Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 84(15): 2870-2881.e5, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39013473

RESUMO

The human silencing hub (HUSH) preserves genome integrity through the epigenetic repression of invasive genetic elements. However, despite our understanding of HUSH as an obligate complex of three subunits, only loss of MPP8 or Periphilin, but not TASOR, triggers interferon signaling following derepression of endogenous retroelements. Here, we resolve this paradox by characterizing a second HUSH complex that shares MPP8 and Periphilin but assembles around TASOR2, an uncharacterized paralog of TASOR. Whereas HUSH represses LINE-1 retroelements marked by the repressive histone modification H3K9me3, HUSH2 is recruited by the transcription factor IRF2 to repress interferon-stimulated genes. Mechanistically, HUSH-mediated retroelement silencing sequesters the limited pool of the shared subunits MPP8 and Periphilin, preventing TASOR2 from forming HUSH2 complexes and hence relieving the HUSH2-mediated repression of interferon-stimulated genes. Thus, competition between two HUSH complexes intertwines retroelement silencing with the induction of an immune response, coupling epigenetic and immune aspects of genome defense.


Assuntos
Inativação Gênica , Humanos , Células HEK293 , Histonas/metabolismo , Histonas/genética , Retroelementos/genética , Epigênese Genética , Elementos Nucleotídeos Longos e Dispersos/genética , Transdução de Sinais , Interferons/metabolismo , Interferons/imunologia , Interferons/genética , Células HeLa
2.
J Cell Sci ; 137(14)2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38904097

RESUMO

PTPRK is a receptor tyrosine phosphatase that is linked to the regulation of growth factor signalling and tumour suppression. It is stabilized at the plasma membrane by trans homophilic interactions upon cell-cell contact. PTPRK regulates cell-cell adhesion but is also reported to regulate numerous cancer-associated signalling pathways. However, the signalling mechanism of PTPRK remains to be determined. Here, we find that PTPRK regulates cell adhesion signalling, suppresses invasion and promotes collective, directed migration in colorectal cancer cells. In vivo, PTPRK supports recovery from inflammation-induced colitis. In addition, we confirm that PTPRK functions as a tumour suppressor in the mouse colon and in colorectal cancer xenografts. PTPRK regulates growth factor and adhesion signalling, and suppresses epithelial to mesenchymal transition (EMT). Contrary to the prevailing notion that PTPRK directly dephosphorylates EGFR, we find that PTPRK regulation of both EGFR and EMT is independent of its catalytic function. This suggests that additional adaptor and scaffold functions are important features of PTPRK signalling.


Assuntos
Transição Epitelial-Mesenquimal , Humanos , Animais , Camundongos , Receptores ErbB/metabolismo , Receptores ErbB/genética , Transdução de Sinais , Adesão Celular/genética , Movimento Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Colite/patologia , Colite/metabolismo , Colite/genética , Colite/induzido quimicamente , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Intestinos/patologia
3.
Blood ; 141(14): 1737-1754, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-36577137

RESUMO

HOXA9 is commonly upregulated in acute myeloid leukemia (AML), in which it confers a poor prognosis. Characterizing the protein interactome of endogenous HOXA9 in human AML, we identified a chromatin complex of HOXA9 with the nuclear matrix attachment protein SAFB. SAFB perturbation phenocopied HOXA9 knockout to decrease AML proliferation, increase differentiation and apoptosis in vitro, and prolong survival in vivo. Integrated genomic, transcriptomic, and proteomic analyses further demonstrated that the HOXA9-SAFB (H9SB)-chromatin complex associates with nucleosome remodeling and histone deacetylase (NuRD) and HP1γ to repress the expression of factors associated with differentiation and apoptosis, including NOTCH1, CEBPδ, S100A8, and CDKN1A. Chemical or genetic perturbation of NuRD and HP1γ-associated catalytic activity also triggered differentiation, apoptosis, and the induction of these tumor-suppressive genes. Importantly, this mechanism is operative in other HOXA9-dependent AML genotypes. This mechanistic insight demonstrates the active HOXA9-dependent differentiation block as a potent mechanism of disease maintenance in AML that may be amenable to therapeutic intervention by targeting the H9SB interface and/or NuRD and HP1γ activity.


