismo , Linfonodos/patologia , Moléculas de Adesão Celular/metabolismo , Antígenos CD34/metabolismo
3.
Nature ; 608(7924): 733-740, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35978187

RESUMO

Single-cell transcriptomics (scRNA-seq) has greatly advanced our ability to characterize cellular heterogeneity1. However, scRNA-seq requires lysing cells, which impedes further molecular or functional analyses on the same cells. Here, we established Live-seq, a single-cell transcriptome profiling approach that preserves cell viability during RNA extraction using fluidic force microscopy2,3, thus allowing to couple a cell's ground-state transcriptome to its downstream molecular or phenotypic behaviour. To benchmark Live-seq, we used cell growth, functional responses and whole-cell transcriptome read-outs to demonstrate that Live-seq can accurately stratify diverse cell types and states without inducing major cellular perturbations. As a proof of concept, we show that Live-seq can be used to directly map a cell's trajectory by sequentially profiling the transcriptomes of individual macrophages before and after lipopolysaccharide (LPS) stimulation, and of adipose stromal cells pre- and post-differentiation. In addition, we demonstrate that Live-seq can function as a transcriptomic recorder by preregistering the transcriptomes of individual macrophages that were subsequently monitored by time-lapse imaging after LPS exposure. This enabled the unsupervised, genome-wide ranking of genes on the basis of their ability to affect macrophage LPS response heterogeneity, revealing basal Nfkbia expression level and cell cycle state as important phenotypic determinants, which we experimentally validated. Thus, Live-seq can address a broad range of biological questions by transforming scRNA-seq from an end-point to a temporal analysis approach.


Assuntos
Sobrevivência Celular , Perfilação da Expressão Gênica , Macrófagos , RNA-Seq , Análise de Célula Única , Transcriptoma , Tecido Adiposo/citologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular , Perfilação da Expressão Gênica/métodos , Perfilação da Expressão Gênica/normas , Genoma/efeitos dos fármacos , Genoma/genética , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Inibidor de NF-kappaB alfa/genética , Especificidade de Órgãos , Fenótipo , RNA/genética , RNA/isolamento & purificação , RNA-Seq/métodos , RNA-Seq/normas , Reprodutibilidade dos Testes , Análise de Sequência de RNA/métodos , Análise de Sequência de RNA/normas , Análise de Célula Única/métodos , Células Estromais/citologia , Células Estromais/metabolismo , Fatores de Tempo , Transcriptoma/genética
4.
Nat Commun ; 10(1): 1809, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-31000713

RESUMO

Krüppel-associated box zinc finger proteins (KZFPs) constitute the largest family of mammalian transcription factors, but most remain completely uncharacterized. While initially proposed to primarily repress transposable elements, recent reports have revealed that KFZPs contribute to a wide variety of other biological processes. Using murine and human in vitro and in vivo models, we demonstrate here that one poorly studied KZFP, ZFP30, promotes adipogenesis by directly targeting and activating a retrotransposon-derived Pparg2 enhancer. Through mechanistic studies, we further show that ZFP30 recruits the co-regulator KRAB-associated protein 1 (KAP1), which, surprisingly, acts as a ZFP30 co-activator in this adipogenic context. Our findings provide an understanding of both adipogenic and KZFP-KAP1 complex-mediated gene regulation, showing that the KZFP-KAP1 axis can also function in a non-repressive manner.


Assuntos
Adipogenia/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Dedos de Zinco/fisiologia , Células 3T3 , Adipócitos/fisiologia , Animais , Biologia Computacional , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Feminino , Regulação da Expressão Gênica/fisiologia , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , PPAR gama/genética , Regiões Promotoras Genéticas/genética , Retroelementos/genética , Fatores de Transcrição/genética
5.
Cell Metab ; 28(2): 282-288.e3, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-29909970

RESUMO

Adipose tissue in the mammary gland undergoes dramatic remodeling during reproduction. Adipocytes are replaced by mammary alveolar structures during pregnancy and lactation, then reappear upon weaning. The fate of the original adipocytes during lactation and the developmental origin of the re-appearing adipocyte post involution are unclear. Here, we reveal that adipocytes in the mammary gland de-differentiate into Pdgfrα+ preadipocyte- and fibroblast-like cells during pregnancy and remain de-differentiated during lactation. Upon weaning, de-differentiated fibroblasts proliferate and re-differentiate into adipocytes. This cycle occurs over multiple pregnancies. These observations reveal the potential of terminally differentiated adipocytes to undergo repeated cycles of de-differentiation and re-differentiation in a physiological setting.


