Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Trends Cell Biol ; 34(3): 198-210, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37474375

RESUMEN

Lysosomes degrade and recycle macromolecules that are delivered through the biosynthetic, endocytic, and autophagic routes. Hydrolysis of the different classes of macromolecules is catalyzed by about 70 soluble enzymes that are transported from the Golgi apparatus to lysosomes in a mannose 6-phosphate (M6P)-dependent process. The molecular machinery that generates M6P tags for receptor-mediated targeting of lysosomal enzymes was thought to be understood in detail. However, recent studies on the M6P pathway have identified a previously uncharacterized core component, yielded structural insights in known components, and uncovered functions in various human diseases. Here we review molecular mechanisms of lysosomal enzyme trafficking and discuss its relevance for rare lysosomal disorders, cancer, and viral infection.


Asunto(s)
Proteínas Portadoras , Lisosomas , Humanos , Proteínas Portadoras/metabolismo , Lisosomas/metabolismo
2.
Cell Death Dis ; 13(11): 969, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36400754

RESUMEN

Multiple myeloma (MM) causes approximately 20% of deaths from blood cancers. Notwithstanding significant therapeutic progress, such as with proteasome inhibitors (PIs), MM remains incurable due to the development of resistance. mTORC1 is a key metabolic regulator, which frequently becomes dysregulated in cancer. While mTORC1 inhibitors reduce MM viability and synergize with other therapies in vitro, clinically, mTORC1 inhibitors are not effective for MM. Here we show that the inactivation of mTORC1 is an intrinsic response of MM to PI treatment. Genetically enforced hyperactivation of mTORC1 in MM was sufficient to compromise tumorigenicity in mice. In vitro, mTORC1-hyperactivated MM cells gained sensitivity to PIs and hypoxia. This was accompanied by increased mitochondrial stress and activation of the eIF2α kinase HRI, which initiates the integrated stress response. Deletion of HRI elevated the toxicity of PIs in wt and mTORC1-activated MM. Finally, we identified the drug PMA as a robust inducer of mTORC1 activity, which synergized with PIs in inducing MM cell death. These results help explain the clinical inefficacy of mTORC1 inhibitors in MM. Our data implicate mTORC1 induction and/or HRI inhibition as pharmacological strategies to enhance MM therapy by PIs.


Asunto(s)
Mieloma Múltiple , Inhibidores de Proteasoma , Animales , Ratones , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Transducción de Señal , eIF-2 Quinasa/metabolismo
3.
Science ; 378(6615): eabn5637, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36074822

RESUMEN

Mammalian cells can generate amino acids through macropinocytosis and lysosomal breakdown of extracellular proteins, which is exploited by cancer cells to grow in nutrient-poor tumors. Through genetic screens in defined nutrient conditions, we characterized LYSET, a transmembrane protein (TMEM251) selectively required when cells consume extracellular proteins. LYSET was found to associate in the Golgi with GlcNAc-1-phosphotransferase, which targets catabolic enzymes to lysosomes through mannose-6-phosphate modification. Without LYSET, GlcNAc-1-phosphotransferase was unstable because of a hydrophilic transmembrane domain. Consequently, LYSET-deficient cells were depleted of lysosomal enzymes and impaired in turnover of macropinocytic and autophagic cargoes. Thus, LYSET represents a core component of the lysosomal enzyme trafficking pathway, underlies the pathomechanism for hereditary lysosomal storage disorders, and may represent a target to suppress metabolic adaptations in cancer.


Asunto(s)
Aparato de Golgi , Enfermedades por Almacenamiento Lisosomal , Lisosomas , Proteínas , Animales , Aparato de Golgi/metabolismo , Humanos , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/metabolismo , Lisosomas/metabolismo , Ratones , Transporte de Proteínas , Proteínas/genética , Proteínas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo
4.
Nat Commun ; 13(1): 4848, 2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-35977928

RESUMEN

Mammalian cells can acquire exogenous amino acids through endocytosis and lysosomal catabolism of extracellular proteins. In amino acid-replete environments, nutritional utilization of extracellular proteins is suppressed by the amino acid sensor mechanistic target of rapamycin complex 1 (mTORC1) through an unknown process. Here, we show that mTORC1 blocks lysosomal degradation of extracellular proteins by suppressing V-ATPase-mediated acidification of lysosomes. When mTORC1 is active, peripheral V-ATPase V1 domains reside in the cytosol where they are stabilized by association with the chaperonin TRiC. Consequently, most lysosomes display low catabolic activity. When mTORC1 activity declines, V-ATPase V1 domains move to membrane-integral V-ATPase Vo domains at lysosomes to assemble active proton pumps. The resulting drop in luminal pH increases protease activity and degradation of protein contents throughout the lysosomal population. These results uncover a principle by which cells rapidly respond to changes in their nutrient environment by mobilizing the latent catabolic capacity of lysosomes.


Asunto(s)
ATPasas de Translocación de Protón Vacuolares , Aminoácidos/metabolismo , Animales , Endocitosis , Lisosomas/metabolismo , Mamíferos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
5.
Cancer Res ; 82(18): 3263-3274, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-35857801

RESUMEN

The mTOR is a key regulator of cell growth that integrates growth factor signaling and nutrient availability and is a downstream effector of oncogenic receptor tyrosine kinases (RTK) and PI3K/Akt signaling. Thus, activating mTOR mutations would be expected to enhance growth in many tumor types. However, tumor sequencing data have shown that mTOR mutations are enriched only in renal clear cell carcinoma, a clinically hypervascular tumor unlikely to be constrained by nutrient availability. To further define this cancer-type-specific restriction, we studied activating mutations in mTOR. All mTOR mutants tested enhanced growth in a cell-type agnostic manner under nutrient-replete conditions but were detrimental to cell survival in nutrient-poor conditions. Consistently, analysis of tumor data demonstrated that oncogenic mutations in the nutrient-sensing arm of the mTOR pathway display a similar phenotype and were exceedingly rare in human cancers of all types. Together, these data suggest that maintaining the ability to turn off mTOR signaling in response to changing nutrient availability is retained in most naturally occurring tumors. SIGNIFICANCE: This study suggests that cells need to inactivate mTOR to survive nutrient stress, which could explain the rarity of mTOR mutations and the limited clinical activity of mTOR inhibitors in cancer.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Mutación , Nutrientes , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Tirosina/genética
6.
Subcell Biochem ; 98: 143-167, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35378707

RESUMEN

Macropinocytosis is an evolutionarily conserved endocytic pathway that mediates non-selective uptake of extracellular fluid in bulk. Macropinocytosis is initiated by localized polymerization of the actin cytoskeleton, which generates plasma membrane protrusions that enclose part of the environment into large endocytic vesicles. From amoebae to mammalian cells, the actin dynamics that drive macropinosome formation are regulated by a conserved set of intracellular signaling proteins including Ras superfamily GTPases and PI3-kinases. In mammalian cells, multiple upstream signaling pathways control activity of these core regulators in response to cell-extrinsic and cell-intrinsic stimuli. Growth factor signaling pathways play a central role in macropinocytosis induction. In addition, an increasing number of functionally diverse processes has been identified as macropinocytosis regulators, including several nutrient-sensing and developmental signaling pathways. Many of these signaling pathways have proto-oncogenic properties, and their dysregulation drives the high macropinocytic activity that is commonly observed in cancer cells. These regulatory principles illustrate how macropinocytosis is controlled by complex upstream inputs to exert diverse cellular functions in physiological and pathological contexts.


Asunto(s)
Pinocitosis , Transducción de Señal , Citoesqueleto de Actina , Animales , Endosomas , Péptidos y Proteínas de Señalización Intercelular , Mamíferos , Pinocitosis/fisiología , Transducción de Señal/fisiología
7.
EMBO J ; 40(8): e108070, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33763859

RESUMEN

Glycosphingolipids are a structurally diverse class of lipids that regulate plasma membrane protein function. Rizzo et al (2021) now show that GOLPH3 promotes intra-Golgi transport of several enzymes that function at branching points of sphingolipid biosynthesis. By regulating the cellular sphingolipidome, GOLPH3 promotes growth factor signaling and cell proliferation, which may explain its oncogenic properties.


Asunto(s)
Aparato de Golgi , Proteínas de la Membrana , Proliferación Celular , Glicoesfingolípidos , Proteínas de la Membrana/genética , Transducción de Señal
9.
Cell ; 184(3): 655-674.e27, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33497611

RESUMEN

Ras GTPase-activating protein-binding proteins 1 and 2 (G3BP1 and G3BP2, respectively) are widely recognized as core components of stress granules (SGs). We report that G3BPs reside at the cytoplasmic surface of lysosomes. They act in a non-redundant manner to anchor the tuberous sclerosis complex (TSC) protein complex to lysosomes and suppress activation of the metabolic master regulator mechanistic target of rapamycin complex 1 (mTORC1) by amino acids and insulin. Like the TSC complex, G3BP1 deficiency elicits phenotypes related to mTORC1 hyperactivity. In the context of tumors, low G3BP1 levels enhance mTORC1-driven breast cancer cell motility and correlate with adverse outcomes in patients. Furthermore, G3bp1 inhibition in zebrafish disturbs neuronal development and function, leading to white matter heterotopia and neuronal hyperactivity. Thus, G3BPs are not only core components of SGs but also a key element of lysosomal TSC-mTORC1 signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , ADN Helicasas/metabolismo , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Esclerosis Tuberosa/metabolismo , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Gránulos Citoplasmáticos/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , ADN Helicasas/química , Evolución Molecular , Femenino , Humanos , Insulina/farmacología , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenotipo , Proteínas de Unión a Poli-ADP-Ribosa/química , ARN Helicasas/química , Proteínas con Motivos de Reconocimiento de ARN/química , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Pez Cebra/metabolismo
10.
Development ; 147(24)2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33355243

RESUMEN

Lipids exert diverse functions in living organisms. They form cellular membranes, store and transport energy and play signalling roles. Some lipid species function in all of these processes, making them ideal candidates to coordinate metabolism with cellular homeostasis and animal development. This theme was central to Suzanne Eaton's research in the fruit fly, Drosophila Here, we discuss her work on membrane lipid homeostasis in changing environments and on functions for lipids in the Hedgehog signalling pathway. We further highlight lipoproteins as inter-organ carriers of lipids and lipid-linked morphogens, which communicate dietary and developmental signals throughout the organism.


Asunto(s)
Proteínas Hedgehog/genética , Metabolismo de los Lípidos/genética , Lípidos/genética , Lipoproteínas/genética , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Proteínas Hedgehog/metabolismo , Homeostasis/genética , Lipoproteínas/metabolismo , Transducción de Señal/genética
11.
Genes Dev ; 34(19-20): 1345-1358, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32912902

RESUMEN

The uptake of macromolecules and cellular debris through macropinocytosis has emerged as an important nutrient acquisition strategy of cancer cells. Genetic alterations commonly found in human cancers (e.g. mutations in KRAS or loss of PTEN) have been shown to increase macropinocytosis. To identify additional effectors that enable cell growth dependent on the uptake of extracellular proteins, pancreatic ductal adenocarcinoma (PDA) cells were selected for growth in medium where extracellular albumin was the obligate source of the essential amino acid leucine. Analysis of global changes in chromatin availability and gene expression revealed that PDA cells selected under these conditions exhibited elevated activity of the transcriptional activators Yap/Taz. Knockout of Yap/Taz prevented growth of PDA cells in leucine-deficient medium, but not in complete medium. Furthermore, constitutively active forms of Yap or Taz were sufficient to stimulate macropinocytosis of extracellular protein. In addition to promoting the uptake of plasma proteins, Yap/Taz also promoted the scavenging of apoptotic cell bodies and necrotic debris by PDA cells. The Yap/Taz transcriptional target Axl was found to be essential for cell growth dependent on the uptake of dead cells and cell debris. Together, these studies suggest that the Hippo pathway effectors Yap and Taz are important transcriptional regulators of endocytic nutrient uptake.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Nutrientes/metabolismo , Pinocitosis/fisiología , Factores de Transcripción/metabolismo , Aciltransferasas , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Espacio Extracelular/metabolismo , Humanos , Ratones , Proteínas Señalizadoras YAP
12.
J Cell Biol ; 219(3)2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-32040547

RESUMEN

Cell proliferation exerts a high demand on protein synthesis, yet the mechanisms coupling the two processes are not fully understood. A kinase and phosphatase screen for activators of translation, based on the formation of stress granules in human cells, revealed cell cycle-associated kinases as major candidates. CDK1 was identified as a positive regulator of global translation, and cell synchronization experiments showed that this is an extramitotic function of CDK1. Different pathways including eIF2α, 4EBP, and S6K1 signaling contribute to controlling global translation downstream of CDK1. Moreover, Ribo-Seq analysis uncovered that CDK1 exerts a particularly strong effect on the translation of 5'TOP mRNAs, which includes mRNAs encoding ribosomal proteins and several translation factors. This effect requires the 5'TOP mRNA-binding protein LARP1, concurrent to our finding that LARP1 phosphorylation is strongly dependent on CDK1. Thus, CDK1 provides a direct means to couple cell proliferation with biosynthesis of the translation machinery and the rate of protein synthesis.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proliferación Celular , Neoplasias del Cuello Uterino/enzimología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Proteína Quinasa CDC2/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Femenino , Fibroblastos/enzimología , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Cinética , Ratones Endogámicos C57BL , Fosforilación , Biosíntesis de Proteínas , Secuencia de Oligopirimidina en la Región 5' Terminal del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Antígeno SS-B
13.
Philos Trans R Soc Lond B Biol Sci ; 374(1765): 20180285, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30967008

RESUMEN

Macropinocytosis is an evolutionarily conserved form of endocytosis that mediates non-selective uptake of extracellular fluid and the solutes contained therein. In mammalian cells, macropinocytosis is initiated by growth factor-mediated activation of the Ras and PI3-kinase signalling pathways. In malignant cells, oncogenic activation of growth factor signalling sustains macropinocytosis cell autonomously. Recent studies of cancer metabolism, discussed here, have begun to define a role for macropinocytosis as a nutrient uptake route. Macropinocytic cancer cells ingest macromolecules in bulk and break them down in the lysosome to support metabolism and macromolecular synthesis. Thereby, macropinocytosis allows cells to tap into the copious nutrient stores of extracellular macromolecules when canonical nutrients are scarce. These findings demonstrate that macropinocytosis promotes metabolic flexibility and resilience, which enables cancer cells to survive and grow in nutrient-poor environments. Implications for physiological roles of growth factor-stimulated macropinocytosis in cell metabolism and its relationship with other nutrient uptake pathways are considered. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Pinocitosis/fisiología , Transducción de Señal/fisiología , Animales , Humanos
14.
Cancer Res ; 78(16): 4658-4670, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29871936

RESUMEN

Macropinocytosis has emerged as an important pathway of protein acquisition in cancer cells, particularly in tumors with activated Ras such as pancreatic and colon cancer. Macropinocytosis is also the route of entry of Bacillus Calmette-Guerin (BCG) and other microbial therapies of cancer. Despite this important role in tumor biology and therapy, the full mechanisms by which cancer cells can activate macropinocytosis remain incompletely defined. Using BCG uptake to assay macropinocytosis, we executed a genome-wide shRNA screen for macropinocytosis activators and identified Wnt pathway activation as a strong driver of macropinocytosis. Wnt-driven macropinocytosis was downstream of the ß-catenin-dependent canonical Wnt pathway, was PAK1 dependent, and supported albumin-dependent growth in Ras-WT cells. In cells with activated Ras-dependent macropinocytosis, pharmacologic or genetic inhibition of Wnt signaling suppressed macropinocytosis. In a mouse model of Wnt-driven colonic hyperplasia via APC silencing, Wnt-activated macropinocytosis stimulated uptake of luminal microbiota, a process reversed by topical pharmacologic inhibition of macropinocytosis. Our findings indicate that Wnt pathway activation drives macropinocytosis in cancer, and its inhibition could provide a therapeutic vulnerability in Wnt-driven intestinal polyposis and cancers with Wnt activation.Significance: The Wnt pathway drives macropinocytosis in cancer cells, thereby contributing to cancer growth in nutrient-deficient conditions and, in the context of colon cancer, to the early phases of oncogenesis. Cancer Res; 78(16); 4658-70. ©2018 AACR.


Asunto(s)
Neoplasias/tratamiento farmacológico , Pinocitosis/genética , Vía de Señalización Wnt/genética , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Silenciador del Gen , Genoma Humano/genética , Humanos , Ratones , Mycobacterium bovis/genética , Neoplasias/genética , Neoplasias/patología , ARN Interferente Pequeño/genética , beta Catenina/genética
15.
Proc Natl Acad Sci U S A ; 114(41): E8628-E8636, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28973876

RESUMEN

Ras-transformed cells can grow in amino acid-poor environments by recovering amino acids through macropinocytosis and lysosomal catabolism of extracellular proteins. However, when studying nontransformed fibroblasts, we found that Ras GTPases are dispensable for growth-factor-stimulated macropinocytosis and lysosomal catabolism of extracellular proteins. Instead, we establish a critical role for phosphatidylinositol 3-kinase (PI3-kinase) signaling in cell proliferation that is supported by protein macropinocytosis. Downstream of PI3-kinase, distinct effectors have opposing roles in regulating uptake and catabolism of extracellular proteins. Rac1 and PLC are required for nutritional use of extracellular proteins. In contrast, Akt suppresses lysosomal catabolism of ingested proteins when free amino acids are abundant. The interplay between these pathways allows cells with oncogenic PIK3CA mutations or PTEN deletion to grow using diverse amino acid sources. Thus, the prevalence of PI3-kinase and PTEN mutations in cancer may result in part because they allow cells to cope with fluctuating nutrient availability.


Asunto(s)
Aminoácidos/metabolismo , Proliferación Celular , Fibroblastos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Pinocitosis/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Células Cultivadas , Fibroblastos/citología , Ratones , Fosforilación , Transducción de Señal , Proteínas ras/metabolismo
16.
Nature ; 546(7657): 234-242, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28593971

RESUMEN

Mammalian cells are surrounded by diverse nutrients, such as glucose, amino acids, various macromolecules and micronutrients, which they can import through transmembrane transporters and endolysosomal pathways. By using different nutrient sources, cells gain metabolic flexibility to survive periods of starvation. Quiescent cells take up sufficient nutrients to sustain homeostasis. However, proliferating cells depend on growth-factor-induced increases in nutrient uptake to support biomass formation. Here, we review cellular nutrient acquisition strategies and their regulation by growth factors and cell-intrinsic nutrient sensors. We also discuss how oncogenes and tumour suppressors promote nutrient uptake and thereby support the survival and growth of cancer cells.


Asunto(s)
Células/metabolismo , Animales , Proliferación Celular , Células/citología , Endocitosis , Glucosa/metabolismo , Glutamina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lisosomas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Pinocitosis , Proteínas/química , Proteínas/metabolismo , Microambiente Tumoral
17.
Dev Cell ; 38(5): 536-47, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27623384

RESUMEN

The scavenging of extracellular macromolecules by engulfment can sustain cell growth in a nutrient-depleted environment. Engulfed macromolecules are contained within vacuoles that are targeted for lysosome fusion to initiate degradation and nutrient export. We have shown that vacuoles containing engulfed material undergo mTORC1-dependent fission that redistributes degraded cargo back into the endosomal network. Here we identify the lipid kinase PIKfyve as a regulator of an alternative pathway that distributes engulfed contents in support of intracellular macromolecular synthesis during macropinocytosis, entosis, and phagocytosis. We find that PIKfyve regulates vacuole size in part through its downstream effector, the cationic transporter TRPML1. Furthermore, PIKfyve promotes recovery of nutrients from vacuoles, suggesting a potential link between PIKfyve activity and lysosomal nutrient export. During nutrient depletion, PIKfyve activity protects Ras-mutant cells from starvation-induced cell death and supports their proliferation. These data identify PIKfyve as a critical regulator of vacuole maturation and nutrient recovery during engulfment.


Asunto(s)
Fosfatidilinositol 3-Quinasas/genética , Canales de Potencial de Receptor Transitorio/genética , Vacuolas/genética , Animales , Caenorhabditis elegans , Línea Celular Tumoral , Endosomas/genética , Endosomas/metabolismo , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Fagocitosis/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inanición , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Vacuolas/metabolismo
18.
Cell ; 166(4): 963-976, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27477511

RESUMEN

Pancreatic cancer is a deadly malignancy that lacks effective therapeutics. We previously reported that oncogenic Kras induced the redox master regulator Nfe2l2/Nrf2 to stimulate pancreatic and lung cancer initiation. Here, we show that NRF2 is necessary to maintain pancreatic cancer proliferation by regulating mRNA translation. Specifically, loss of NRF2 led to defects in autocrine epidermal growth factor receptor (EGFR) signaling and oxidation of specific translational regulatory proteins, resulting in impaired cap-dependent and cap-independent mRNA translation in pancreatic cancer cells. Combined targeting of the EGFR effector AKT and the glutathione antioxidant pathway mimicked Nrf2 ablation to potently inhibit pancreatic cancer ex vivo and in vivo, representing a promising synthetic lethal strategy for treating the disease.


Asunto(s)
Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/metabolismo , Biosíntesis de Proteínas , Animales , Comunicación Autocrina , Cisteína/metabolismo , Glutatión/metabolismo , Humanos , Ratones , Organoides/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal
19.
Artículo en Inglés | MEDLINE | ID: mdl-28396524

RESUMEN

A key feature of multicellular life is the sharing of nutritional resources by all cells of the organism. In animals, the allocation of nutrients to individual cells is not determined in a cell-autonomous fashion. Instead, growth factor and hormonal signaling pathways have evolved to regulate cellular nutrient uptake, which prevents individual cells from parasitizing the organism's metabolic reserves. Cancer is a condition where the highly ordered regulation of nutrient distribution is disrupted. During carcinogenesis, transformed cells acquire mutations in signaling pathways that render nutrient uptake cell-autonomous. A deeper understanding of how the acquisition of potential nutrients is regulated may help develop improved approaches to cancer therapy.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Transducción de Señal/fisiología , Animales , Alimentos , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Mutación/genética , Neoplasias/genética
20.
Genes Dev ; 29(22): 2331-6, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26543160

RESUMEN

Mammalian cells possess two amino acid-sensing kinases: general control nonderepressible 2 (GCN2) and mechanistic target of rapamycin complex 1 (mTORC1). Their combined effects orchestrate cellular adaptation to amino acid levels, but how their activities are coordinated remains poorly understood. Here, we demonstrate an important link between GCN2 and mTORC1 signaling. Upon deprivation of various amino acids, activated GCN2 up-regulates ATF4 to induce expression of the stress response protein Sestrin2, which is required to sustain repression of mTORC1 by blocking its lysosomal localization. Moreover, Sestrin2 induction is necessary for cell survival during glutamine deprivation, indicating that Sestrin2 is a critical effector of GCN2 signaling that regulates amino acid homeostasis through mTORC1 suppression.


Asunto(s)
Aminoácidos/metabolismo , Regulación de la Expresión Génica , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Células HEK293 , Humanos , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Proteínas Nucleares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...