Assuntos
Leucemia Mieloide Aguda , Proteínas de Ligação à Região de Interação com a Matriz , Humanos , Proteômica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Fatores de Transcrição/genética , Proteínas Associadas à Matriz Nuclear , Cromatina , Receptores de Estrogênio/genética , Receptores de Estrogênio/uso terapêutico , Proteínas de Ligação à Região de Interação com a Matriz/genética
4.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35105802

RESUMO

Human cytomegalovirus (HCMV) is an important human pathogen and a paradigm of viral immune evasion, targeting intrinsic, innate, and adaptive immunity. We have employed two orthogonal multiplexed tandem mass tag-based proteomic screens to identify host proteins down-regulated by viral factors expressed during the latest phases of viral infection. This approach revealed that the HIV-1 restriction factor Schlafen-11 (SLFN11) was degraded by the poorly characterized, late-expressed HCMV protein RL1, via recruitment of the Cullin4-RING E3 Ubiquitin Ligase (CRL4) complex. SLFN11 potently restricted HCMV infection, inhibiting the formation and spread of viral plaques. Overall, we show that a restriction factor previously thought only to inhibit RNA viruses additionally restricts HCMV. We define the mechanism of viral antagonism and also describe an important resource for revealing additional molecules of importance in antiviral innate immunity and viral immune evasion.


Assuntos
Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Evasão da Resposta Imune , Proteínas Nucleares/imunologia , Proteólise , Proteínas do Envelope Viral/imunologia , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Humanos , Proteínas Nucleares/genética , Complexos Ubiquitina-Proteína Ligase/genética , Complexos Ubiquitina-Proteína Ligase/imunologia , Proteínas do Envelope Viral/genética
5.
J Gen Virol ; 105(4)2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38687323

RESUMO

The human cytomegalovirus (HCMV) pUS2 glycoprotein exploits the host's endoplasmic reticulum (ER)-associated degradation (ERAD) pathway to degrade major histocompatibility complex class I (MHC-I) and prevent antigen presentation. Beyond MHC-I, pUS2 has been shown to target a range of cellular proteins for degradation, preventing their cell surface expression. Here we have identified a novel pUS2 target, ER-resident protein lectin mannose binding 2 like (LMAN2L). pUS2 expression was both necessary and sufficient for the downregulation of LMAN2L, which was dependent on the cellular E3 ligase TRC8. Given the hypothesized role of LMAN2L in the trafficking of glycoproteins, we employed proteomic plasma membrane profiling to measure LMAN2L-dependent changes at the cell surface. A known pUS2 target, integrin alpha-6 (ITGA6), was downregulated from the surface of LMAN2L-deficient cells, but not other integrins. Overall, these results suggest a novel strategy of pUS2-mediated protein degradation whereby pUS2 targets LMAN2L to impair trafficking of ITGA6. Given that pUS2 can directly target other integrins, we propose that this single viral protein may exhibit both direct and indirect mechanisms to downregulate key cell surface molecules.


Assuntos
Citomegalovirus , Retículo Endoplasmático , Proteínas do Envelope Viral , Proteínas Virais , Humanos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteólise , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Lectinas de Ligação a Manose/metabolismo , Lectinas de Ligação a Manose/genética , Degradação Associada com o Retículo Endoplasmático , Interações Hospedeiro-Patógeno , Membrana Celular/metabolismo , Membrana Celular/virologia
6.
PLoS Pathog ; 18(6): e1010612, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35727847

RESUMO

The interaction between immune cells and virus-infected targets involves multiple plasma membrane (PM) proteins. A systematic study of PM protein modulation by vaccinia virus (VACV), the paradigm of host regulation, has the potential to reveal not only novel viral immune evasion mechanisms, but also novel factors critical in host immunity. Here, >1000 PM proteins were quantified throughout VACV infection, revealing selective downregulation of known T and NK cell ligands including HLA-C, downregulation of cytokine receptors including IFNAR2, IL-6ST and IL-10RB, and rapid inhibition of expression of certain protocadherins and ephrins, candidate activating immune ligands. Downregulation of most PM proteins occurred via a proteasome-independent mechanism. Upregulated proteins included a decoy receptor for TRAIL. Twenty VACV-encoded PM proteins were identified, of which five were not recognised previously as such. Collectively, this dataset constitutes a valuable resource for future studies on antiviral immunity, host-pathogen interaction, poxvirus biology, vector-based vaccine design and oncolytic therapy.


Assuntos
Doenças Transmissíveis , Poxviridae , Vacínia , Humanos , Evasão da Resposta Imune , Proteínas de Membrana/metabolismo , Vaccinia virus
7.
Proc Natl Acad Sci U S A ; 117(31): 18771-18779, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32690704

RESUMO

Human cytomegalovirus (HCMV) is an important human pathogen and a paradigm of intrinsic, innate, and adaptive viral immune evasion. Here, we employed multiplexed tandem mass tag-based proteomics to characterize host proteins targeted for degradation late during HCMV infection. This approach revealed that mixed lineage kinase domain-like protein (MLKL), a key terminal mediator of cellular necroptosis, was rapidly and persistently degraded by the minimally passaged HCMV strain Merlin but not the extensively passaged strain AD169. The strain Merlin viral inhibitor of apoptosis pUL36 was necessary and sufficient both to degrade MLKL and to inhibit necroptosis. Furthermore, mutation of pUL36 Cys131 abrogated MLKL degradation and restored necroptosis. As the same residue is also required for pUL36-mediated inhibition of apoptosis by preventing proteolytic activation of procaspase-8, we define pUL36 as a multifunctional inhibitor of both apoptotic and necroptotic cell death.


Assuntos
Apoptose/fisiologia , Citomegalovirus , Necroptose/fisiologia , Proteínas Virais/metabolismo , Células Cultivadas , Citomegalovirus/química , Citomegalovirus/metabolismo , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Humanos , Ligação Proteica , Proteólise
8.
J Virol ; 95(20): e0069821, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34346763

RESUMO

Long noncoding RNAs (lncRNAs) are frequently associated with broad modulation of gene expression and thus provide the cell with the ability to synchronize entire metabolic processes. We used transcriptomic approaches to investigate whether the most abundant human cytomegalovirus-encoded lncRNA, RNA2.7, has this characteristic. By comparing cells infected with wild-type virus (WT) to cells infected with RNA2.7 deletion mutants, RNA2.7 was implicated in regulating a large number of cellular genes late in lytic infection. Pathway analysis indicated that >100 of these genes are associated with promoting cell movement, and the 10 most highly regulated of these were validated in further experiments. Morphological analysis and live cell tracking of WT- and RNA2.7 mutant-infected cells indicated that RNA2.7 is involved in promoting the movement and detachment of infected cells late in infection, and plaque assays using sparse cell monolayers indicated that RNA2.7 is also involved in promoting cell-to-cell spread of virus. Consistent with the observation that upregulated mRNAs are relatively A+U-rich, which is a trait associated with transcript instability, and that they are also enriched in motifs associated with mRNA instability, transcriptional inhibition experiments on WT- and RNA2.7 mutant-infected cells showed that four upregulated transcripts lived longer in the presence of RNA2.7. These findings demonstrate that RNA2.7 is required for promoting cell movement and viral spread late in infection and suggest that this may be due to general stabilization of A+U-rich transcripts. IMPORTANCE In addition to messenger RNAs (mRNAs), the human genome encodes a large number of long noncoding RNAs (lncRNAs). Many lncRNAs that have been studied in detail are associated with broad modulation of gene expression and have important biological roles. Human cytomegalovirus, which is a large, clinically important DNA virus, specifies four lncRNAs, one of which (RNA2.7) is expressed at remarkably high levels during lytic infection. Our studies show that RNA2.7 is required for upregulating a large number of human genes, about 100 of which are associated with cell movement, and for promoting the movement of infected cells and the spread of virus from one cell to another. Further bioinformatic and experimental analyses indicated that RNA2.7 may exert these effects by stabilizing mRNAs that are relatively rich in A and U nucleotides. These findings increase our knowledge of how human cytomegalovirus regulates the infected cell to promote its own success.


Assuntos
Citomegalovirus/genética , RNA Longo não Codificante/genética , Movimento Celular/genética , Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Humanos , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Viral/genética , Ativação Transcricional/genética , Transcriptoma , Regulação para Cima , Replicação Viral/genética
9.
Diabetologia ; 64(4): 890-902, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33501603

RESUMO

AIMS/HYPOTHESIS: Levels of the microRNA (miRNA) miR-126-3p are programmed cell-autonomously in visceral adipose tissue of adult offspring born to obese female C57BL/6J mice. The spectrum of miR-126-3p targets and thus the consequences of its dysregulation for adipocyte metabolism are unknown. Therefore, the aim of the current study was to identify novel targets of miR-126-3p in vitro and then establish the outcomes of their dysregulation on adipocyte metabolism in vivo using a well-established maternal obesity mouse model. METHODS: miR-126-3p overexpression in 3T3-L1 pre-adipocytes followed by pulsed stable isotope labelling by amino acids in culture (pSILAC) was performed to identify novel targets of the miRNA. Well-established bioinformatics algorithms and luciferase assays were then employed to confirm those that were direct targets of miR-126-3p. Selected knockdown experiments were performed in vitro to define the consequences of target dysregulation. Quantitative real-time PCR, immunoblotting, histology, euglycaemic-hyperinsulinaemic clamps and glucose tolerance tests were performed to determine the phenotypic and functional outcomes of maternal programmed miR-126-3p levels in offspring adipose tissue. RESULTS: The proteomic approach confirmed the identity of known targets of miR-126-3p (including IRS-1) and identified Lunapark, an endoplasmic reticulum (ER) protein, as a novel one. We confirmed by luciferase assay that Lunapark was a direct target of miR-126-3p. Overexpression of miR-126-3p in vitro led to a reduction in Lunapark protein levels and increased Perk (also known as Eif2ak3) mRNA levels and small interference-RNA mediated knockdown of Lunapark led to increased Xbp1, spliced Xbp1, Chop (also known as Ddit3) and Perk mRNA levels and an ER stress transcriptional response in 3T3-L1 pre-adipocytes. Consistent with the results found in vitro, increased miR-126-3p expression in adipose tissue from adult mouse offspring born to obese dams was accompanied by decreased Lunapark and IRS-1 protein levels and increased markers of ER stress. At the whole-body level the animals displayed glucose intolerance. CONCLUSIONS/INTERPRETATION: Concurrently targeting IRS-1 and Lunapark, a nutritionally programmed increase in miR-126-3p causes adipose tissue insulin resistance and an ER stress response, both of which may contribute to impaired glucose tolerance. These findings provide a novel mechanism by which obesity during pregnancy leads to increased risk of type 2 diabetes in the offspring and therefore identify miR-126-3p as a potential therapeutic target.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Estresse do Retículo Endoplasmático , Proteínas de Homeodomínio/metabolismo , MicroRNAs/metabolismo , Obesidade Materna/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Células 3T3-L1 , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Glicemia/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Proteínas de Homeodomínio/genética , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Obesidade Materna/genética , Obesidade Materna/patologia , Fenótipo , Gravidez , Transdução de Sinais
10.
PLoS Biol ; 16(1): e2004411, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29381698