Assuntos
Adipócitos Brancos/metabolismo , Adipogenia , Tecido Adiposo , Lactação/metabolismo , Glândulas Mamárias Animais , Adipócitos Brancos/citologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Feminino , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Desmame
6.
Mol Cell Biol ; 38(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061731

RESUMO

The c-Jun gene encodes a transcription factor that has been implicated in many physiological and pathological processes. c-Jun is a highly unstable protein that is degraded through a ubiquitination/proteasome-dependent mechanism. However, the deubiquitinating enzyme (DUB) that regulates the stability of the c-Jun protein requires further investigation. Here, by screening a DUB expression library, we identified ubiquitin-specific protease 6 (USP6) and showed that it regulates the stability of the c-Jun protein in a manner depending on its enzyme activity. USP6 interacts with c-Jun and antagonizes its ubiquitination. USP6 overexpression upregulates the activity of the downstream signaling pathway mediated by c-Jun/AP-1 and promotes cell invasion. Moreover, many aberrant genes that are upregulated in USP6 translocated nodular fasciitis are great potential targets regulated by c-Jun. Based on our data, USP6 is an enzyme that deubiquitinates c-Jun and regulates its downstream cellular functions.


Assuntos
Fasciite/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina Tiolesterase/metabolismo , Movimento Celular , Feminino , Regulação da Expressão Gênica , Células HeLa , Humanos , Células MCF-7 , Estabilidade Proteica , Transporte Proteico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-jun/genética , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitinação
7.
Curr Protoc Mouse Biol ; 7(3): 145-175, 2017 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-28884792

RESUMO

Complex biological systems are composed of multiple cell types whose transcriptional activity can vary due to differences in cell state, environmental stimulation, or intrinsic programs. Conventional bulk analysis methods capture the average transcriptional programs of the cell population, thus missing the unique cellular signature of each single cell. In recent years, the development of single-cell RNA-sequencing (scRNA-seq) technologies has provided a powerful approach to dissect the cellular heterogeneity of complex biological systems. However, such approaches require specialized equipment or are costly. In this article, we describe an improved Smart-seq2-based method to profile the transcriptome of hundreds of single cells simultaneously, without utilizing commercial kits or requiring any specialized single-cell capture/library preparation tools. Moreover, we introduce the Automated Single-cell Analysis Pipeline (ASAP), which allows researchers without strong computational expertise to explore scRNA-seq data using a wide range of commonly used algorithms and sophisticated visualization tools. © 2017 by John Wiley & Sons, Inc.


Assuntos
Análise de Célula Única , Transcriptoma , Animais , Sequência de Bases , Biblioteca Gênica , Humanos , Camundongos , Análise de Sequência de RNA
8.
Sci Rep ; 7: 42130, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28181539

RESUMO

Brown adipocytes regulate energy expenditure via mitochondrial uncoupling, which makes them attractive therapeutic targets to tackle obesity. However, the regulatory mechanisms underlying brown adipogenesis are still poorly understood. To address this, we profiled the transcriptome and chromatin state during mouse brown fat cell differentiation, revealing extensive gene expression changes and chromatin remodeling, especially during the first day post-differentiation. To identify putatively causal regulators, we performed transcription factor binding site overrepresentation analyses in active chromatin regions and prioritized factors based on their expression correlation with the bona-fide brown adipogenic marker Ucp1 across multiple mouse and human datasets. Using loss-of-function assays, we evaluated both the phenotypic effect as well as the transcriptomic impact of several putative regulators on the differentiation process, uncovering ZFP467, HOXA4 and Nuclear Factor I A (NFIA) as novel transcriptional regulators. Of these, NFIA emerged as the regulator yielding the strongest molecular and cellular phenotypes. To examine its regulatory function, we profiled the genomic localization of NFIA, identifying it as a key early regulator of terminal brown fat cell differentiation.


Assuntos
Adipócitos Marrons/metabolismo , Metabolismo Energético/genética , Fatores de Transcrição NFI/genética , Proteína Desacopladora 1/genética , Adipogenia/genética , Animais , Diferenciação Celular/genética , Montagem e Desmontagem da Cromatina/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento , Genômica , Proteínas de Homeodomínio , Humanos , Camundongos , Fatores de Transcrição , Transcriptoma/genética
9.
Biochem Biophys Res Commun ; 463(3): 275-9, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26009488

RESUMO

Clnk, as a third member of the Blnk/SLP-76 adapter family, is involved in the positive regulation of immunoreceptor signaling. Here we provide findings that Clnk may be is required for TNF induced cell death, it functions downstream of RIP3 and promotes TNF- induced ROS generation and MLKL tetramer formation and subsequent necrosis of L929 cells. Therefore, Clnk, as an adaptor protein, may take part in the other cellular processes.


Assuntos
Proteínas de Transporte/imunologia , Fibrossarcoma/imunologia , Espécies Reativas de Oxigênio/imunologia , Fator de Necrose Tumoral alfa/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Morte Celular , Linhagem Celular Tumoral , Fibrossarcoma/patologia , Células HeLa , Humanos , Camundongos , Necrose/imunologia , Proteínas Quinases/química , Proteínas Quinases/imunologia , Multimerização Proteica , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Transdução de Sinais
10.
Nat Cell Biol ; 17(4): 434-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25751141

RESUMO

The auto-phosphorylation of murine receptor-interacting protein 3 (Rip3) on Thr 231 and Ser 232 in the necrosome is required to trigger necroptosis. However, how Rip3 phosphorylation is regulated is still largely unknown. Here we identified protein phosphatase 1B (Ppm1b) as a Rip3 phosphatase and found that Ppm1b restricts necroptosis in two settings: spontaneous necroptosis caused by Rip3 auto-phosphorylation in resting cells, and tumour necrosis factor-α (TNF)-induced necroptosis in cultured cells. We revealed that Ppm1b selectively suppresses necroptosis through the dephosphorylation of Rip3, which then prevents the recruitment of mixed lineage kinase domain-like protein (Mlkl) to the necrosome. We further showed that Ppm1b deficiency (Ppm1b(d/d)) in mice enhanced TNF-induced death in a Rip3-dependent manner, and the role of Ppm1b in inhibiting necroptosis was evidenced by elevated Rip3 phosphorylation and tissue damage in the caecum of TNF-treated Ppm1b(d/d) mice. These data indicate that Ppm1b negatively regulates necroptosis through dephosphorylating Rip3 in vitro and in vivo.


Assuntos
Apoptose/genética , Fosfoproteínas Fosfatases/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Células 3T3 , Animais , Ceco/citologia , Linhagem Celular , Técnicas de Inativação de Genes , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Quinases/metabolismo , Proteína Fosfatase 2C , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Fator de Necrose Tumoral alfa
11.
Cell Host Microbe ; 17(2): 229-42, 2015 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-25674982

RESUMO

Necroptosis is a form of programmed necrosis that is mediated by signaling complexes containing the receptor-interacting protein 3 (RIP3) and RIP1 kinases. We show that RIP3 and its interaction with the herpes simplex virus type 1 (HSV-1) protein ICP6 triggers necroptosis in infected mouse cells and limits viral propagation in mice. ICP6 interacts with RIP1/RIP3 through its RHIM domain and forms dimers/oliogmers by its C-terminal R1 domain. These binding events result in RIP1-RIP3 hetero- and RIP3-RIP3 homo-interactions and subsequent necroptosis of HSV-1-infected mouse cells. However, ICP6 RHIM cannot trigger necroptosis and even inhibits TNF-induced necroptosis in human cells. As the RHIM domain in murine cytomegalovirus protein vIRA can inhibit necroptosis in both human and mouse cells, these data suggest that both viral and host RHIM sequences determine whether the virus-host RHIM interaction is pro- or anti-necroptotic and that some viruses may evolve to escape this restriction.


Assuntos
Morte Celular , Proteínas Ativadoras de GTPase/metabolismo , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno , Mapas de Interação de Proteínas , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Herpesvirus Humano 1/fisiologia , Humanos , Evasão da Resposta Imune , Camundongos , Ligação Proteica , Replicação Viral
12.
PLoS Pathog ; 10(4): e1004034, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24722736

RESUMO

Viruses hijack host factors for their high speed protein synthesis, but information about these factors is largely unknown. In searching for genes that are involved in viral replication, we carried out a forward genetic screen for Drosophila mutants that are more resistant or sensitive to Drosophila C virus (DCV) infection-caused death, and found a virus-resistant line in which the expression of pelo gene was deficient. Our mechanistic studies excluded the viral resistance of pelo deficient flies resulting from the known Drosophila anti-viral pathways, and revealed that pelo deficiency limits the high level synthesis of the DCV capsid proteins but has no or very little effect on the expression of some other viral proteins, bulk cellular proteins, and transfected exogenous genes. The restriction of replication of other types of viruses in pelo deficient flies was also observed, suggesting pelo is required for high level production of capsids of all kinds of viruses. We show that both pelo deficiency and high level DCV protein synthesis increase aberrant 80S ribosomes, and propose that the preferential requirement of pelo for high level synthesis of viral capsids is at least partly due to the role of pelo in dissociation of stalled 80S ribosomes and clearance of aberrant viral RNA and proteins. Our data demonstrated that pelo is a host factor that is required for high efficiency translation of viral capsids and targeting pelo could be a strategy for general inhibition of viral infection.


Assuntos
Dicistroviridae/fisiologia , Proteínas de Drosophila/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Proteínas Nucleares/metabolismo , Biossíntese de Proteínas/fisiologia , Proteínas Virais/biossíntese , Replicação Viral/fisiologia , Animais , Capsídeo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Mutação , Proteínas Nucleares/genética , Proteínas Virais/genética
13.
Cell Res ; 24(4): 417-32, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24513853

RESUMO

Formation of multi-component signaling complex necrosomes is essential for tumor necrosis factor α (TNF)-induced programmed necrosis (also called necroptosis). However, the mechanisms of necroptosis are still largely unknown. We isolated a TNF-resistant L929 mutant cell line generated by retrovirus insertion and identified that disruption of the guanine nucleotide-binding protein γ 10 (Gγ10) gene is responsible for this phenotype. We further show that Gγ10 is involved in TNF-induced necroptosis and Gß2 is the partner of Gγ10. Src is the downstream effector of Gß2γ10 in TNF-induced necroptosis because TNF-induced Src activation was impaired upon Gγ10 knockdown. Gγ10 does not affect TNF-induced activation of NF-κB and MAPKs and the formation of necrosomes, but is required for trafficking of necrosomes to their potential functioning site, an unidentified subcellular organelle that can be fractionated into heterotypic membrane fractions. The TNF-induced Gßγ-Src signaling pathway is independent of RIP1/RIP3 kinase activity and necrosome formation, but is required for the necrosome to function.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/genética , Vesículas Citoplasmáticas/efeitos dos fármacos , Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Quinases da Família src/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Vesículas Citoplasmáticas/metabolismo , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Necrose/induzido quimicamente , Necrose/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
14.
Cell Res ; 23(8): 994-1006, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23835476

RESUMO

Mixed lineage kinase domain-like protein (Mlkl) was recently found to interact with receptor interacting protein 3 (Rip3) and to be essential for tumor necrosis factor (TNF)-induced programmed necrosis (necroptosis) in cultured cell lines. We have generated Mlkl-deficient mice by transcription activator-like effector nucleases (TALENs)-mediated gene disruption and found Mlkl to be dispensable for normal mouse development as well as immune cell development. Mlkl-deficient mouse embryonic fibroblasts (MEFs) and macrophages both showed resistance to necrotic but not apoptotic stimuli. Mlkl-deficient MEFs and macrophages were indistinguishable from wild-type cells in their ability to activate NF-κB, ERK, JNK, and p38 in response to TNF and lipopolysaccharides (LPS), respectively. Consistently, Mlkl-deficient macrophages and mice exhibited normal interleukin-1ß (IL-1ß), IL-6, and TNF production after LPS treatment. Mlkl deficiency protects mice from cerulean-induced acute pancreatitis, a necrosis-related disease, but has no effect on polymicrobial septic shock-induced animal death. Our results provide genetic evidence for the role of Mlkl in necroptosis.


