Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 607(7919): 593-603, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35768510

RESUMEN

Aggressive and metastatic cancers show enhanced metabolic plasticity1, but the precise underlying mechanisms of this remain unclear. Here we show how two NOP2/Sun RNA methyltransferase 3 (NSUN3)-dependent RNA modifications-5-methylcytosine (m5C) and its derivative 5-formylcytosine (f5C) (refs.2-4)-drive the translation of mitochondrial mRNA to power metastasis. Translation of mitochondrially encoded subunits of the oxidative phosphorylation complex depends on the formation of m5C at position 34 in mitochondrial tRNAMet. m5C-deficient human oral cancer cells exhibit increased levels of glycolysis and changes in their mitochondrial function that do not affect cell viability or primary tumour growth in vivo; however, metabolic plasticity is severely impaired as mitochondrial m5C-deficient tumours do not metastasize efficiently. We discovered that CD36-dependent non-dividing, metastasis-initiating tumour cells require mitochondrial m5C to activate invasion and dissemination. Moreover, a mitochondria-driven gene signature in patients with head and neck cancer is predictive for metastasis and disease progression. Finally, we confirm that this metabolic switch that allows the metastasis of tumour cells can be pharmacologically targeted through the inhibition of mitochondrial mRNA translation in vivo. Together, our results reveal that site-specific mitochondrial RNA modifications could be therapeutic targets to combat metastasis.


Asunto(s)
5-Metilcitosina , Citosina/análogos & derivados , Glucólisis , Mitocondrias , Metástasis de la Neoplasia , Fosforilación Oxidativa , ARN Mitocondrial , 5-Metilcitosina/biosíntesis , 5-Metilcitosina/metabolismo , Antígenos CD36 , Supervivencia Celular , Citosina/metabolismo , Progresión de la Enfermedad , Glucólisis/efectos de los fármacos , Humanos , Metilación/efectos de los fármacos , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Fosforilación Oxidativa/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mitocondrial/genética , ARN Mitocondrial/metabolismo , ARN de Transferencia de Metionina/genética , ARN de Transferencia de Metionina/metabolismo
2.
Hepatology ; 77(4): 1211-1227, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35776660

RESUMEN

BACKGROUND AND AIMS: In hereditary hemorrhagic telangiectasia (HHT), severe liver vascular malformations are associated with mutations in the Activin A Receptor-Like Type 1 ( ACVRL1 ) gene encoding ALK1, the receptor for bone morphogenetic protein (BMP) 9/BMP10, which regulates blood vessel development. Here, we established an HHT mouse model with exclusive liver involvement and adequate life expectancy to investigate ALK1 signaling in liver vessel formation and metabolic function. APPROACH AND RESULTS: Liver sinusoidal endothelial cell (LSEC)-selective Cre deleter line, Stab2-iCreF3 , was crossed with Acvrl1 -floxed mice to generate LSEC-specific Acvrl1 -deficient mice ( Alk1HEC-KO ). Alk1HEC-KO mice revealed hepatic vascular malformations and increased posthepatic flow, causing right ventricular volume overload. Transcriptomic analyses demonstrated induction of proangiogenic/tip cell gene sets and arterialization of hepatic vessels at the expense of LSEC and central venous identities. Loss of LSEC angiokines Wnt2 , Wnt9b , and R-spondin-3 ( Rspo3 ) led to disruption of metabolic liver zonation in Alk1HEC-KO mice and in liver specimens of patients with HHT. Furthermore, prion-like protein doppel ( Prnd ) and placental growth factor ( Pgf ) were upregulated in Alk1HEC-KO hepatic endothelial cells, representing candidates driving the organ-specific pathogenesis of HHT. In LSEC in vitro , stimulation or inhibition of ALK1 signaling counter-regulated Inhibitors of DNA binding (ID)1-3, known Alk1 transcriptional targets. Stimulation of ALK1 signaling and inhibition of ID1-3 function confirmed regulation of Wnt2 and Rspo3 by the BMP9/ALK1/ID axis. CONCLUSIONS: Hepatic endothelial ALK1 signaling protects from development of vascular malformations preserving organ-specific endothelial differentiation and angiocrine signaling. The long-term surviving Alk1HEC-KO HHT model offers opportunities to develop targeted therapies for this severe disease.