RESUMO

The AP-5 adaptor protein complex is presumed to function in membrane traffic, but so far nothing is known about its pathway or its cargo. We have used CRISPR-Cas9 to knock out the AP-5 ζ subunit gene, AP5Z1, in HeLa cells, and then analysed the phenotype by subcellular fractionation profiling and quantitative mass spectrometry. The retromer complex had an altered steady-state distribution in the knockout cells, and several Golgi proteins, including GOLIM4 and GOLM1, were depleted from vesicle-enriched fractions. Immunolocalisation showed that loss of AP-5 led to impaired retrieval of the cation-independent mannose 6-phosphate receptor (CIMPR), GOLIM4, and GOLM1 from endosomes back to the Golgi region. Knocking down the retromer complex exacerbated this phenotype. Both the CIMPR and sortilin interacted with the AP-5-associated protein SPG15 in pull-down assays, and we propose that sortilin may act as a link between Golgi proteins and the AP-5/SPG11/SPG15 complex. Together, our findings suggest that AP-5 functions in a novel sorting step out of late endosomes, acting as a backup pathway for retromer. This provides a mechanistic explanation for why mutations in AP-5/SPG11/SPG15 cause cells to accumulate aberrant endolysosomes, and highlights the role of endosome/lysosome dysfunction in the pathology of hereditary spastic paraplegia and other neurodegenerative disorders.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sistemas CRISPR-Cas , Endossomos/fisiologia , Complexo de Golgi/fisiologia , Células HeLa , Humanos , Lisossomos/genética , Lisossomos/fisiologia , Espectrometria de Massas , Proteínas de Membrana/metabolismo , Fenótipo , Transporte Proteico , Paraplegia Espástica Hereditária/genética , Proteínas de Transporte Vesicular/metabolismo
11.
Proc Natl Acad Sci U S A ; 115(19): 4998-5003, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29691324

RESUMO

CD58 is an adhesion molecule that is known to play a critical role in costimulation of effector cells and is intrinsic to immune synapse structure. Herein, we describe a virally encoded gene that inhibits CD58 surface expression. Human cytomegalovirus (HCMV) UL148 was necessary and sufficient to promote intracellular retention of CD58 during HCMV infection. Blocking studies with antagonistic anti-CD58 mAb and an HCMV UL148 deletion mutant (HCMV∆UL148) with restored CD58 expression demonstrated that the CD2/CD58 axis was essential for the recognition of HCMV-infected targets by CD8+ HCMV-specific cytotoxic T lymphocytes (CTLs). Further, challenge of peripheral blood mononuclear cells ex vivo with HCMV∆UL148 increased both CTL and natural killer (NK) cell degranulation against HCMV-infected cells, including NK-driven antibody-dependent cellular cytotoxicity, showing that UL148 is a modulator of the function of multiple effector cell subsets. Our data stress the effect of HCMV immune evasion functions on shaping the immune response, highlighting the capacity for their potential use in modulating immunity during the development of anti-HCMV vaccines and HCMV-based vaccine vectors.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Evasão da Resposta Imune , Imunidade Celular , Células Matadoras Naturais/imunologia , Proteínas Virais de Fusão/imunologia , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Transformada , Citomegalovirus/genética , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/patologia , Humanos , Células Matadoras Naturais/patologia , Proteínas Virais de Fusão/genética
12.
J Virol ; 93(16)2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31142673

RESUMO

BK polyomavirus (BKPyV) is a small DNA virus that establishes a life-long persistent infection in the urinary tract of most people. BKPyV is known to cause severe morbidity in renal transplant recipients and can lead to graft rejection. The simple 5.2-kbp double-stranded DNA (dsDNA) genome expresses just seven known proteins; thus, it relies heavily on the host machinery to replicate. How the host proteome changes over the course of infection is key to understanding this host-virus interplay. Here, for the first time quantitative temporal viromics has been used to quantify global changes in >9,000 host proteins in two types of primary human epithelial cells throughout 72 h of BKPyV infection. These data demonstrate the importance of cell cycle progression and pseudo-G2 arrest in effective BKPyV replication, along with a surprising lack of an innate immune response throughout the whole virus replication cycle. BKPyV thus evades pathogen recognition to prevent activation of innate immune responses in a sophisticated manner.IMPORTANCE BK polyomavirus can cause serious problems in immune-suppressed patients, in particular, kidney transplant recipients who can develop polyomavirus-associated kidney disease. In this work, we have used advanced proteomics techniques to determine the changes to protein expression caused by infection of two independent primary cell types of the human urinary tract (kidney and bladder) throughout the replication cycle of this virus. Our findings have uncovered new details of a specific form of cell cycle arrest caused by this virus, and, importantly, we have identified that this virus has a remarkable ability to evade detection by host cell defense systems. In addition, our data provide an important resource for the future study of kidney epithelial cells and their infection by urinary tract pathogens.


Assuntos
Vírus BK/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular , Imunidade Inata , Infecções por Polyomavirus/imunologia , Infecções por Polyomavirus/metabolismo , Infecções por Polyomavirus/virologia , Proteoma , Proteômica , Biomarcadores , Proteínas de Ciclo Celular/metabolismo , Resistência à Doença , Suscetibilidade a Doenças/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Proteômica/métodos , Fluxo de Trabalho
13.
PLoS Genet ; 13(4): e1006698, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28403141

RESUMO

The cellular machinery required for the fusion of constitutive secretory vesicles with the plasma membrane in metazoans remains poorly defined. To address this problem we have developed a powerful, quantitative assay for measuring secretion and used it in combination with combinatorial gene depletion studies in Drosophila cells. This has allowed us to identify at least three SNARE complexes mediating Golgi to PM transport (STX1, SNAP24/29 and Syb; STX1, SNAP24/29 and YKT6; STX4, SNAP24 and Syb). RNAi mediated depletion of YKT6 and VAMP3 in mammalian cells also blocks constitutive secretion suggesting that YKT6 has an evolutionarily conserved role in this process. The unexpected role of YKT6 in plasma membrane fusion may in part explain why RNAi and gene disruption studies have failed to produce the expected phenotypes in higher eukaryotes.


Assuntos
Membrana Celular/genética , Proteínas de Drosophila/genética , Proteínas R-SNARE/genética , Proteínas SNARE/genética , Proteína 3 Associada à Membrana da Vesícula/genética , Animais , Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Heterozigoto , Humanos , Fusão de Membrana/genética , Transporte Proteico/genética , Proteínas R-SNARE/metabolismo , Interferência de RNA , Proteínas SNARE/metabolismo , Toxina Shiga I/genética , Toxina Shiga I/metabolismo , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/genética , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Proteína 3 Associada à Membrana da Vesícula/metabolismo
14.
PLoS Genet ; 13(3): e1006620, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28267784

RESUMO

Tubulointerstitial kidney disease is an important cause of progressive renal failure whose aetiology is incompletely understood. We analysed a large pedigree with maternally inherited tubulointerstitial kidney disease and identified a homoplasmic substitution in the control region of the mitochondrial genome (m.547A>T). While mutations in mtDNA coding sequence are a well recognised cause of disease affecting multiple organs, mutations in the control region have never been shown to cause disease. Strikingly, our patients did not have classical features of mitochondrial disease. Patient fibroblasts showed reduced levels of mitochondrial tRNAPhe, tRNALeu1 and reduced mitochondrial protein translation and respiration. Mitochondrial transfer demonstrated mitochondrial transmission of the defect and in vitro assays showed reduced activity of the heavy strand promoter. We also identified further kindreds with the same phenotype carrying a homoplasmic mutation in mitochondrial tRNAPhe (m.616T>C). Thus mutations in mitochondrial DNA can cause maternally inherited renal disease, likely mediated through reduced function of mitochondrial tRNAPhe.


Assuntos
DNA Mitocondrial/genética , Nefropatias/genética , Túbulos Renais/patologia , Mutação , Acetilglucosaminidase/urina , Biópsia , Feminino , Fibroblastos/metabolismo , Ligação Genética , Humanos , Leucina/química , Masculino , Mitocôndrias/metabolismo , Consumo de Oxigênio , Linhagem , Fenótipo , Fenilalanina/química , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Músculo Quadríceps/patologia , RNA de Transferência/genética
15.
Proc Natl Acad Sci U S A ; 114(35): 9421-9426, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28811369

RESUMO

An intergenic region of human chromosome 2 (2p25.3) harbors genetic variants which are among those most strongly and reproducibly associated with obesity. The gene closest to these variants is TMEM18, although the molecular mechanisms mediating these effects remain entirely unknown. Tmem18 expression in the murine hypothalamic paraventricular nucleus (PVN) was altered by changes in nutritional state. Germline loss of Tmem18 in mice resulted in increased body weight, which was exacerbated by high fat diet and driven by increased food intake. Selective overexpression of Tmem18 in the PVN of wild-type mice reduced food intake and also increased energy expenditure. We provide evidence that TMEM18 has four, not three, transmembrane domains and that it physically interacts with key components of the nuclear pore complex. Our data support the hypothesis that TMEM18 itself, acting within the central nervous system, is a plausible mediator of the impact of adjacent genetic variation on human adiposity.


Assuntos
Apetite/genética , Peso Corporal/genética , Proteínas de Membrana/metabolismo , Obesidade/genética , Animais , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas de Transporte Vesicular
16.
Mol Syst Biol ; 14(8): e8376, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-30072421

RESUMO

Circadian rhythms are cell-autonomous biological oscillations with a period of about 24 h. Current models propose that transcriptional feedback loops are the primary mechanism for the generation of circadian oscillations. Within this framework, Drosophila S2 cells are regarded as "non-rhythmic" cells, as they do not express several canonical circadian components. Using an unbiased multi-omics approach, we made the surprising discovery that Drosophila S2 cells do in fact display widespread daily rhythms. Transcriptomics and proteomics analyses revealed that hundreds of genes and their products, and in particular metabolic enzymes, are rhythmically expressed in a 24-h cycle. Metabolomics analyses extended these findings and demonstrate that central carbon metabolism and amino acid metabolism are core metabolic pathways driven by protein rhythms. We thus demonstrate that 24-h metabolic oscillations, coupled to gene and protein cycles, take place in nucleated cells without the contribution of any known circadian regulators. These results therefore suggest a reconsideration of existing models of the clockwork in Drosophila and other eukaryotic systems.


Assuntos
Relógios Biológicos/genética , Ritmo Circadiano/genética , Drosophila melanogaster/genética , Transcriptoma/genética , Animais , Drosophila melanogaster/metabolismo , Metabolômica , Proteoma/genética
17.
PLoS Pathog ; 11(4): e1004811, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25875600

RESUMO

Human cytomegalovirus (HCMV) US2, US3, US6 and US11 act in concert to prevent immune recognition of virally infected cells by CD8+ T-lymphocytes through downregulation of MHC class I molecules (MHC-I). Here we show that US2 function goes far beyond MHC-I degradation. A systematic proteomic study using Plasma Membrane Profiling revealed US2 was unique in downregulating additional cellular targets, including: five distinct integrin α-chains, CD112, the interleukin-12 receptor, PTPRJ and thrombomodulin. US2 recruited the cellular E3 ligase TRC8 to direct the proteasomal degradation of all its targets, reminiscent of its degradation of MHC-I. Whereas integrin α-chains were selectively degraded, their integrin ß1 binding partner accumulated in the ER. Consequently integrin signaling, cell adhesion and migration were strongly suppressed. US2 was necessary and sufficient for degradation of the majority of its substrates, but remarkably, the HCMV NK cell evasion function UL141 requisitioned US2 to enhance downregulation of the NK cell ligand CD112. UL141 retained CD112 in the ER from where US2 promoted its TRC8-dependent retrotranslocation and degradation. These findings redefine US2 as a multifunctional degradation hub which, through recruitment of the cellular E3 ligase TRC8, modulates diverse immune pathways involved in antigen presentation, NK cell activation, migration and coagulation; and highlight US2's impact on HCMV pathogenesis.


Assuntos
Evasão da Resposta Imune/imunologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cromatografia Líquida de Alta Pressão , Citomegalovirus/imunologia , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Espectrometria de Massas , Proteínas de Membrana/metabolismo , Proteômica/métodos , RNA Interferente Pequeno , Transdução Genética
18.
Lancet ; 385 Suppl 1: S66, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26312888

RESUMO

BACKGROUND: Less than 100 years since it was first transmitted to the human population HIV-1 infects more than 30 million people worldwide and causes almost 2 million AIDS-related deaths every year. Viruses manipulate cellular genes and pathways to benefit their survival, and the study of cell surface proteins downregulated by viruses has provided insights into both viral pathogenesis and crucial aspects of cell biology. We aimed to identify novel cell surface proteins targeted for downregulation by HIV-1. METHODS: We combined plasma membrane enrichment through selective aminooxybiotinylation with tandem mass tag-based quantitative proteomics (plasma membrane profiling) to map expression timecourses of more than 800 plasma membrane proteins in T cells infected in vitro with HIV-1. Novel substrates of the viral accessory proteins Vpu and Nef were sought by use of deletion viruses and single gene overexpression. FINDINGS: Our proteomic datasets defined more than 100 previously unsuspected cell surface targets of HIV-1, particularly proteins involved in T-cell activation, cell adhesion, and aminoacid transport. In addition to its known targets, Vpu was found to be necessary and sufficient for the downregulation of the aminoacid transporter TOV3. Downregulation of TOV3 was post transcriptional, mediated by the ß-TrCP ubiquitin E3 ligase and occurred via an endolysosomal pathway. TOV3 was highly expressed in primary human CD4 T cells, and depletion of TOV3 by RNA interference markedly impaired the mitogenic response to CD3/CD28 stimulation. We identified alanine as an endogenous TOV3 substrate, and showed that extracellular alanine was crucial for T-cell proliferation. INTERPRETATION: This study suggests that TOV3 downregulation is restricted to Vpu variants from the lineage of HIV-1 group M viruses giving rise to pandemic AIDS. Antagonism of alanine transport in CD4 T cells might contribute to HIV-1 pathogenesis through modulation of virus production, impairment of the adaptive immune response, or enhancement of CD4 T-cell loss. FUNDING: Wellcome Trust, Addenbrooke's Charitable Trust, Raymond and Beverly Sackler Foundation.

19.
Proc Natl Acad Sci U S A ; 110(35): 14290-5, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23929775

RESUMO

Misfolded MHC class I heavy chains (MHC I HCs) are targeted for endoplasmic reticulum (ER)-associated degradation (ERAD) by the ubiquitin E3 ligase HRD1, and E2 ubiquitin conjugating enzyme UBE2J1, and represent one of the few known endogenous ERAD substrates. The mechanism by which misfolded proteins are dislocated across the ER membrane into the cytosol is unclear. Here, we investigate the requirements for MHC I ubiquitination and degradation and show that endogenous misfolded MHC I HCs are recognized in the ER lumen by EDEM1 in a glycan-dependent manner and targeted to the core SEL1L/HRD1/UBE2J1 complex. A soluble MHC I HC lacking its transmembrane domain and cytosolic tail uses the same ERAD components and is degraded as efficiently as wild-type MHC I. Unexpectedly, HRD1-dependent polyubiquitination is preferentially targeted to the ER luminal domain of full-length MHC I HCs, despite the presence of an exposed cytosolic C-terminal tail. MHC I luminal domain ubiquitination occurs before p97 ATPase-mediated extraction from the ER membrane and can be targeted to nonlysine, as well as lysine, residues. A subset of integral membrane proteins, therefore, requires an early dislocation event to expose part of their luminal domain to the cytosol, before HRD1-mediated polyubiquitination and dislocation.


Assuntos
Retículo Endoplasmático/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Adenosina Trifosfatases/metabolismo , Sequência de Bases , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Ubiquitinação
20.
Proc Natl Acad Sci U S A ; 110(7): 2557-62, 2013 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-23359686

RESUMO

SNPs in the first intron of FTO (fat mass and obesity associated) are strongly associated with human obesity. While it is not yet formally established that this effect is mediated through the actions of the FTO protein itself, loss of function mutations in FTO or its murine homologue Fto result in severe growth retardation, and mice globally overexpressing FTO are obese. The mechanisms through which FTO influences growth and body composition are unknown. We describe a role for FTO in the coupling of amino acid levels to mammalian target of rapamycin complex 1 signaling. These findings suggest that FTO may influence body composition through playing a role in cellular nutrient sensing.


Assuntos
Aminoácidos/metabolismo , Composição Corporal/genética , Obesidade/genética , Proteínas/genética , Proteínas/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Fracionamento Celular , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Fibroblastos , Células HEK293 , Humanos , Imunoprecipitação , Camundongos , Polimorfismo de Nucleotídeo Único/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa