Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-39012319

ABSTRACT

The Mammalian Target of Rapamycin Complex 1 (mTORC1) is a serine threonine kinase that couples nutrient and growth factor signaling to the cellular control of metabolism and plays a fundamental role in aberrant proliferation in cancer. mTORC1 has previously been considered an "on/off" switch, capable of phosphorylating the entire pool of its substrates when activated. However recent studies have indicated that mTORC1 may be active towards its canonical substrates, 4EBP1 and S6K, involved in mRNA translation and protein synthesis, and inactive towards TFEB and TFE3, transcription factors involved in the regulation of lysosome biogenesis, in several pathological contexts. Among these conditions are Birt Hogg Dube (BHD) and recently, Tuberous Sclerosis Complex (TSC). Furthermore, TFEB and TFE3 hyperactivation in these syndromes, and in translocation Renal Cell Carcinomas (tRCC), drives mTORC1 activity towards the canonical substrates, through the transcriptional activation of the Rag GTPases, thereby positioning TFEB and TFE3 upstream of mTORC1 activity towards 4EBP1 and S6K. The expanding importance of TFEB and TFE3 in the pathogenesis of these renal diseases warrants a novel clinical grouping that we term "TFEopathies". Currently, there no therapeutic options directly targeting TFEB and TFE3, which represents a challenging and critically required avenue for cancer research.

2.
Nat Commun ; 15(1): 406, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38195686

ABSTRACT

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.


Subject(s)
Kidney Neoplasms , Tuberous Sclerosis , Animals , Mice , Mechanistic Target of Rapamycin Complex 1 , Mice, Knockout , Sirolimus/pharmacology , Tuberous Sclerosis/genetics
4.
Nat Commun ; 12(1): 4245, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34253722

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Organelle Biogenesis , Tuberous Sclerosis Complex 2 Protein/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinoma, Renal Cell/pathology , Cell Nucleus/metabolism , Cell Proliferation , Female , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Kidney Neoplasms/pathology , Lysosomes/ultrastructure , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Phosphoserine/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Tuberous Sclerosis Complex 2 Protein/deficiency , Tumor Suppressor Proteins/metabolism
6.
Sci Rep ; 8(1): 14161, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242175

ABSTRACT

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.


Subject(s)
Carcinogenesis/metabolism , Class III Phosphatidylinositol 3-Kinases/metabolism , Pinocytosis/physiology , Tuberous Sclerosis/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Autophagy/drug effects , Autophagy/physiology , Autophagy-Related Protein 5/metabolism , Carcinogenesis/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dextrans/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Pinocytosis/drug effects , Protein Kinase Inhibitors/pharmacology , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis Complex 2 Protein/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
7.
JCI Insight ; 3(8)2018 04 19.
Article in English | MEDLINE | ID: mdl-29669930

ABSTRACT

Tuberous sclerosis complex (TSC) is an incurable multisystem disease characterized by mTORC1-hyperactive tumors. TSC1/2 mutations also occur in other neoplastic disorders, including lymphangioleiomyomatosis (LAM) and bladder cancer. Whether TSC-associated tumors will respond to immunotherapy is unknown. We report here that the programmed death 1 coinhibitory receptor (PD-1) is upregulated on T cells in renal angiomyolipomas (AML) and pulmonary lymphangioleiomyomatosis (LAM). In C57BL/6J mice injected with syngeneic TSC2-deficient cells, anti-PD-1 alone decreased 105K tumor growth by 67% (P < 0.0001); the combination of PD-1 and CTLA-4 blockade was even more effective in suppressing tumor growth. Anti-PD-1 induced complete rejection of TSC2-deficient 105K tumors in 37% of mice (P < 0.05). Double blockade of PD-1 and CTLA-4 induced rejection in 62% of mice (P < 0.01). TSC2 reexpression in TSC2-deficient TMKOC cells enhanced antitumor immunity by increasing T cell infiltration and production of IFN-γ/TNF-α by T cells, suggesting that TSC2 and mTORC1 play specific roles in the induction of antitumor immunity. Finally, 1 month of anti-PD-1 blockade reduced renal tumor burden by 53% (P < 0.01) in genetically engineered Tsc2+/- mice. Taken together, these data demonstrate for the first time to our knowledge that checkpoint blockade may have clinical efficacy for TSC and LAM, and possibly other benign tumor syndromes, potentially yielding complete and durable clinical responses.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy/methods , Mechanistic Target of Rapamycin Complex 1/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis/genetics , Angiomyolipoma/complications , Angiomyolipoma/genetics , Angiomyolipoma/immunology , Animals , CTLA-4 Antigen/metabolism , Drug Therapy, Combination , Lymphangioleiomyomatosis/complications , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/immunology , Male , Mice , Mice, Inbred C57BL , Mutation , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/etiology , Tuberous Sclerosis/immunology , Tuberous Sclerosis Complex 1 Protein , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/complications , Urinary Bladder Neoplasms/pathology
8.
Oncotarget ; 8(39): 64714-64727, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29029388

ABSTRACT

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.

9.
Oncotarget ; 8(24): 38099-38112, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28498820

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Lysosomes/metabolism , Membrane Glycoproteins/metabolism , Receptors, LDL/metabolism , Tuberous Sclerosis/metabolism , Cell Line , Homeostasis/physiology , Humans , Intracellular Signaling Peptides and Proteins , Niemann-Pick C1 Protein
10.
Cancer Res ; 77(12): 3255-3267, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28512249

ABSTRACT

p62/sequestosome-1 (SQSTM1) is a multifunctional adaptor protein and autophagic substrate that accumulates in cells with hyperactive mTORC1, such as kidney cells with mutations in the tumor suppressor genes tuberous sclerosis complex (TSC)1 or TSC2. Here we report that p62 is a critical mediator of TSC2-driven tumorigenesis, as Tsc2+/- and Tsc2f/f Ksp-CreERT2+ mice crossed to p62-/- mice were protected from renal tumor development. Metabolic profiling revealed that depletion of p62 in Tsc2-null cells decreased intracellular glutamine, glutamate, and glutathione (GSH). p62 positively regulated the glutamine transporter Slc1a5 and increased glutamine uptake in Tsc2-null cells. We also observed p62-dependent changes in Gcl, Gsr, Nqo1, and Srxn1, which were decreased by p62 attenuation and implicated in GSH production and utilization. p62 attenuation altered mitochondrial morphology, reduced mitochondrial membrane polarization and maximal respiration, and increased mitochondrial reactive oxygen species and mitophagy marker PINK1. These mitochondrial phenotypes were rescued by addition of exogenous GSH and overexpression of Sod2, which suppressed indices of mitochondrial damage and promoted growth of Tsc2-null cells. Finally, p62 depletion sensitized Tsc2-null cells to both oxidative stress and direct inhibition of GSH biosynthesis by buthionine sulfoximine. Our findings show how p62 helps maintain intracellular pools of GSH needed to limit mitochondrial dysfunction in tumor cells with elevated mTORC1, highlighting p62 and redox homeostasis as nodal vulnerabilities for therapeutic targeting in these tumors. Cancer Res; 77(12); 3255-67. ©2017 AACR.


Subject(s)
Carcinogenesis/metabolism , Mitochondria/pathology , Multiprotein Complexes/metabolism , Sequestosome-1 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Animals , Carcinogenesis/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Glutathione/biosynthesis , Immunohistochemistry , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
11.
PLoS Genet ; 12(8): e1006242, 2016 08.
Article in English | MEDLINE | ID: mdl-27494029

ABSTRACT

Renal angiomyolipoma is a kidney tumor in the perivascular epithelioid (PEComa) family that is common in patients with Tuberous Sclerosis Complex (TSC) and Lymphangioleiomyomatosis (LAM) but occurs rarely sporadically. Though histologically benign, renal angiomyolipoma can cause life-threatening hemorrhage and kidney failure. Both angiomyolipoma and LAM have mutations in TSC2 or TSC1. However, the frequency and contribution of other somatic events in tumor development is unknown. We performed whole exome sequencing in 32 resected tumor samples (n = 30 angiomyolipoma, n = 2 LAM) from 15 subjects, including three with TSC. Two germline and 22 somatic inactivating mutations in TSC2 were identified, and one germline TSC1 mutation. Twenty of 32 (62%) samples showed copy neutral LOH (CN-LOH) in TSC2 or TSC1 with at least 8 different LOH regions, and 30 of 32 (94%) had biallelic loss of either TSC2 or TSC1. Whole exome sequencing identified a median of 4 somatic non-synonymous coding region mutations (other than in TSC2/TSC1), a mutation rate lower than nearly all other cancer types. Three genes with mutations were known cancer associated genes (BAP1, ARHGAP35 and SPEN), but they were mutated in a single sample each, and were missense variants with uncertain functional effects. Analysis of sixteen angiomyolipomas from a TSC subject showed both second hit point mutations and CN-LOH in TSC2, many of which were distinct, indicating that they were of independent clonal origin. However, three tumors had two shared mutations in addition to private somatic mutations, suggesting a branching evolutionary pattern of tumor development following initiating loss of TSC2. Our results indicate that TSC2 and less commonly TSC1 alterations are the primary essential driver event in angiomyolipoma/LAM, whereas other somatic mutations are rare and likely do not contribute to tumor development.


Subject(s)
Angiomyolipoma/genetics , Kidney Neoplasms/genetics , Lymphangioleiomyomatosis/genetics , Tumor Suppressor Proteins/genetics , Adult , Angiomyolipoma/pathology , Carcinogenesis/genetics , Exome/genetics , Female , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Humans , Kidney Neoplasms/pathology , Loss of Heterozygosity/genetics , Lymphangioleiomyomatosis/pathology , Male , Mutation , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
12.
Curr Drug Metab ; 12(3): 278-86, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21470106

ABSTRACT

It is well known that interindividual variability can affect the response to many drugs in relation to age, gender, diet, and organ function. Pharmacogenomic studies have also documented that genetic polymorphisms can exert clinically significant effects in terms of drug resistance, efficacy and toxicity by modifying the expression of critical gene products (drug-metabolizing enzymes, transporters, and target molecules) as well as pharmacokinetic and pharmacodynamic parameters. A growing body of in vitro and clinical evidence suggests that common polymorphisms in the folate gene pathway are associated with an altered response to methotrexate (MTX) in patients with malignancy and autoimmune disease. Such polymorphisms may also induce significant MTX toxicity requiring expensive monitoring and treatment. Although the available data are not conclusive, they suggest that in the future MTX pharmacogenetics could play a key role in clinical practice by improving and tailoring treatment. This review describes the genetic polymorphisms that significantly influence MTX resistance, efficacy, and toxicity.


Subject(s)
Methotrexate/metabolism , Methotrexate/pharmacology , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacology , Drug Resistance , Folic Acid Antagonists/adverse effects , Folic Acid Antagonists/metabolism , Folic Acid Antagonists/pharmacology , Humans , Methotrexate/adverse effects , Pharmacogenetics , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...