Assuntos
Apoptose , Proteínas Quinases/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Sequência de Bases , Linhagem Celular , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Necrose , Proteínas Quinases/deficiência , Proteínas Quinases/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Fatores de Necrose Tumoral/metabolismo
15.
J Biol Chem ; 288(23): 16247-16261, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23612963

RESUMO

Receptor interacting protein 3 (RIP3) is a protein kinase essential for TNF-induced necroptosis. Phosphorylation on Ser-227 in human RIP3 (hRIP3) is required for its interaction with human mixed lineage kinase domain-like (MLKL) in the necrosome, a signaling complex induced by TNF stimulation. RIP1 and RIP3 mediate necrosome aggregation leading to the formation of amyloid-like signaling complexes. We found that TNF induces Thr-231 and Ser-232 phosphorylation in mouse RIP3 (mRIP3) and this phosphorylation is required for mRIP3 to interact with mMLKL. Ser-232 in mRIP3 corresponds to Ser-227 in hRIP3, whereas Thr-231 is not conserved in hRIP3. Although the RIP3-MLKL interaction is required for necroptosis in both human and mouse cells, hRIP3 does not interact with mMLKL and mRIP3 cannot bind to hMLKL. The species specificity of the RIP3-MLKL interaction is primarily determined by the sequence differences in the phosphorylation sites and the flanking sequence around the phosphorylation sites in hRIP3 and mRIP3. It appears that the RIP3-MLKL interaction has been selected as an evolutionarily conserved mechanism in mediating necroptosis signaling despite that differing structural and mechanistic bases for this interaction emerged simultaneously in different organisms. In addition, we further revealed that the interaction of RIP3 with MLKL prevented massive abnormal RIP3 aggregation, and therefore should be crucial for formation of the amyloid signaling complex of necrosomes. We also found that the interaction between RIP3 and MLKL is required for the translocation of necrosomes to mitochondria-associated membranes. Our data demonstrate the importance of the RIP3-MLKL interaction in the formation of functional necrosomes and suggest that translocation of necrosomes to mitochondria-associated membranes is essential for necroptosis signaling.


Assuntos
Células Musculares/enzimologia , Proteínas Musculares/metabolismo , Proteínas Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais , Amiloide/genética , Amiloide/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Células Musculares/patologia , Proteínas Musculares/genética , Necrose/enzimologia , Necrose/genética , Necrose/patologia , Fosforilação/genética , Proteínas Quinases/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética
16.
Mol Biol Cell ; 18(11): 4681-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17855512

RESUMO

Mitochondrial adenine nucleotide translocase (ANT) is believed to be a component or a regulatory component of the mitochondrial permeability transition pore (mtPTP), which controls mitochondrial permeability transition during apoptosis. However, the role of ANT in apoptosis is still uncertain, because hepatocytes isolated from ANT knockout and wild-type mice are equally sensitive to TNF- and Fas-induced apoptosis. In a screen for genes required for tumor necrosis factor alpha (TNF-alpha)-induced apoptosis in MCF-7 human breast cancer cells using retrovirus insertion-mediated random mutagenesis, we discovered that the ANT3 gene is involved in TNF-alpha-induced cell death in MCF-7 cells. We further found that ANT3 is selectively required for TNF- and oxidative stress-induced cell death in MCF-7 cells, but it is dispensable for cell death induced by several other inducers. This data supplements previous data obtained from ANT knockout studies, indicating that ANT is involved in some apoptotic processes. We found that the resistance to TNF-alpha-induced apoptosis observed in ANT3 mutant (ANT3(mut)) cells is associated with a deficiency in the regulation of the mitochondrial membrane potential and cytochrome c release. It is not related to intracellular ATP levels or survival pathways, supporting a previous model in which ANT regulates mtPTP. Our study provides genetic evidence supporting a role of ANT in apoptosis and suggests that the involvement of ANT in cell death is cell type- and stimulus-dependent.


Assuntos
Translocador 3 do Nucleotídeo Adenina/metabolismo , Apoptose/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Translocador 3 do Nucleotídeo Adenina/genética , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mutação/genética , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA