Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Ann Rheum Dis ; 2022 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-35922125

RESUMO

OBJECTIVES: Rheumatic immune-related adverse events (irAE) such as (poly)arthritis in patients undergoing immune checkpoint inhibitor (ICI) treatment pose a major clinical challenge. ICI therapy improves CD8+ T cell (CD8) function, but CD8 contributes to chronic inflammation in autoimmune arthritis (AA). Thus, we investigated whether immune functional and metabolic changes in CD8 explain the development of musculoskeletal irAE in ICI-treated patients. METHODS: Peripheral CD8 obtained from ICI-treated patients with and without arthritis irAEs and from AA patients with and without a history of malignancy were stimulated in media containing 13C-labelled glucose with and without tofacitinib or infliximab. Changes in metabolism, immune-mediator release, expression of effector cell-surface molecules and inhibition of tumour cell growth were quantified. RESULTS: CD8 from patients with irAE showed significantly lower frequency and expression of cell-surface molecule characteristic for activation, effector-functions, homing, exhaustion and apoptosis and reduced release of cytotoxic and proinflammatory immune mediators compared with CD8 from ICI patients who did not develop irAE. This was accompanied by a higher glycolytic rate and ATP production. Gene-expression analysis of pre-ICI-treated CD8 revealed several differentially expressed transcripts in patients who later developed arthritis irAEs. In vitro tofacitinib or infliximab treatment did not significantly change the immune-metabolic profile nor the capacity to release cytolytic mediators that inhibit the growth of the human lung cancer cell line H838. CONCLUSIONS: Our study shows that CD8 from ICI-treated patients who develop a musculoskeletal irAE has a distinct immune-effector and metabolic profile from those that remain irAE free. This specific irAE profile overlaps with the one observed in CD8 from AA patients and may prove useful for novel therapeutic strategies to manage ICI-induced irAEs.

2.
Sci Rep ; 9(1): 12911, 2019 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-31501484

RESUMO

Induced morphology changes of cells and organelles are by far the easiest way to determine precise protein sub-locations and organelle quantities in light microscopy. By using hypotonic solutions to swell mammalian cell organelles we demonstrate that precise membrane, lumen or matrix protein locations within the endoplasmic reticulum, Golgi and mitochondria can reliably be established. We also show the benefit of this approach for organelle quantifications, especially for clumped or intertwined organelles like peroxisomes and mitochondria. Since cell and organelle swelling is reversible, it can be applied to live cells for successive high-resolution analyses. Our approach outperforms many existing imaging modalities with respect to resolution, ease-of-use and cost-effectiveness without excluding any co-utilization with existing optical (super)resolution techniques.


Assuntos
Forma Celular , Soluções Hipotônicas , Organelas , Animais , Linhagem Celular , Humanos , Microscopia de Fluorescência , Imagem Óptica , Organelas/metabolismo , Transporte Proteico
3.
EMBO Rep ; 20(1)2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30467237

RESUMO

CDC14A codes for a conserved proline-directed phosphatase, and mutations in the gene are associated with autosomal-recessive severe to profound deafness, due to defective kinocilia. A role of CDC14A in cilia formation has also been described in other organisms. However, how human CDC14A impacts on cilia formation remains unclear. Here, we show that human RPE1 hCDC14APD cells, encoding a phosphatase dead version of hCDC14A, have longer cilia than wild-type cells, while hCDC14A overexpression reduces cilia formation. Phospho-proteome analysis of ciliated RPE1 cells identified actin-associated and microtubule binding proteins regulating cilia length as hCDC14A substrates, including the actin-binding protein drebrin. Indeed, we find that hCDC14A counteracts the CDK5-dependent phosphorylation of drebrin at S142 during ciliogenesis. Further, we show that drebrin and hCDC14A regulate the recruitment of the actin organizer Arp2 to centrosomes. In addition, during ciliogenesis hCDC14A also regulates endocytosis and targeting of myosin Va vesicles to the basal body in a drebrin-independent manner, indicating that it impacts primary cilia formation in a multilayered manner.


Assuntos
Proteína 2 Relacionada a Actina/genética , Cílios/genética , Neuropeptídeos/genética , Monoéster Fosfórico Hidrolases/genética , Actinas/genética , Linhagem Celular , Movimento Celular/genética , Centrossomo/metabolismo , Cílios/metabolismo , Quinase 5 Dependente de Ciclina/genética , Endocitose/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Microtúbulos/genética , Mutação , Cadeias Pesadas de Miosina/genética , Miosina Tipo V/genética , Fosforilação , Ligação Proteica , Proteínas Tirosina Fosfatases , Proteoma/genética
4.
J Biol Chem ; 294(8): 2786-2800, 2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30578301

RESUMO

The endoplasmic reticulum (ER), as a multifunctional organelle, plays crucial roles in lipid biosynthesis and calcium homeostasis as well as the synthesis and folding of secretory and membrane proteins. Therefore, it is of high importance to maintain ER homeostasis and to adapt ER function and morphology to cellular needs. Here, we show that signal peptide peptidase (SPP) modulates the ER shape through degradation of morphogenic proteins. Elevating SPP activity induces rapid rearrangement of the ER and formation of dynamic ER clusters. Inhibition of SPP activity rescues the phenotype without the need for new protein synthesis, and this rescue depends on a pre-existing pool of proteins in the Golgi. With the help of organelle proteomics, we identified certain membrane proteins to be diminished upon SPP expression and further show that the observed morphology changes depend on SPP-mediated cleavage of ER morphogenic proteins, including the SNARE protein syntaxin-18. Thus, we suggest that SPP-mediated protein abundance control by a regulatory branch of ER-associated degradation (ERAD-R) has a role in shaping the early secretory pathway.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Degradação Associada com o Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Organelas/metabolismo , Proteínas Qa-SNARE/metabolismo , Células HEK293 , Humanos , Proteólise , Proteômica
5.
Mol Biol Cell ; 29(26): 3105-3118, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30354798

RESUMO

Microtubule nucleation was uncovered as a key principle of spindle assembly. However, the mechanistic details about microtubule nucleation and the organization of spindle formation and symmetry are currently being revealed. Here we describe the function of coiled-coil domain containing 61 (Ccdc61), a so far uncharacterized centrosomal protein, in spindle assembly and symmetry. Our data describe that Ccdc61 is required for spindle assembly and precise chromosome alignments in mitosis. Microtubule tip-tracking experiments in the absence of Ccdc61 reveal a clear loss of the intrinsic symmetry of microtubule tracks within the spindle. Furthermore, we show that Ccdc61 controls the centrosomal localization of centrosomal protein of 170 kDa (Cep170), a protein that was shown previously to localize to centrosomes as well as spindle microtubules and promotes microtubule organization and microtubule assembly. Interestingly, selective disruption of Ccdc61 impairs the binding between Cep170 and TANK binding kinase 1, an interaction that is required for microtubule stability. In summary, we have discovered Ccdc61 as a centrosomal protein with an important function in mitotic microtubule organization.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Mitose , Proteínas Serina-Treonina Quinases/genética , Fuso Acromático/metabolismo , Fatores de Transcrição/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Centrossomo , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Fuso Acromático/ultraestrutura , Fatores de Transcrição/metabolismo
6.
Stroke ; 49(6): 1479-1487, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29760276

RESUMO

BACKGROUND AND PURPOSE: Detection and localization of the early phase of blood-brain barrier disruption (BBBD) in vivo during cerebral ischemia/reperfusion injury remain a major challenge but may be a relevant outcome parameter in stroke. METHODS: We studied early BBBD in mice after transient middle cerebral artery occlusion by multimodal, high-field (9.4T) in vivo magnetic resonance imaging, including the contrast agent gadofluorineM as an albumin-binding tracer. GadofluorineM contrast-enhanced magnetic resonance imaging was performed to determine BBBD at 2, 6, and 24 hours after reperfusion. BBBD was confirmed and localized along the microvascular tree by using fluorescent gadofluorineM and immunofluorescence stainings (cluster of differentiation 31, ephrin type-B receptor 4, alpha smooth muscle actin, ionized calcium binding adaptor molecule 1). RESULTS: GadofluorineM contrast-enhanced magnetic resonance imaging revealed a multifocal spatial distribution of early BBBD and its close association with the microvasculature at a resolution of 40 µm. GadofluorineM leakage was closely associated with ephrin type-B receptor 4-positive but not alpha smooth muscle actin-positive vessels. The multifocal pattern of early BBBD (already at 2 hours after reperfusion) thus occurred in the distal capillary and venular microvascular bed. These multifocal zones showed distinct imaging signs indicative of early vasogenic edema. The total volume of multifocal early BBBD accurately predicted infarct size at 24 hours after reperfusion. CONCLUSIONS: Early BBBD in focal cerebral ischemia initiates multifocally in the distal capillary and venular bed of the cerebral microvasculature. It is closely associated with perimicrovascular vasogenic edema and microglial activation and predicts the extent of final infarction.


Assuntos
Barreira Hematoencefálica/patologia , Isquemia Encefálica/patologia , Capilares/patologia , Acidente Vascular Cerebral/patologia , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Edema Encefálico/patologia , Circulação Cerebrovascular/fisiologia , Infarto da Artéria Cerebral Média/patologia , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/patologia
7.
Elife ; 5: e11794, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26943317

RESUMO

Ribosome stalling during translation can potentially be harmful, and is surveyed by a conserved quality control pathway that targets the associated mRNA and nascent polypeptide chain (NC). In this pathway, the ribosome-associated quality control (RQC) complex promotes the ubiquitylation and degradation of NCs remaining stalled in the 60S subunit. NC stalling is recognized by the Rqc2/Tae2 RQC subunit, which also stabilizes binding of the E3 ligase, Listerin/Ltn1. Additionally, Rqc2 modifies stalled NCs with a carboxy-terminal, Ala- and Thr-containing extension-the 'CAT tail'. However, the function of CAT tails and fate of CAT tail-modified ('CATylated') NCs has remained unknown. Here we show that CATylation mediates formation of detergent-insoluble NC aggregates. CATylation and aggregation of NCs could be observed either by inactivating Ltn1 or by analyzing NCs with limited ubiquitylation potential, suggesting that inefficient targeting by Ltn1 favors the Rqc2-mediated reaction. These findings uncover a translational stalling-dependent protein aggregation mechanism, and provide evidence that proteins can become specifically marked for aggregation.


Assuntos
Peptídeos/metabolismo , Agregação Patológica de Proteínas , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas de Ligação a RNA/metabolismo , Ribossomos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo
8.
Cancer Res ; 76(1): 96-107, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26669866

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) carries the most dismal prognosis of all solid tumors and is generally strongly resistant to currently available chemo- and/or radiotherapy regimens, including targeted molecular therapies. Therefore, unraveling the molecular mechanisms underlying the aggressive behavior of pancreatic cancer is a necessary prerequisite for the development of novel therapeutic approaches. We previously identified the protein placenta-specific 8 (PLAC8, onzin) in a genome-wide search for target genes associated with pancreatic tumor progression and demonstrated that PLAC8 is strongly ectopically expressed in advanced preneoplastic lesions and invasive human PDAC. However, the molecular function of PLAC8 remained unclear, and accumulating evidence suggested its role is highly dependent on cellular and physiologic context. Here, we demonstrate that in contrast to other cellular systems, PLAC8 protein localizes to the inner face of the plasma membrane in pancreatic cancer cells, where it interacts with specific membranous structures in a temporally and spatially stable manner. Inhibition of PLAC8 expression strongly inhibited pancreatic cancer cell growth by attenuating cell-cycle progression, which was associated with transcriptional and/or posttranslational modification of the central cell-cycle regulators CDKN1A, retinoblastoma protein, and cyclin D1 (CCND1), but did not impact autophagy. Moreover, Plac8 deficiency significantly inhibited tumor formation in genetically engineered mouse models of pancreatic cancer. Together, our findings establish PLAC8 as a central mediator of tumor progression in PDAC and as a promising candidate gene for diagnostic and therapeutic targeting.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Prognóstico , Proteínas/genética , Análise Serial de Tecidos , Transfecção
9.
Aging (Albany NY) ; 7(11): 911-27, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26546739

RESUMO

The TERT gene encodes for the reverse transcriptase activity of the telomerase complex and mutations in TERT can lead to dysfunctional telomerase activity resulting in diseases such as dyskeratosis congenita (DKC). Here, we describe a novel TERT mutation at position T1129P leading to DKC with progressive bone marrow (BM) failure in homozygous members of a consanguineous family. BM hematopoietic stem cells (HSCs) of an affected family member were 300-fold reduced associated with a significantly impaired colony forming capacity in vitro and impaired repopulation activity in mouse xenografts. Recent data in yeast suggested improved cellular checkpoint controls by mTOR inhibition preventing cells with short telomeres or DNA damage from dividing. To evaluate a potential therapeutic option for the patient, we treated her primary skin fibroblasts and BM HSCs with the mTOR inhibitor rapamycin. This led to prolonged survival and decreased levels of senescence in T1129P mutant fibroblasts. In contrast, the impaired HSC function could not be improved by mTOR inhibition, as colony forming capacity and multilineage engraftment potential in xenotransplanted mice remained severely impaired. Thus, rapamycin treatment did not rescue the compromised stem cell function of TERTT1129P mutant patient HSCs and outlines limitations of a potential DKC therapy based on rapamycin.


Assuntos
Antígenos CD34/análise , Senescência Celular , Disceratose Congênita/genética , Células-Tronco Hematopoéticas/fisiologia , Mutação , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Telomerase/genética , Animais , Feminino , Células HeLa , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Telômero
11.
Sci Rep ; 5: 12457, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26208256

RESUMO

We introduce Map3-2D, a freely available software to accurately project up to five-dimensional (5D) fluorescence microscopy image data onto full-content 2D maps. Similar to the Earth's projection onto cartographic maps, Map3-2D unfolds surface information from a stack of images onto a single, structurally connected map. We demonstrate its applicability for visualization and quantitative analyses of spherical and uneven surfaces in fixed and dynamic live samples by using mammalian and yeast cells, and giant unilamellar vesicles. Map3-2D software is available at http://www.zmbh.uni-heidelberg.de//Central_Services/Imaging_Facility/Map3-2D.html.

12.
Autophagy ; 11(9): 1484-98, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26101826

RESUMO

Mutations in PINK1 and PARK2/Parkin are a main risk factor for familial Parkinson disease. While the physiological mechanism of their activation is unclear, these proteins have been shown in tissue culture cells to serve as a key trigger for autophagy of depolarized mitochondria. Here we show that ablation of the mitochondrial rhomboid protease PARL leads to retrograde translocation of an intermembrane space-bridging PINK1 import intermediate. Subsequently, it is rerouted to the outer membrane in order to recruit PARK2, which phenocopies mitophagy induction by uncoupling agents. Consistent with a role of this retrograde translocation mechanism in neurodegenerative disease, we show that pathogenic PINK1 mutants which are not cleaved by PARL affect PINK1 kinase activity and the ability to induce PARK2-mediated mitophagy. Altogether we suggest that PARL is an important intrinsic player in mitochondrial quality control, a system substantially impaired in Parkinson disease as indicated by reduced removal of damaged mitochondria in affected patients.


Assuntos
Membranas Intracelulares/enzimologia , Metaloproteases/metabolismo , Proteínas Mitocondriais/metabolismo , Mitofagia , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Biocatálise , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Células HEK293 , Humanos , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Modelos Biológicos , Proteínas Mutantes/metabolismo , Doença de Parkinson/patologia , Processamento de Proteína Pós-Traducional , Solubilidade
13.
J Cell Biol ; 202(1): 81-95, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23816619

RESUMO

Meiotic maturation in vertebrate oocytes is an excellent model system for microtubule reorganization during M-phase spindle assembly. Here, we surveyed changes in the pattern of microtubule-interacting proteins upon Xenopus laevis oocyte maturation by quantitative proteomics. We identified the synovial sarcoma X breakpoint protein (SSX2IP) as a novel spindle protein. Using X. laevis egg extracts, we show that SSX2IP accumulated at spindle poles in a Dynein-dependent manner and interacted with the γ-tubulin ring complex (γ-TuRC) and the centriolar satellite protein PCM-1. Immunodepletion of SSX2IP impeded γ-TuRC loading onto centrosomes. This led to reduced microtubule nucleation and spindle assembly failure. In rapidly dividing blastomeres of medaka (Oryzias latipes) and in somatic cells, SSX2IP knockdown caused fragmentation of pericentriolar material and chromosome segregation errors. We characterize SSX2IP as a novel centrosome maturation and maintenance factor that is expressed at the onset of vertebrate development. It preserves centrosome integrity and faithful mitosis during the rapid cleavage division of blastomeres and in somatic cells.


Assuntos
Centríolos/metabolismo , Centrossomo/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Blastômeros/metabolismo , Blastômeros/patologia , Centríolos/genética , Segregação de Cromossomos , Cromossomos/genética , Cromossomos/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Técnicas de Silenciamento de Genes , Mitose , Proteínas de Neoplasias/genética , Oócitos/metabolismo , Oryzias/embriologia , Oryzias/genética , Oryzias/metabolismo , Proteômica , Proteínas Repressoras/genética , Fuso Acromático/genética , Fuso Acromático/metabolismo , Imagem com Lapso de Tempo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Xenopus laevis/genética
14.
Nat Cell Biol ; 15(2): 179-88, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23314863

RESUMO

Eps8, a bi-functional actin cytoskeleton remodeller, is a positive regulator of cell proliferation and motility. Here, we describe an unrecognized mechanism regulating Eps8 that is required for proper mitotic progression: whereas Eps8 is stable in G1 and S phase, its half-life drops sharply in G2. This requires G2-specific proteasomal degradation mediated by the ubiquitin E3 ligase SCF(Fbxw5). Consistent with a short window of degradation, Eps8 disappears from the cell cortex early in mitosis, but reappears at the midzone of dividing cells. Failure to reduce Eps8 levels in G2 prolongs its localization at the cell cortex and markedly delays cell rounding and prometaphase duration. However, during late stages of mitosis and cytokinesis, Eps8 capping activity is required to prevent membrane blebbing and cell-shape deformations. Our findings identify SCF(Fbxw5)-driven fluctuation of Eps8 levels as an important mechanism that contributes to cell-shape changes during entry into-and exit from-mitosis.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Forma Celular , Proteínas F-Box/metabolismo , Mitose , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Ligação Competitiva , Proteínas F-Box/genética , Pontos de Checagem da Fase G2 do Ciclo Celular , Células HEK293 , Meia-Vida , Células HeLa , Humanos , Metáfase , Camundongos , Camundongos Knockout , Microscopia de Vídeo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Estabilidade Proteica , Proteólise , Interferência de RNA , Fatores de Tempo , Transfecção , Ubiquitinação
15.
PLoS One ; 7(7): e39703, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22808051

RESUMO

The endoplasmic reticulum (ER) forms contacts with the plasma membrane. These contacts are known to function in non-vesicular lipid transport and signaling. Ist2 resides in specific domains of the ER in Saccharomyces cerevisiae where it binds phosphoinositide lipids at the cytosolic face of the plasma membrane. Here, we report that Ist2 recruits domains of the yeast ER to the plasma membrane. Ist2 determines the amount of cortical ER present and the distance between the ER and the plasma membrane. Deletion of IST2 resulted in an increased distance between ER and plasma membrane and allowed access of ribosomes to the space between the two membranes. Cells that overexpress Ist2 showed an association of the nucleus with the plasma membrane. The morphology of the ER and yeast growth were sensitive to the abundance of Ist2. Moreover, Ist2-dependent effects on cytosolic pH and genetic interactions link Ist2 to the activity of the H(+) pump Pma1 in the plasma membrane during cellular adaptation to the growth phase of the culture. Consistently we found a partial colocalization of Ist2-containing cortical ER and Pma1-containing domains of the plasma membrane. Hence Ist2 may be critically positioned in domains that couple functions of the ER and the plasma membrane.


Assuntos
Membrana Celular/genética , Retículo Endoplasmático/genética , Regulação Fúngica da Expressão Gênica , ATPases Translocadoras de Prótons/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Citosol/metabolismo , Citosol/ultraestrutura , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Concentração de Íons de Hidrogênio , Microscopia Confocal , Microscopia de Fluorescência , Fosfatidilinositóis/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Ribossomos/metabolismo , Ribossomos/ultraestrutura , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais
16.
J Cell Sci ; 124(Pt 8): 1301-7, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21444755

RESUMO

Tail-anchored (TA) proteins are post-translationally targeted to and inserted into the endoplasmic reticulum (ER) membrane through their single C-terminal transmembrane domain. Membrane insertion of TA proteins in mammalian cells is mediated by the ATPase TRC40/Asna1 (Get3 in yeast) and a receptor in the ER membrane. We have identified tryptophan-rich basic protein (WRB), also known as congenital heart disease protein 5 (CHD5), as the ER membrane receptor for TRC40/Asna1. WRB shows sequence similarity to Get1, a subunit of the membrane receptor complex for yeast Get3. Using biochemical and cell imaging approaches, we demonstrate that WRB is an ER-resident membrane protein that interacts with TRC40/Asna1 and recruits it to the ER membrane. We identify the coiled-coil domain of WRB as the binding site for TRC40/Asna1 and show that a soluble form of the coiled-coil domain interferes with TRC40/Asna1-mediated membrane insertion of TA proteins. The identification of WRB as a component of the TRC (Get) pathway for membrane insertion of TA proteins raises new questions concerning the proposed roles of WRB (CHD5) in congenital heart disease, and heart and eye development.


Assuntos
ATPases Transportadoras de Arsenito/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas Nucleares/metabolismo , ATPases Transportadoras de Arsenito/química , ATPases Transportadoras de Arsenito/genética , Retículo Endoplasmático/química , Retículo Endoplasmático/genética , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico
17.
J Neurochem ; 117(5): 856-67, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21426348

RESUMO

Intramembrane proteolysis is a conserved mechanism that regulates a variety of cellular processes ranging from transcription control to signaling. In mitochondria, the inner membrane rhomboid protease PARL has been implicated in the control of life span and apoptosis by a so far uncharacterized mechanism. Here, we show that PARL cleaves human Pink1, which is implicated in Parkinson's disease, within its conserved membrane anchor. Mature Pink1 is then free to be released into the cytosol or the mitochondrial intermembrane space. Upon depolarization of the mitochondrial membrane potential, the canonical import of Pink1 and PARL-catalyzed processing is blocked, leading to accumulation of the Pink1 precursor. As targeting of this precursor to the outer mitochondrial membrane has been shown to trigger mitophagy, we suggest that the PARL-catalyzed removal of the Pink1 signal sequence in the canonical import pathway acts as a cellular checkpoint for mitochondrial integrity. Furthermore, we show that two Parkinson's disease-causing mutations decrease the processing of Pink1 by PARL, with attendant implications for pathogenesis.


Assuntos
Metaloproteases/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Quinases/metabolismo , Sequência de Aminoácidos , Western Blotting , Carbonatos/química , Linhagem Celular , Metabolismo Energético/fisiologia , Células HEK293 , Humanos , Potenciais da Membrana/fisiologia , Membranas Mitocondriais/fisiologia , Dados de Sequência Molecular , Mutação/genética , Peptídeo Hidrolases/química , Plasmídeos/genética , Proteínas Quinases/biossíntese , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Frações Subcelulares/metabolismo , Transfecção
18.
Curr Protoc Cell Biol ; Chapter 5: Unit 5.7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21154552

RESUMO

Correct localization and topology are crucial for a protein's cellular function. To determine topologies of membrane proteins, a new technique, called fluorescence protease protection (FPP) assay, has recently been established. The sole requirements for FPP are the expression of fluorescent-protein fusion proteins and the selective permeabilization of the plasma membrane, permitting a wide range of cell types and organelles to be investigated. Proteins topologies in organelles like endoplasmic reticulum, Golgi apparatus, mitochondria, peroxisomes, and autophagosomes have already been determined by FPP. Here, two different step-by-step protocols of the FPP assay are provided. First, we describe the FPP assay using fluorescence microscopy for single adherent cells, and second, we outline the FPP assay for high-throughput screening applications.


Assuntos
Técnicas Citológicas , Proteínas de Membrana/química , Microscopia de Fluorescência/métodos , Animais , Linhagem Celular , Ensaios de Triagem em Larga Escala/métodos , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Peptídeo Hidrolases/metabolismo , Transporte Proteico , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Análise de Célula Única
19.
Nature ; 466(7305): E1, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20651645

RESUMO

During mitosis in Saccharomyces cerevisiae, senescence factors such as extrachromosomal ribosomal DNA circles (ERCs) are retained in the mother cell and excluded from the bud/daughter cell. Shcheprova et al. proposed a model suggesting segregation of ERCs through their association with nuclear pore complexes (NPCs) and retention of pre-existing NPCs in the mother cell during mitosis. However, this model is inconsistent with previous data and we demonstrate here that NPCs do efficiently migrate from the mother into the bud. Therefore, binding to NPCs does not seem to explain the retention of ERCs in the mother cell.


Assuntos
DNA Fúngico/metabolismo , Mitose , Poro Nuclear/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Segregação de Cromossomos/fisiologia , DNA Ribossômico/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/análise , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/análise , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
20.
Methods Mol Biol ; 440: 227-33, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18369949

RESUMO

Determining a protein's correct topological distribution within the cell is essential for understanding the proper functioning of many proteins. Here, we describe a fluorescence-based technique, termed FPP for fluorescence protease protection, to determine protein topology in living cells. The FPP assay uses the restricted proteolytic digestibility of green fluorescent protein-tagged membrane proteins to reveal their intramembrane orientation. Membrane protein topology can be assessed using this technique for proteins residing in organelles as diverse as the Golgi apparatus, the endoplasmic reticulum (ER), peroxisomes, mitochondria, and autophagosomes. To illustrate the technique, we describe its use for deciphering the topology of a membrane protein in the ER.


Assuntos
Bioensaio/métodos , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Peptídeo Hidrolases/metabolismo , Animais , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular , Células Cultivadas , Digitonina/farmacologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinética , Proteínas de Membrana/química , Conformação Proteica , Desnaturação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA