Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 406, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195686

RESUMEN

Tuberous Sclerosis Complex (TSC) is caused by TSC1 or TSC2 mutations, leading to hyperactivation of mechanistic target of rapamycin complex 1 (mTORC1) and lesions  in multiple organs including lung (lymphangioleiomyomatosis) and kidney (angiomyolipoma and renal cell carcinoma). Previously, we found that TFEB is constitutively active in TSC. Here, we generated two mouse models of TSC in which kidney pathology is the primary phenotype. Knockout of TFEB rescues kidney pathology and overall survival, indicating that TFEB is the primary driver of renal disease in TSC. Importantly, increased mTORC1 activity in the TSC2 knockout kidneys is normalized by TFEB knockout. In TSC2-deficient cells, Rheb knockdown or Rapamycin treatment paradoxically increases TFEB phosphorylation at the mTORC1-sites and relocalizes TFEB from nucleus to cytoplasm. In mice, Rapamycin treatment normalizes lysosomal gene expression, similar to TFEB knockout, suggesting that Rapamycin's benefit in TSC is TFEB-dependent. These results change the view of the mechanisms of mTORC1 hyperactivation in TSC and may lead to therapeutic avenues.


Asunto(s)
Neoplasias Renales , Esclerosis Tuberosa , Animales , Ratones , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Noqueados , Sirolimus/farmacología , Esclerosis Tuberosa/genética
2.
iScience ; 25(10): 105185, 2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36238896

RESUMEN

Lipid phosphate phosphatases are a family of enzymes with diverse cellular metabolic functions. Phospholipid phosphatase 6 (PLPP6) is a regulator of cellular polyisoprenyl phosphates; however, its in vivo functions remain to be determined. Here, mouse PLPP6 was characterized to possess similar catalytic properties as the human enzyme. Plpp6 knockout mice (Plpp6 -/- ) were generated and displayed decreased airway allergen sensitization, pointing to a role for PLPP6 in the early events of lung allergic responses. Dendritic cell (DC) responses were investigated and endocytosis of allergen via macropinocytosis was decreased in Plpp6 -/- DCs that had lower cholesterol content. When reversed by cholesterol loading, the DC macropinocytosis defect is corrected. Adoptive transfer of Plpp6 -/- DCs to wild-type mice during sensitization was sufficient to decrease allergen-induced responses. Together, our findings have identified PLPP6 as a pivotal regulator of DC cholesterol content and macropinocytosis, cellular mechanisms that are important for pathologic responses in allergen-induced lung inflammation.

3.
Proc Natl Acad Sci U S A ; 119(28): e2122840119, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35867762

RESUMEN

Chromophobe (Ch) renal cell carcinoma (RCC) arises from the intercalated cell in the distal nephron. There are no proven treatments for metastatic ChRCC. A distinguishing characteristic of ChRCC is strikingly high levels of reduced (GSH) and oxidized (GSSG) glutathione. Here, we demonstrate that ChRCC-derived cells exhibit higher sensitivity to ferroptotic inducers compared with clear-cell RCC. ChRCC-derived cells are critically dependent on cystine via the cystine/glutamate antiporter xCT to maintain high levels of glutathione, making them sensitive to inhibitors of cystine uptake and cyst(e)inase. Gamma-glutamyl transferase 1 (GGT1), a key enzyme in glutathione homeostasis, is markedly suppressed in ChRCC relative to normal kidney. Importantly, GGT1 overexpression inhibits the proliferation of ChRCC cells in vitro and in vivo, suppresses cystine uptake, and decreases levels of GSH and GSSG. Collectively, these data identify ferroptosis as a metabolic vulnerability in ChRCC, providing a potential avenue for targeted therapy for these distinctive tumors.


Asunto(s)
Sistema de Transporte de Aminoácidos y+ , Carcinoma de Células Renales , Cistina , Ferroptosis , Glutatión , Neoplasias Renales , Sistema de Transporte de Aminoácidos y+/metabolismo , Transporte Biológico , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Cistina/metabolismo , Glutatión/metabolismo , Disulfuro de Glutatión/deficiencia , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Terapia Molecular Dirigida , gamma-Glutamiltransferasa/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34544857

RESUMEN

Tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM) are caused by aberrant mechanistic Target of Rapamycin Complex 1 (mTORC1) activation due to loss of either TSC1 or TSC2 Cytokine profiling of TSC2-deficient LAM patient-derived cells revealed striking up-regulation of Interleukin-6 (IL-6). LAM patient plasma contained increased circulating IL-6 compared with healthy controls, and TSC2-deficient cells showed up-regulation of IL-6 transcription and secretion compared to wild-type cells. IL-6 blockade repressed the proliferation and migration of TSC2-deficient cells and reduced oxygen consumption and extracellular acidification. U-13C glucose tracing revealed that IL-6 knockout reduced 3-phosphoserine and serine production in TSC2-deficient cells, implicating IL-6 in de novo serine metabolism. IL-6 knockout reduced expression of phosphoserine aminotransferase 1 (PSAT1), an essential enzyme in serine biosynthesis. Importantly, recombinant IL-6 treatment rescued PSAT1 expression in the TSC2-deficient, IL-6 knockout clones selectively and had no effect on wild-type cells. Treatment with anti-IL-6 (αIL-6) antibody similarly reduced cell proliferation and migration and reduced renal tumors in Tsc2+/- mice while reducing PSAT1 expression. These data reveal a mechanism through which IL-6 regulates serine biosynthesis, with potential relevance to the therapy of tumors with mTORC1 hyperactivity.


Asunto(s)
Interleucina-6/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Serina/metabolismo , Transaminasas/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Interleucina-6/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transaminasas/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/fisiología
5.
J Vis Exp ; (174)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34398147

RESUMEN

Macropinocytosis is a highly conserved, actin-dependent endocytic process that allows the uptake of extracellular material, including proteins and lipids. In proliferating cells, macropinocytosis can deliver extracellular nutrients to the lysosome, processed into critical macromolecule building blocks. Recent studies have highlighted the dependence of multiple cancers on macropinocytosis, including breast, colorectal and pancreatic cancer. Ras mutations are thought to be the driver events behind macropinocytosis initiation, leading to the activation of cellular anabolic processes via the mTORC1 signaling pathway. Interestingly, mTORC1 can also be activated by macropinocytosis independently of Ras. Therefore, macropinocytosis represents a metabolic vulnerability that can be leveraged to target macropinocytic tumors by limiting their access to nutrients therapeutically. In Tuberous Sclerosis Complex (TSC) and Lymphangioleiomyomatosis (LAM), mTORC1-hyperactivation leads to enhanced macropinocytosis and metabolic reprogramming. Here, we describe a flow cytometry-based protocol to assess macropinocytosis in mammalian cells quantitatively. TSC2-deficient MEFs are employed, which exhibit aberrant activation of mTORC1 and have been shown to have increased macropinocytosis compared to TSC2-expressing cells. Cells treated with pharmacologic inhibitors of macropinocytosis are incubated with fluorescently labeled, lysine-fixable, 70 kDa dextran, or fluorescently labeled bovine serum albumin (BSA) assayed by flow cytometry. To date, robust image-based techniques have been developed to quantitatively assess macropinocytosis in tumor cells in vitro and in vivo. This analysis provides a quantitative assessment of macropinocytosis in multiple experimental conditions and complements existing image-based techniques.


Asunto(s)
Linfangioleiomiomatosis , Pinocitosis , Animales , Citometría de Flujo , Lisosomas , Diana Mecanicista del Complejo 1 de la Rapamicina
6.
EMBO Mol Med ; 13(9): e13929, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34378323

RESUMEN

Inhibition of mTOR is the standard of care for lymphangioleiomyomatosis (LAM). However, this therapy has variable tolerability and some patients show progressive decline of lung function despite treatment. LAM diagnosis and monitoring can also be challenging due to the heterogeneity of symptoms and insufficiency of non-invasive tests. Here, we propose monoamine-derived biomarkers that provide preclinical evidence for novel therapeutic approaches. The major histamine-derived metabolite methylimidazoleacetic acid (MIAA) is relatively more abundant in LAM plasma, and MIAA values are independent of VEGF-D. Higher levels of histamine are associated with poorer lung function and greater disease burden. Molecular and cellular analyses, and metabolic profiling confirmed active histamine signaling and metabolism. LAM tumorigenesis is reduced using approved drugs targeting monoamine oxidases A/B (clorgyline and rasagiline) or histamine H1 receptor (loratadine), and loratadine synergizes with rapamycin. Depletion of Maoa or Hrh1 expression, and administration of an L-histidine analog, or a low L-histidine diet, also reduce LAM tumorigenesis. These findings extend our knowledge of LAM biology and suggest possible ways of improving disease management.


Asunto(s)
Neoplasias Pulmonares , Linfangioleiomiomatosis , Biomarcadores , Histamina , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Linfangioleiomiomatosis/tratamiento farmacológico , Transducción de Señal
7.
Nat Commun ; 12(1): 4245, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253722

RESUMEN

Tuberous Sclerosis Complex (TSC) is caused by TSC1 or TSC2 mutations, resulting in hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). Transcription factor EB (TFEB), a master regulator of lysosome biogenesis, is negatively regulated by mTORC1 through a RAG GTPase-dependent phosphorylation. Here we show that lysosomal biogenesis is increased in TSC-associated renal tumors, pulmonary lymphangioleiomyomatosis, kidneys from Tsc2+/- mice, and TSC1/2-deficient cells via a TFEB-dependent mechanism. Interestingly, in TSC1/2-deficient cells, TFEB is hypo-phosphorylated at mTORC1-dependent sites, indicating that mTORC1 is unable to phosphorylate TFEB in the absence of the TSC1/2 complex. Importantly, overexpression of folliculin (FLCN), a GTPase activating protein for RAGC, increases TFEB phosphorylation at the mTORC1 sites in TSC2-deficient cells. Overexpression of constitutively active RAGC is sufficient to relocalize TFEB to the cytoplasm. These findings establish the TSC proteins as critical regulators of lysosomal biogenesis via TFEB and RAGC and identify TFEB as a driver of the proliferation of TSC2-deficient cells.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Lisosomas/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Biogénesis de Organelos , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Carcinoma de Células Renales/patología , Núcleo Celular/metabolismo , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Neoplasias Renales/patología , Lisosomas/ultraestructura , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosforilación , Fosfoserina/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Transcripción Genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/deficiencia , Proteínas Supresoras de Tumor/metabolismo
8.
Mol Cancer Res ; 19(8): 1389-1397, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33888601

RESUMEN

Tuberous sclerosis complex (TSC) is caused by mutations of either the TSC1 or TSC2 tumor suppressor gene. TSC causes tumors of the brain, heart, kidney, skin and lymphangioleiomyomatosis (LAM). Here we report that the TSC2 protein physically binds to high-density lipoprotein binding protein (HDLBP), also called vigilin, a core stress granule (SG) protein, and that TSC2 localizes to SGs. SGs contain mRNAs and translation initiation complexes, and regulate gene expression by sequestering specific transcripts, thereby serving a cytoprotective role. TSC2 has never before been shown to localize to SGs and knocking down vigilin impacts SG translocation of TSC2. TSC2-deficient cells showed a striking increase in the number of SGs after thermal shock and arsenite treatment relative to Tsc2-expressing cells. Our findings also show that murine kidney lysates from a model of TSC have increased levels of SG components including G3BP1 and Caprin1. G3BP1 and Caprin are elevated in renal angiomyolipomas (a renal tumor common in patients with TSC) compared with control normal kidney. G3BP1 is also elevated in TSC-associated subependymal giant cell astrocytomas. We found that genetic inhibition of G3BP1 inhibits the proliferation of TSC2-deficient cells in vitro. Finally, in a mouse model of TSC, genetic inhibition of SGs suppresses cell growth, suggesting that targeting SGs may have efficacy in the therapy of TSC. IMPLICATIONS: This study demonstrates that TSC2 physically interacts with HDLBP/vigilin, a component of SGs, that TSC2 localizes to SG and that TSC2-deficient cells have more SGs, suggesting that SGs represent a novel therapeutic target in TSC.


Asunto(s)
Proteínas de Unión al ARN/metabolismo , Gránulos de Estrés/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Angiomiolipoma/metabolismo , Angiomiolipoma/patología , Animales , Línea Celular , Línea Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Linfangioleiomiomatosis/metabolismo , Linfangioleiomiomatosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , ARN Mensajero/metabolismo , Gránulos de Estrés/patología , Proteínas Supresoras de Tumor/metabolismo
9.
Cancer Res ; 81(8): 2086-2100, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33593821

RESUMEN

Lymphangioleiomyomatosis is a rare destructive lung disease affecting primarily women and is the primary lung manifestation of tuberous sclerosis complex (TSC). In lymphangioleiomyomatosis, biallelic loss of TSC1/2 leads to hyperactivation of mTORC1 and inhibition of autophagy. To determine how the metabolic vulnerabilities of TSC2-deficient cells can be targeted, we performed a high-throughput screen utilizing the "Repurposing" library at the Broad Institute of MIT and Harvard (Cambridge, MA), with or without the autophagy inhibitor chloroquine. Ritanserin, an inhibitor of diacylglycerol kinase alpha (DGKA), was identified as a selective inhibitor of proliferation of Tsc2-/- mouse embryonic fibroblasts (MEF), with no impact on Tsc2+/+ MEFs. DGKA is a lipid kinase that metabolizes diacylglycerol to phosphatidic acid, a key component of plasma membranes. Phosphatidic acid levels were increased 5-fold in Tsc2-/- MEFs compared with Tsc2+/+ MEFs, and treatment of Tsc2-/- MEFs with ritanserin led to depletion of phosphatidic acid as well as rewiring of phospholipid metabolism. Macropinocytosis is known to be upregulated in TSC2-deficient cells. Ritanserin decreased macropinocytic uptake of albumin, limited the number of lysosomes, and reduced lysosomal activity in Tsc2-/- MEFs. In a mouse model of TSC, ritanserin treatment decreased cyst frequency and volume, and in a mouse model of lymphangioleiomyomatosis, genetic downregulation of DGKA prevented alveolar destruction and airspace enlargement. Collectively, these data indicate that DGKA supports macropinocytosis in TSC2-deficient cells to maintain phospholipid homeostasis and promote proliferation. Targeting macropinocytosis with ritanserin may represent a novel therapeutic approach for the treatment of TSC and lymphangioleiomyomatosis. SIGNIFICANCE: This study identifies macropinocytosis and phospholipid metabolism as novel mechanisms of metabolic homeostasis in mTORC1-hyperactive cells and suggest ritanserin as a novel therapeutic strategy for use in mTORC1-hyperactive tumors, including pancreatic cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2086/F1.large.jpg.


Asunto(s)
Diacilglicerol Quinasa/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Linfangioleiomiomatosis/tratamiento farmacológico , Pinocitosis/efectos de los fármacos , Ritanserina/farmacología , Proteína 2 del Complejo de la Esclerosis Tuberosa/deficiencia , Esclerosis Tuberosa/tratamiento farmacológico , Angiolipoma/genética , Animales , Autofagia/efectos de los fármacos , Proliferación Celular , Cloroquina/farmacología , Diacilglicerol Quinasa/genética , Diacilglicerol Quinasa/metabolismo , Regulación hacia Abajo , Sinergismo Farmacológico , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica , Neoplasias Renales/genética , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis/etiología , Linfangioleiomiomatosis/patología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Desnudos , Nutrientes/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfolípidos/metabolismo , Pinocitosis/fisiología , Esclerosis Tuberosa/complicaciones
10.
Sci Rep ; 8(1): 14161, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30242175

RESUMEN

Tuberous Sclerosis Complex (TSC), a rare genetic disorder with mechanistic target of rapamycin complex 1 (mTORC1) hyperactivation, is characterized by multi-organ hamartomatous benign tumors including brain, skin, kidney, and lung (Lymphangioleiomyomatosis). mTORC1 hyperactivation drives metabolic reprogramming including glucose and glutamine utilization, protein, nucleic acid and lipid synthesis. To investigate the mechanisms of exogenous nutrients uptake in Tsc2-deficient cells, we measured dextran uptake, a polysaccharide internalized via macropinocytosis. Tsc2-deficient cells showed a striking increase in dextran uptake (3-fold, p < 0.0001) relative to Tsc2-expressing cells, which was decreased (3-fold, p < 0.0001) with mTOR inhibitor, Torin1. Pharmacologic and genetic inhibition of the lipid kinase Vps34 markedly abrogated uptake of Dextran in Tsc2-deficient cells. Macropinocytosis was further increased in Tsc2-deficient cells that lack autophagic mechanisms, suggesting that autophagy inhibition leads to dependence on exogenous nutrient uptake in Tsc2-deficient cells. Treatment with a macropinocytosis inhibitor, ethylisopropylamiloride (EIPA), resulted in selective growth inhibition of Atg5-deficient, Tsc2-deficient cells (50%, p < 0.0001). Genetic inhibition of autophagy (Atg5-/- MEFs) sensitized cells with Tsc2 downregulation to the Vps34 inhibitor, SAR405, resulting in growth inhibition (75%, p < 0.0001). Finally, genetic downregulation of Vps34 inhibited tumor growth and increased tumor latency in an in vivo xenograft model of TSC. Our findings show that macropinocytosis is upregulated with Tsc2-deficiency via a Vps34-dependent mechanism to support their anabolic state. The dependence of Tsc2-deficient cells on exogenous nutrients may provide novel approaches for the treatment of TSC.


Asunto(s)
Carcinogénesis/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Pinocitosis/fisiología , Esclerosis Tuberosa/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Autofagia/efectos de los fármacos , Autofagia/fisiología , Proteína 5 Relacionada con la Autofagia/metabolismo , Carcinogénesis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Dextranos/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pinocitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteína 2 del Complejo de la Esclerosis Tuberosa/deficiencia , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
11.
Proc Natl Acad Sci U S A ; 115(27): E6274-E6282, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29891694

RESUMEN

Chromophobe renal cell carcinoma (ChRCC) accounts for 5% of all sporadic renal cancers and can also occur in genetic syndromes including Birt-Hogg-Dube (BHD) and tuberous sclerosis complex (TSC). ChRCC has a distinct accumulation of abnormal mitochondria, accompanied by characteristic chromosomal imbalances and relatively few "driver" mutations. Metabolomic profiling of ChRCC and oncocytomas (benign renal tumors that share pathological features with ChRCC) revealed both similarities and differences between these tumor types, with principal component analysis (PCA) showing a distinct separation. ChRCC have a striking decrease in intermediates of the glutathione salvage pathway (also known as the gamma-glutamyl cycle) compared with adjacent normal kidney, as well as significant changes in glycolytic and pentose phosphate pathway intermediates. We also found that gamma glutamyl transferase 1 (GGT1), the key enzyme of the gamma-glutamyl cycle, is expressed at ∼100-fold lower levels in ChRCC compared with normal kidney, while no change in GGT1 expression was found in clear cell RCC (ccRCC). Significant differences in specific metabolite abundance were found in ChRCC vs. ccRCC, including the oxidative stress marker ophthalmate. Down-regulation of GGT1 enhanced the sensitivity to oxidative stress and treatment with buthionine sulfoximine (BSO), which was associated with changes in glutathione-pathway metabolites. These data indicate that impairment of the glutathione salvage pathway, associated with enhanced oxidative stress, may have key therapeutic implications for this rare tumor type for which there are currently no specific targeted therapies.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/enzimología , Neoplasias Renales/enzimología , Proteínas de Neoplasias/metabolismo , Oligopéptidos/metabolismo , gamma-Glutamiltransferasa/metabolismo , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Femenino , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Proteínas de Neoplasias/genética , Oligopéptidos/genética , Estrés Oxidativo/genética , Transducción de Señal/genética , gamma-Glutamiltransferasa/genética
12.
Oncotarget ; 8(39): 64714-64727, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-29029388

RESUMEN

mTORC1 hyperactivation drives the multi-organ hamartomatous disease tuberous sclerosis complex (TSC). Rapamycin inhibits mTORC1, inducing partial tumor responses; however, the tumors regrow following treatment cessation. We discovered that the oncogenic miRNA, miR-21, is increased in Tsc2-deficient cells and, surprisingly, further increased by rapamycin. To determine the impact of miR-21 in TSC, we inhibited miR-21 in vitro. miR-21 inhibition significantly repressed the tumorigenic potential of Tsc2-deficient cells and increased apoptosis sensitivity. Tsc2-deficient cells' clonogenic and anchorage independent growth were reduced by ∼50% (p<0.01) and ∼75% (p<0.0001), respectively, and combined rapamycin treatment decreased soft agar growth by ∼90% (p<0.0001). miR-21 inhibition also increased sensitivity to apoptosis. Through a network biology-driven integration of RNAseq data, we discovered that miR-21 promotes mitochondrial adaptation and homeostasis in Tsc2-deficient cells. miR-21 inhibition reduced mitochondrial polarization and function in Tsc2-deficient cells, with and without co-treatment with rapamycin. Importantly, miR-21 inhibition limited Tsc2-deficient tumor growth in vivo, reducing tumor size by approximately 3-fold (p<0.0001). When combined with rapamcyin, miR-21 inhibition showed even more striking efficacy, both during treatment and after treatment cessation, with a 4-fold increase in median survival following rapamycin cessation (p=0.0008). We conclude that miR-21 promotes mTORC1-driven tumorigenesis via a mechanism that involves the mitochondria, and that miR-21 is a potential therapeutic target for TSC-associated hamartomas and other mTORC1-driven tumors, with the potential for synergistic efficacy when combined with rapalogs.

13.
Oncotarget ; 8(24): 38099-38112, 2017 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-28498820

RESUMEN

Tuberous sclerosis complex (TSC) is a multisystem disease associated with hyperactive mTORC1. The impact of TSC1/2 deficiency on lysosome-mediated processes is not fully understood. We report here that inhibition of lysosomal function using chloroquine (CQ) upregulates cholesterol homeostasis genes in TSC2-deficient cells. This TSC2-dependent transcriptional signature is associated with increased accumulation and intracellular levels of both total cholesterol and cholesterol esters. Unexpectedly, engaging this CQ-induced cholesterol uptake pathway together with inhibition of de novo cholesterol synthesis allows survival of TSC2-deficient, but not TSC2-expressing cells. The underlying mechanism of TSC2-deficient cell survival is dependent on exogenous cholesterol uptake via LDL-R, and endosomal trafficking mediated by Vps34. Simultaneous inhibition of lysosomal and endosomal trafficking inhibits uptake of esterified cholesterol and cell growth in TSC2-deficient, but not TSC2-expressing cells, highlighting the TSC-dependent lysosome-mediated regulation of cholesterol homeostasis and pointing toward the translational potential of these pathways for the therapy of TSC.


Asunto(s)
Proteínas Portadoras/metabolismo , Colesterol/metabolismo , Lisosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Esclerosis Tuberosa/metabolismo , Línea Celular , Homeostasis/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteína Niemann-Pick C1
14.
Curr Cancer Drug Targets ; 15(8): 684-702, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26452384

RESUMEN

Survival rates of patients with metastatic or recurrent cancers have remained virtually unchanged during the past 30 years. This fact makes the need for new therapeutic options even more urgent. An attractive option would be to target autophagy, an essential quality control process that degrades toxic aggregates, damaged organelles, and signaling proteins, and acts as a tumor suppressor pathway of tumor initiation. Conversely, other fascinating observations suggest that autophagy supports cancer progression, relapse, metastasis, dormancy and resistance to therapy. This review provides an overview of the contradictory roles that autophagy plays in cancer initiation and progression and discusses the promises and challenges of current strategies that target autophagy for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Autofagia , Metástasis de la Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Pruebas en el Punto de Atención , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Metástasis de la Neoplasia/patología , Recurrencia Local de Neoplasia/patología
15.
Am J Respir Cell Mol Biol ; 53(1): 33-41, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25780943

RESUMEN

Lymphangioleiomyomatosis (LAM) is a destructive lung disease affecting women. LAM is caused by mutations in the tuberous sclerosis complex (TSC) genes. The TSC protein complex inhibits the mechanistic/mammalian target of rapamycin complex 1 (mTORC1), which is a master regulator of cellular metabolism. Using mass spectrometry-based lipid profiling, we analyzed plasma from patients with LAM and discovered elevated levels of four lysophosphatidylcholine (LPC) species (C16:0, C18:0, C18:1, and C20:4) compared with those in healthy control women. To investigate whether these lipids are generated in a TSC2-dependent manner, we profiled in vitro preclinical models of TSC/LAM and found significant LPC accumulation in TSC2-deficient cells relative to TSC2-expressing control cells. These lysoglycerophospholipid changes occurred alongside changes in other phospholipid and neutral lipid species. Treatment with rapamycin or torin1 or down-regulation of sterol regulatory element-binding protein (SREBP), a lipogenic transcription factor, did not suppress LPC in TSC2-deficient cells. Inhibition of distinct isoforms of phospholipase A2 decreased the proliferation of TSC2-deficient cells. Collectively, these results demonstrate that TSC2-deficient cells have enhanced choline phospholipid metabolism and reveal a novel function of the TSC proteins in choline lysoglycerophospholipid metabolism, with implications for disease pathogenesis and targeted therapeutic strategies.


Asunto(s)
Metabolismo de los Lípidos , Linfangioleiomiomatosis/metabolismo , Lisofosfatidilcolinas/biosíntesis , Proteínas Supresoras de Tumor/deficiencia , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Femenino , Humanos , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/patología , Lisofosfatidilcolinas/genética , Espectrometría de Masas , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Naftiridinas/farmacología , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo , Ratas , Sirolimus/farmacología , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa
16.
J Virol ; 87(24): 13179-92, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24067972

RESUMEN

We previously demonstrated that simian virus 40 (SV40) large T antigen (LT) binds to the Bub1 kinase, a key regulator of the spindle checkpoint and chromosome segregation. Bub1 mutations or altered expression patterns are linked to chromosome missegregation and are considered to be a driving force in some human cancers. Here we report that LT, dependent on Bub1 binding, causes micronuclei, lagging chromatin, and anaphase bridges, which are hallmarks of chromosomal instability (CIN) and Bub1 insufficiency. Using time-lapse microscopy, we demonstrate that LT imposes a Bub1 binding-dependent delay in the metaphase-to-anaphase transition. Kinetochore fibers reveal that LT, via Bub1 binding, causes aberrant kinetochore (KT)-microtubule (MT) attachments and a shortened interkinetochore distance, consistent with a lack of tension. Previously, we showed that LT also induces the DNA damage response (DDR) via Bub1 binding. Using inducible LT cell lines, we show that an activated DDR was observed before the appearance of anaphase bridges and micronuclei. Furthermore, LT induction in serum-starved cells demonstrated γ-H2AX accumulation in cells that had not yet entered mitosis. Thus, DDR activation can occur independently of chromosome segregation defects. Replication stress pathways may be responsible, because signatures of replication stress were observed, which were attenuated by exogenous supplementation with nucleosides. Our observations allow us to propose a model that explains and integrates the diverse manifestations of genomic instability induced by LT.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Replicación del ADN , Infecciones por Polyomavirus/genética , Infecciones por Polyomavirus/virología , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/virología , Anafase , Antígenos Virales de Tumores/genética , Cromatina/genética , Cromatina/metabolismo , Inestabilidad Cromosómica , Daño del ADN , Humanos , Cinetocoros/metabolismo , Mitosis , Infecciones por Polyomavirus/enzimología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Virus 40 de los Simios/genética , Virus 40 de los Simios/metabolismo , Infecciones Tumorales por Virus/enzimología
17.
Biochim Biophys Acta ; 1813(10): 1872-82, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21782855

RESUMEN

Human cytomegalovirus utilizes cellular signal transduction pathways to activate viral or cellular transcription factors involved in the control of viral gene expression and DNA replication. In the present study, we demonstrate that Harvey-ras-transformed cells show increased permissiveness to human cytomegalovirus when compared to their parental non-transformed cells. Both the progeny viral yield and the protein levels were elevated in the human cytomegalovirus-infected Harvey-ras-transformed cells requiring active viral gene replication, as shown by the infection with UV-inactivated human cytomegalovirus. Inhibition of Ras or of key molecules of the Ras pathway, effectively suppressed viral infection in the Harvey-ras-transformed cells. On a cellular level, the human cytomegalovirus-infected Harvey-ras-transformed cells formed larger cellular foci, which were significantly higher in number, compared to the uninfected cells and preferentially recruited human cytomegalovirus virions, thereby incriminating human cytomegalovirus infection for the increased transformation of these cells. Furthermore, proliferation assays revealed a higher rate for the human cytomegalovirus-infected Harvey-ras-transformed cells compared to mock-infected cells, whereas human cytomegalovirus infection had no considerable effect on the proliferation of the non-transformed cells. Higher susceptibility to apoptosis was also detected in the human cytomegalovirus-infected ras-transformed cells, which in combination with the higher progeny virus reveals a mode by which human cytomegalovirus achieves efficient spread of infection in the cells expressing the oncogenic Harvey-ras (12V) gene. Collectively, our data suggest that human cytomegalovirus employs the host-cell Ras signaling pathway to ensue viral expression and ultimately successful propagation. Transformed cells with an activated Ras signaling pathway are therefore particularly susceptible to human cytomegalovirus infection.


Asunto(s)
Citomegalovirus/fisiología , Genes ras/fisiología , Interacciones Huésped-Patógeno/genética , Animales , Línea Celular Transformada , Células Cultivadas , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/genética , Predisposición Genética a la Enfermedad , Humanos , Ratones , Células 3T3 NIH , Ratas , Ratas Endogámicas F344 , Transducción de Señal/genética , Transducción de Señal/fisiología , Transfección , Replicación Viral/genética
18.
Eur J Med Chem ; 46(6): 2586-95, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21497957

RESUMEN

The methoxylated trans-stilbene resveratrol analogue, (E)-3,4,5,4'-tetramethoxystilbene (1), has shown promising antiproliferative activity in in vitro cell line and in vivo models. In vivo 1 gives rise to several metabolic products through demethylation or hydroxylation reactions at the stilbene moiety. In the present study we examined the anticancer activity of 1 and the metabolites (E)-3'-hydroxy-3,4,5,4'-tetramethoxystilbene (2), (E)-4'-hydroxy-3,4,5-trimethoxystilbene (3), (E)-4-hydroxy-3,5,4'-trimethoxystilbene (4) and (E)-3-hydroxy-4,5,4'-trimethoxystilbene (5) by means of cell viability testing, cell cycle analysis, immunostaining and Western blotting. Compounds 1 and 2 exhibited submicromolar toxicity in MCF-7 breast adenocarcinoma and HepG2 hepatoma cells, whereas 3, 4 and 5 were inactive in terms of inhibition of cellular proliferation. Incubation with 1 or 2 at 10 µM for 24h induced apoptosis and G2/M cell cycle arrest in MCF-7 and HepG2 cells. Immunostaining of MCF-7 cells for ß-tubulin in the presence of either 1 or 2 revealed shorter localization of the protein around the nucleus, as compared to control cells. Western blot analyses further demonstrated that treatment with either 1 or 2 at concentrations between 30 and 50 µM for 24 h caused a downregulation in the levels of ß-tubulin and cyclin D1 expression and an upregulation in the levels of p53 expression in MCF-7 and HepG2 cells. 2 further increased the ratio of mRNA levels of the apoptosis-related genes Bax/Bcl-xL in both MCF-7 and HepG2 cells in a dose-dependent manner. We conclude that 2 inhibits HepG2 and MCF-7 cellular proliferation by inducing apoptosis and G2/M arrest through p53 and Bax/Bcl-xL upregulation. Our findings further demonstrate that trimethoxy substitutions along with the presence of a methoxy group at position 4' are necessary for retaining the activity of 1.


Asunto(s)
Antineoplásicos/farmacología , Estilbenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Biología Computacional , Simulación por Computador , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células Hep G2 , Humanos , Modelos Moleculares , Estructura Molecular , Estereoisomerismo , Estilbenos/química , Estilbenos/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
19.
Biochim Biophys Acta ; 1803(7): 777-85, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20303365

RESUMEN

Cancer is the final result of the accumulation of several genetic alterations occurring in a cell. Several herpesviruses and especially gamma-herpesviruses have played an important role in Cancer Biology, contributing significantly to our comprehension of cell signaling and growth control pathways which lead to malignancy. Unlike other infectious agents, herpesviruses persist in the host by establishing a latent infection, so that they can reactivate periodically. Interestingly, some herpesviruses are able to either deliver or induce the expression of cellular oncogenes. Such alterations can result in the derailment of the normal cell cycle and ultimately shift the balance between continuous proliferation and programmed cell death. Herpesvirus infection employs key molecules of cellular signaling cascades mostly to enhance viral replication. However, most of these molecules are also involved in essential cellular functions, such as proliferation, cellular differentiation and migration, as well as in DNA repair mechanisms. Ras proteins are key molecules that regulate a wide range of cellular functions, including differentiation, proliferation and cell survival. A broad field of medical research is currently focused on elucidating the role of ras oncogenes in human tumor initiation as well as tumor progression and metastasis. Upon activation, Ras proteins employ several downstream effector molecules such as phosphatidylinositol 3-kinase (PI3-K) and Raf and Ral guanine nucleotide-dissociation stimulators (RALGDS) to regulate a cascade of events ranging from cell proliferation and survival to apoptosis and cellular death. In this review, we give an overview of the impact that herpesvirus infection has on the host-cell Ras signaling pathway, providing an outline of their interactions with the key cascade molecules with which they associate. Several of these interactions of viral proteins with member of the Ras signaling pathway may be crucial in determining herpesviruses' oncogenic potential or their oncomodulatory behavior. The questions that emerge concern the potential role of these molecules as therapeutic targets both for viral infections and cancer. Understanding the means by which viruses may cause oncogenesis would therefore provide a deeper knowledge of the overall oncogenic process.


Asunto(s)
Herpesviridae/metabolismo , Transducción de Señal/fisiología , Proteínas ras/metabolismo , Animales , Transformación Celular Neoplásica , Herpesviridae/patogenicidad , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...