Asunto(s)
Telangiectasia Hemorrágica Hereditaria , Ratones , Femenino , Animales , Telangiectasia Hemorrágica Hereditaria/genética , Células Endoteliales/metabolismo , Factor de Crecimiento Placentario/metabolismo , Hígado/patología , Transducción de Señal , Factor 2 de Diferenciación de Crecimiento/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo
3.
Int J Cancer ; 153(1): 173-182, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36444499

RESUMEN

Chronic inflammation, linked to the presence of bovine milk and meat factors (BMMFs) and specific subsets of macrophages, results in oxygen radical synthesis and induction of mutations in DNA of actively replicating cells and replicating single stranded DNA. Cancers arising from this process have been characterized as indirect carcinogenesis by infectious agents (without persistence of genes of the agent in premalignant or cancers cells). Here, we investigate structural properties of pleomorphic vesicles, regularly identified by staining peritumor tissues of colorectal, lung and pancreatic cancer for expression of BMMF Rep. The latter represents a subgroup of BMMF1 proteins involved in replication of small single-stranded circular plasmids of BMMF, but most likely also contributing to pleomorphic vesicular structures found in the periphery of colorectal, lung and pancreatic cancers. Structurally dense regions are demonstrated in preselected areas of colorectal cancer, after staining with monoclonal antibodies against BMMF1 Rep. Similar structures were observed in human embryonic cells (HEK293TT) overexpressing Rep. These data suggest that Rep or Rep isoforms contribute to the structural formation of vesicles.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Pancreáticas , Humanos , Animales , Leche , Replicación del ADN , Plásmidos , Neoplasias Pancreáticas/genética , Pulmón , Carne , Neoplasias Colorrectales/genética
4.
J Cell Sci ; 134(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34704600

RESUMEN

Osteoclasts form special integrin-mediated adhesion structures called sealing zones that enable them to adhere to and resorb bone. Sealing zones consist of densely packed podosomes tightly interconnected by actin fibers. Their formation requires the presence of the hematopoietic integrin regulator kindlin-3 (also known as Fermt3). In this study, we investigated osteoclasts and their adhesion structures in kindlin-3 hypomorphic mice expressing only 5-10% of the kindlin-3 level of wild-type mice. Low kindlin-3 expression reduces integrin activity, results in impaired osteoclast adhesion and signaling, and delays cell spreading. Despite these defects, in vitro-generated kindlin-3-hypomorphic osteoclast-like cells arrange their podosomes into adhesion patches and belts, but their podosome and actin organization is abnormal. Remarkably, kindlin-3-hypomorphic osteoclasts form sealing zones when cultured on calcified matrix in vitro and on bone surface in vivo. However, functional assays, immunohistochemical staining and electron micrographs of bone sections showed that they fail to seal the resorption lacunae properly, which is required for secreted proteinases to digest bone matrix. This results in mild osteopetrosis. Our study reveals a new, hitherto understudied function of kindlin-3 as an essential organizer of integrin-mediated adhesion structures, such as sealing zones.


Asunto(s)
Proteínas del Citoesqueleto , Osteoclastos , Osteopetrosis , Animales , Matriz Ósea , Huesos , Proteínas del Citoesqueleto/genética , Integrinas , Ratones , Osteopetrosis/genética
5.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34638879

RESUMEN

Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is the rate-limiting enzyme in the glycosphingolipid (GSL)-biosynthesis pathway and overexpressed in many human tumors. We suppressed GSL-biosynthesis using the GCS inhibitor Genz-123346 (Genz), NB-DNJ (Miglustat) or by genetic targeting of the GCS-encoding gene UDP-glucose-ceramide-glucosyltransferase- (UGCG). GCS-inhibition or GSL-depletion led to a marked arrest of the cell cycle in Lovo cells. UGCG silencing strongly also inhibited tumor spheroid growth in Lovo cells and moderately in HCT116 cells. MS/MS analysis demonstrated markedly elevated levels of sphingomyelin (SM) and phosphatidylcholine (PC) that occurred in a Genz-concentration dependent manner. Ultrastructural analysis of Genz-treated cells indicated multi-lamellar lipid storage in vesicular compartments. In mice, Genz lowered the incidence of experimentally induced colorectal tumors and in particular the growth of colorectal adenomas. These results highlight the potential for GCS-based inhibition in the treatment of CRC.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Neoplasias del Colon , Dioxanos/farmacología , Glicoesfingolípidos , Pirrolidinas/farmacología , Esferoides Celulares , Animales , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/metabolismo , Glicoesfingolípidos/biosíntesis , Glicoesfingolípidos/genética , Células HCT116 , Humanos , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
6.
J Biol Chem ; 294(8): 2786-2800, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30578301

RESUMEN

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.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Orgánulos/metabolismo , Proteínas Qa-SNARE/metabolismo , Células HEK293 , Humanos , Proteolisis , Proteómica
7.
Cell Mol Life Sci ; 76(23): 4783-4794, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31168659

RESUMEN

RATIONALE: Mitochondrial homeostasis has recently emerged as a focal point in the pathophysiology of idiopathic pulmonary fibrosis (IPF), but conflicting data have been reported regarding its regulation. We speculated that phosphoglycerate mutase family member 5 (PGAM5), a mitochondrial protein at the intersection of multiple cell death and mitochondrial turnover pathways, might be involved in the pathogenesis of IPF. METHODS: PGAM5-deficient mice and human pulmonary epithelial cells were analyzed comparatively with PGAM5-proficient controls in a bleomycin-based model of pulmonary fibrogenesis. Mitochondria were visualized by confocal and transmission electron microscopy. Mitochondrial homeostasis was assessed using JC1 (ΔΨ) and flow cytometry. RESULTS: PGAM5 plays an important role in pulmonary fibrogenesis. Pgam5-/- mice displayed significantly attenuated lung fibrosis compared to controls. Complementary, in vitro studies demonstrated that PGAM5 impaired mitochondrial integrity on a functional and structural level independently of mtROS-production. On a molecular level, reduced mitophagy caused by PGAM5 deficiency improved mitochondrial homeostasis. CONCLUSIONS: Our study identifies PGAM5 as an important regulator of mitochondrial homeostasis in pulmonary fibrosis. Our data further indicate PGAM5-mediated mitophagy itself as a pivotal gateway event in the mediation of self-sustaining mitochondrial damage and membrane depolarization. Our work hereby highlights the importance of mitochondrial dynamics and identifies a potential therapeutic target that warrants further studies. Toxic agents lead to mitochondrial damage resulting in depolarization of the mitochondrial membrane potential (ΔΨ) which is a gateway event for the initiation of PGAM5-mediated mitophagy. PGAM5-mediated mitophagy in turn leads to a self-perpetuating escalation of ΔΨ depolarization. Loss of the mitophagy-based damage-enhancing loop under PGAM5-deficient conditions breaks this vicious cycle, leading to improved mitochondrial homeostasis.


Asunto(s)
Mitocondrias/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fibrosis Pulmonar/patología , Células A549 , Animales , Bleomicina/farmacología , Modelos Animales de Enfermedad , Edición Génica , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Fosfoproteínas Fosfatasas/genética , Proteínas Quinasas/metabolismo , Fibrosis Pulmonar/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
Int J Mol Sci ; 21(15)2020 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-32759826

RESUMEN

Small extracellular vesicles (sEVs) are nanoparticles responsible for cell-to-cell communication released by healthy and cancer cells. Different roles have been described for sEVs in physiological and pathological contexts, including acceleration of tissue regeneration, modulation of tumor microenvironment, or premetastatic niche formation, and they are discussed as promising biomarkers for diagnosis and prognosis in body fluids. Although efforts have been made to standardize techniques for isolation and characterization of sEVs, current protocols often result in co-isolation of soluble protein or lipid complexes and of other extracellular vesicles. The risk of contaminated preparations is particularly high when isolating sEVs from tissues. As a consequence, the interpretation of data aiming at understanding the functional role of sEVs remains challenging and inconsistent. Here, we report an optimized protocol for isolation of sEVs from human and murine lymphoid tissues. sEVs from freshly resected human lymph nodes and murine spleens were isolated comparing two different approaches-(1) ultracentrifugation on a sucrose density cushion and (2) combined ultracentrifugation with size-exclusion chromatography. The purity of sEV preparations was analyzed using state-of-the-art techniques, including immunoblots, nanoparticle tracking analysis, and electron microscopy. Our results clearly demonstrate the superiority of size-exclusion chromatography, which resulted in a higher yield and purity of sEVs, and we show that their functionality alters significantly between the two isolation protocols.


Asunto(s)
Separación Celular/métodos , Exosomas/química , Vesículas Extracelulares/química , Tejido Linfoide/química , Animales , Exosomas/genética , Humanos , Lípidos/química , Ratones , Ultracentrifugación
9.
Dev Biol ; 433(1): 84-93, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29155043

RESUMEN

Spermiogenesis is the final phase during sperm cell development in which round spermatids undergo dramatic morphological changes to generate spermatozoa. Here we report that the serine/threonine kinase Stk33 is essential for the differentiation of round spermatids into functional sperm cells and male fertility. Constitutive Stk33 deletion in mice results in severely malformed and immotile spermatozoa that are particularly characterized by disordered structural tail elements. Stk33 expression first appears in primary spermatocytes, and targeted deletion of Stk33 in these cells recapitulates the defects observed in constitutive knockout mice, confirming a germ cell-intrinsic function. Stk33 protein resides in the cytoplasm and partially co-localizes with the caudal end of the manchette, a transient structure that guides tail elongation, in elongating spermatids, and loss of Stk33 leads to the appearance of a tight, straight and elongated manchette. Together, these results identify Stk33 as an essential regulator of spermatid differentiation and male fertility.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Espermátides/enzimología , Animales , Diferenciación Celular/fisiología , Fertilidad/fisiología , Masculino , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Espermatocitos/citología , Espermatocitos/enzimología , Espermatogénesis/fisiología , Espermatozoides/enzimología , Testículo/enzimología
10.
EMBO J ; 34(6): 778-97, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25672362

RESUMEN

Disruption of the functional protein balance in living cells activates protective quality control systems to repair damaged proteins or sequester potentially cytotoxic misfolded proteins into aggregates. The established model based on Saccharomyces cerevisiae indicates that aggregating proteins in the cytosol of eukaryotic cells partition between cytosolic juxtanuclear (JUNQ) and peripheral deposits. Substrate ubiquitination acts as the sorting principle determining JUNQ deposition and subsequent degradation. Here, we show that JUNQ unexpectedly resides inside the nucleus, defining a new intranuclear quality control compartment, INQ, for the deposition of both nuclear and cytosolic misfolded proteins, irrespective of ubiquitination. Deposition of misfolded cytosolic proteins at INQ involves chaperone-assisted nuclear import via nuclear pores. The compartment-specific aggregases, Btn2 (nuclear) and Hsp42 (cytosolic), direct protein deposition to nuclear INQ and cytosolic (CytoQ) sites, respectively. Intriguingly, Btn2 is transiently induced by both protein folding stress and DNA replication stress, with DNA surveillance proteins accumulating at INQ. Our data therefore reveal a bipartite, inter-compartmental protein quality control system linked to DNA surveillance via INQ and Btn2.


Asunto(s)
Sistemas de Transporte de Aminoácidos/fisiología , Compartimento Celular/fisiología , Citosol/metabolismo , Proteínas de Choque Térmico/fisiología , Agregado de Proteínas/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/fisiología , Sistemas de Transporte de Aminoácidos/metabolismo , Western Blotting , Células HeLa , Proteínas de Choque Térmico/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Modelos Biológicos , Pliegue de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Imagen de Lapso de Tiempo , Ubiquitinación
11.
ACS Nano ; 18(3): 2500-2519, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38207106

RESUMEN

Glioblastoma is a deadly brain tumor for which there is no cure. The presence of glioblastoma stem-like cells (GSCs) contributes to the heterogeneous nature of the disease and makes developing effective therapies challenging. Glioblastoma cells have been shown to influence their environment by releasing biological nanostructures known as extracellular vesicles (EVs). Here, we investigated the role of GSC-derived nanosized EVs (<200 nm) in glioblastoma heterogeneity, plasticity, and aggressiveness, with a particular focus on their protein, metabolite, and fatty acid content. We showed that conditioned medium and small extracellular vesicles (sEVs) derived from cells of one glioblastoma subtype induced transcriptomic and proteomic changes in cells of another subtype. We found that GSC-derived sEVs are enriched in proteins playing a role in the transmembrane transport of amino acids, carboxylic acids, and organic acids, growth factor binding, and metabolites associated with amino acid, carboxylic acid, and sugar metabolism. This suggests a dual role of GSC-derived sEVs in supplying neighboring GSCs with valuable metabolites and proteins responsible for their transport. Moreover, GSC-derived sEVs were enriched in saturated fatty acids, while their respective cells were high in unsaturated fatty acids, supporting that the loading of biological cargos into sEVs is a highly regulated process and that GSC-derived sEVs could be sources of saturated fatty acids for the maintenance of glioblastoma cell metabolism. Interestingly, sEVs isolated from GSCs of the proneural and mesenchymal subtypes are enriched in specific sets of proteins, metabolites, and fatty acids, suggesting a molecular collaboration between transcriptionally different glioblastoma cells. In summary, this study revealed the complexity of GSC-derived sEVs and unveiled their potential contribution to tumor heterogeneity and critical cellular processes commonly deregulated in glioblastoma.


Asunto(s)
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Humanos , Glioblastoma/patología , Ácidos Grasos/análisis , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacología , Proteómica , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Vesículas Extracelulares/química , Neoplasias Encefálicas/patología
12.
Nat Commun ; 15(1): 6067, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39025856

RESUMEN

After recognizing its ligand lipopolysaccharide, Toll-like receptor 4 (TLR4) recruits adaptor proteins to the cell membrane, thereby initiating downstream signaling and triggering inflammation. Whether this recruitment of adaptor proteins is dependent solely on protein-protein interactions is unknown. Here, we report that the sphingolipid sphinganine physically interacts with the adaptor proteins MyD88 and TIRAP and promotes MyD88 recruitment in macrophages. Myeloid cell-specific deficiency in serine palmitoyltransferase long chain base subunit 2, which encodes the key enzyme catalyzing sphingolipid biosynthesis, decreases the membrane recruitment of MyD88 and inhibits inflammatory responses in in vitro bone marrow-derived macrophage and in vivo sepsis models. In a melanoma mouse model, serine palmitoyltransferase long chain base subunit 2 deficiency decreases anti-tumor myeloid cell responses and increases tumor growth. Therefore, sphinganine biosynthesis is required for the initiation of TLR4 signal transduction and serves as a checkpoint for macrophage pattern recognition in sepsis and melanoma mouse models.


Asunto(s)
Macrófagos , Melanoma , Factor 88 de Diferenciación Mieloide , Sepsis , Serina C-Palmitoiltransferasa , Esfingosina , Receptor Toll-Like 4 , Animales , Receptor Toll-Like 4/metabolismo , Sepsis/metabolismo , Macrófagos/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Ratones , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Melanoma/metabolismo , Melanoma/patología , Melanoma/genética , Serina C-Palmitoiltransferasa/metabolismo , Serina C-Palmitoiltransferasa/genética , Humanos , Transducción de Señal , Modelos Animales de Enfermedad , Inflamación/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Células HEK293 , Lipopolisacáridos
13.
J Cell Sci ; 123(Pt 3): 392-400, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20130140

RESUMEN

Promyelocytic leukemia nuclear bodies (PML-NBs) are mobile subnuclear organelles formed by PML and Sp100 protein. They have been reported to have a role in transcription, DNA replication and repair, telomere lengthening, cell cycle control and tumor suppression. We have conducted high-resolution 4Pi fluorescence laser-scanning microscopy studies complemented with correlative electron microscopy and investigations of the accessibility of the PML-NB subcompartment. During interphase PML-NBs adopt a spherical organization characterized by the assembly of PML and Sp100 proteins into patches within a 50- to 100-nm-thick shell. This spherical shell of PML and Sp100 imposes little constraint to the exchange of components between the PML-NB interior and the nucleoplasm. Post-translational SUMO modifications, telomere repeats and heterochromatin protein 1 were found to localize in characteristic patterns with respect to PML and Sp100. From our findings, we derived a model that explains how the three-dimensional organization of PML-NBs serves to concentrate different biological activities while allowing for an efficient exchange of components.


Asunto(s)
Cuerpos de Inclusión Intranucleares/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Antígenos Nucleares/metabolismo , Antígenos Nucleares/ultraestructura , Autoantígenos/metabolismo , Autoantígenos/ultraestructura , Línea Celular Tumoral , Células HeLa , Humanos , Cuerpos de Inclusión Intranucleares/ultraestructura , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Modelos Biológicos , Proteínas Nucleares/ultraestructura , Proteína de la Leucemia Promielocítica , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/ultraestructura , Proteínas Supresoras de Tumor/ultraestructura , Ubiquitinas/metabolismo
14.
J Cell Biol ; 172(3): 373-81, 2006 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-16431928

RESUMEN

The bimolecular fluorescence complementation (BiFC) assay, which allows the investigation of interacting molecules in vivo, was applied to study complex formation between the splicing factor Y14 and nuclear export factor 1 (NXF1), which evidence indicates are functionally associated with nuclear mRNA. Y14 linked to the COOH terminus of yellow fluorescent protein (YFP; YC-Y14), and NXF1 fused to the NH2 terminus of YFP (YN-NXF1) expressed in MCF7 cells yielded BiFC upon specific binding. Fluorescence accumulated within and around nuclear speckles, suggesting the involvement of speckles in mRNA processing and export. Accordingly, BiFC depended on transcription and full-length NXF1. Coimmunoprecipitation of YC-Y14 with YN-NXF1, NXF1, Y14, and RNA indicated that YC-Y14 and YN-NXF1 functionally associate with RNA. Fluorescence recovery after photobleaching and fluorescence loss in photobleaching revealed that roughly half of the accumulated BiFC complexes were immobile in vivo. This immobile fraction was readily depleted by adenosine triphosphate (ATP) administration in permeabilized cells. These results suggest that a fraction of RNA, which remains in the nucleus for several hours despite its association with splicing and export proteins, accumulates in speckles because of an ATP-dependent mechanism.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Mapeo de Interacción de Proteínas , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Digitonina/farmacología , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Espacio Intranuclear/metabolismo , Cinética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mutación/genética , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Unión Proteica , ARN Polimerasa II/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleoproteínas/metabolismo , Factores de Empalme Serina-Arginina , Azida Sódica/farmacología , Transfección
15.
Nat Commun ; 12(1): 6963, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-34845225

RESUMEN

Within the bone marrow microenvironment, endothelial cells (EC) exert important functions. Arterial EC support hematopoiesis while H-type capillaries induce bone formation. Here, we show that BM sinusoidal EC (BM-SEC) actively control erythropoiesis. Mice with stabilized ß-catenin in BM-SEC (Ctnnb1OE-SEC) generated by using a BM-SEC-restricted Cre mouse line (Stab2-iCreF3) develop fatal anemia. While activation of Wnt-signaling in BM-SEC causes an increase in erythroblast subsets (PII-PIV), mature erythroid cells (PV) are reduced indicating impairment of terminal erythroid differentiation/reticulocyte maturation. Transplantation of Ctnnb1OE-SEC hematopoietic stem cells into wildtype recipients confirms lethal anemia to be caused by cell-extrinsic, endothelial-mediated effects. Ctnnb1OE-SEC BM-SEC reveal aberrant sinusoidal differentiation with altered EC gene expression and perisinusoidal ECM deposition and angiocrine dysregulation with de novo endothelial expression of FGF23 and DKK2, elevated in anemia and involved in vascular stabilization, respectively. Our study demonstrates that BM-SEC play an important role in the bone marrow microenvironment in health and disease.


Asunto(s)
Anemia/genética , Médula Ósea/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Endotelio Vascular/metabolismo , Eritroblastos/metabolismo , Eritropoyesis/genética , beta Catenina/genética , Anemia/metabolismo , Anemia/mortalidad , Anemia/patología , Animales , Médula Ósea/irrigación sanguínea , Capilares/citología , Capilares/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Diferenciación Celular , Células Endoteliales/clasificación , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Eritroblastos/clasificación , Eritroblastos/citología , Femenino , Factor-23 de Crecimiento de Fibroblastos/genética , Factor-23 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Integrasas/genética , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Transgénicos , Osteogénesis , Reticulocitos/citología , Reticulocitos/metabolismo , Análisis de Supervivencia , Vía de Señalización Wnt , beta Catenina/metabolismo
16.
Viruses ; 12(10)2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096814

RESUMEN

H-1 protoparvovirus (H-1PV) is a self-propagating virus that is non-pathogenic in humans and has oncolytic and oncosuppressive activities. H-1PV is the first member of the Parvoviridae family to undergo clinical testing as an anticancer agent. Results from clinical trials in patients with glioblastoma or pancreatic carcinoma show that virus treatment is safe, well-tolerated and associated with first signs of efficacy. Characterisation of the H-1PV life cycle may help to improve its efficacy and clinical outcome. In this study, we investigated the entry route of H-1PV in cervical carcinoma HeLa and glioma NCH125 cell lines. Using electron and confocal microscopy, we detected H-1PV particles within clathrin-coated pits and vesicles, providing evidence that the virus uses clathrin-mediated endocytosis for cell entry. In agreement with these results, we found that blocking clathrin-mediated endocytosis using specific inhibitors or small interfering RNA-mediated knockdown of its key regulator, AP2M1, markedly reduced H-1PV entry. By contrast, we found no evidence of viral entry through caveolae-mediated endocytosis. We also show that H-1PV entry is dependent on dynamin, while viral trafficking occurs from early to late endosomes, with acidic pH necessary for a productive infection. This is the first study that characterises the cell entry pathways of oncolytic H-1PV.


Asunto(s)
Clatrina/fisiología , Endocitosis , Parvovirus H-1 , Neoplasias/terapia , Viroterapia Oncolítica , Caveolas/fisiología , Línea Celular Tumoral , Dinaminas/fisiología , Humanos , Concentración de Iones de Hidrógeno , Internalización del Virus
17.
ChemSusChem ; 13(3): 529-538, 2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-31738480

RESUMEN

The addition of Si compounds to graphite anodes has become an attractive way of increasing the practical specific energies in Li-ion cells. Previous studies involving Si/C anodes lacked direct insight into the processes occurring in full cells during low-temperature operation. In this study, a powerful combination of operando neutron diffraction, electrochemical tests, and post-mortem analysis is used for the investigation of Li-ion cells. 18650-type cylindrical cells in two different aging states are investigated by operando neutron diffraction. The experiments reveal deep insights and important trends in low-temperature charging mechanisms involving intercalation, alloying, Li metal deposition, and relaxation processes as a function of charging C-rates and temperatures. Additionally, the main aging mechanism caused by long-term cycling and interesting synergistic effects of Si and graphite are elucidated.

18.
Nat Commun ; 11(1): 3281, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32612175

RESUMEN

Amyloid fibrils result from the aggregation of host cell-encoded proteins, many giving rise to specific human illnesses such as Alzheimer's disease. Here we show that the major virulence factor of Rift Valley fever virus, the protein NSs, forms filamentous structures in the brain of mice and affects mortality. NSs assembles into nuclear and cytosolic disulfide bond-dependent fibrillary aggregates in infected cells. NSs structural arrangements exhibit characteristics typical for amyloids, such as an ultrastructure of 12 nm-width fibrils, a strong detergent resistance, and interactions with the amyloid-binding dye Thioflavin-S. The assembly dynamics of viral amyloid-like fibrils can be visualized in real-time. They form spontaneously and grow in an amyloid fashion within 5 hours. Together, our results demonstrate that viruses can encode amyloid-like fibril-forming proteins and have strong implications for future research on amyloid aggregation and toxicity in general.


Asunto(s)
Amiloide/metabolismo , Proteínas Amiloidogénicas/metabolismo , Fiebre del Valle del Rift/metabolismo , Virus de la Fiebre del Valle del Rift/metabolismo , Proteínas no Estructurales Virales/metabolismo , Amiloide/química , Amiloide/ultraestructura , Proteínas Amiloidogénicas/química , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Chlorocebus aethiops , Células HeLa , Humanos , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Agregación Patológica de Proteínas/metabolismo , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/patogenicidad , Células Vero , Proteínas no Estructurales Virales/química , Virulencia , Factores de Virulencia
19.
Biochim Biophys Acta ; 1783(11): 2100-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18723053

RESUMEN

It is well established, that biochemical reactions are dependent on pH, ionic strength, temperature and the concentration of reactants. However, the steric repulsion among bulky components of biological systems also affect biochemical behavior: The 'excluded volume effect of macromolecular crowding' drives bulky components into structurally compact organizations, increases their thermodynamic activities and slows down diffusion. The very special composition of the cell nucleus, which is packed with high-molecular chromatin, ribonucleo-particles and associated proteins, suggests that crowding-effects are part of nuclear functionality. Realizing that many nuclear processes, notably gene transcription, hnRNA splicing and DNA replication, use macromolecular machines, and taking into account that macromolecular crowding provides a cooperative momentum for the assembly of macromolecular complexes, we here elaborate why macromolecular crowding may be functionally important in supporting the statistical significance of nuclear activities.


Asunto(s)
Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Sustancias Macromoleculares/metabolismo , Línea Celular , Núcleo Celular/genética , Humanos , Sustancias Macromoleculares/química , Matriz Nuclear/metabolismo , Matriz Nuclear/ultraestructura , Proteínas Nucleares/metabolismo , Termodinámica
20.
Nat Commun ; 10(1): 4851, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31649258

RESUMEN

Maintenance of cellular proteostasis is achieved by a multi-layered quality control network, which counteracts the accumulation of misfolded proteins by refolding and degradation pathways. The organized sequestration of misfolded proteins, actively promoted by cellular sequestrases, represents a third strategy of quality control. Here we determine the role of sequestration within the proteostasis network in Saccharomyces cerevisiae and the mechanism by which it occurs. The Hsp42 and Btn2 sequestrases are functionally intertwined with the refolding activity of the Hsp70 system. Sequestration of misfolded proteins by Hsp42 and Btn2 prevents proteostasis collapse and viability loss in cells with limited Hsp70 capacity, likely by shielding Hsp70 from misfolded protein overload. Btn2 has chaperone and sequestrase activity and shares features with small heat shock proteins. During stress recovery Btn2 recruits the Hsp70-Hsp104 disaggregase by directly interacting with the Hsp70 co-chaperone Sis1, thereby shunting sequestered proteins to the refolding pathway.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteostasis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Replegamiento Proteico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA