Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Cancer ; 5(3): 448-462, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38267628

ABSTRACT

Chemotherapy often generates intratumoral senescent cancer cells that strongly modify the tumor microenvironment, favoring immunosuppression and tumor growth. We discovered, through an unbiased proteomics screen, that the immune checkpoint inhibitor programmed cell death 1 ligand 2 (PD-L2) is highly upregulated upon induction of senescence in different types of cancer cells. PD-L2 is not required for cells to undergo senescence, but it is critical for senescent cells to evade the immune system and persist intratumorally. Indeed, after chemotherapy, PD-L2-deficient senescent cancer cells are rapidly eliminated and tumors do not produce the senescence-associated chemokines CXCL1 and CXCL2. Accordingly, PD-L2-deficient pancreatic tumors fail to recruit myeloid-derived suppressor cells and undergo regression driven by CD8 T cells after chemotherapy. Finally, antibody-mediated blockade of PD-L2 strongly synergizes with chemotherapy causing remission of mammary tumors in mice. The combination of chemotherapy with anti-PD-L2 provides a therapeutic strategy that exploits vulnerabilities arising from therapy-induced senescence.


Subject(s)
Pancreatic Neoplasms , Animals , Mice , Pancreatic Neoplasms/metabolism , CD8-Positive T-Lymphocytes/pathology , Immune Tolerance , Immunosuppression Therapy , Cellular Senescence , Tumor Microenvironment
2.
Cancers (Basel) ; 12(7)2020 Jul 04.
Article in English | MEDLINE | ID: mdl-32635473

ABSTRACT

Despite significant efforts to improve pancreatic ductal adenocarcinoma (PDAC) clinical outcomes, overall survival remains dismal. The poor response to current therapies is partly due to the existence of pancreatic cancer stem cells (PaCSCs), which are efficient drivers of PDAC tumorigenesis, metastasis and relapse. To find new therapeutic agents that could efficiently kill PaCSCs, we screened a chemical library of 680 compounds for candidate small molecules with anti-CSC activity, and identified two compounds of a specific chemical series with potent activity in vitro and in vivo against patient-derived xenograft (PDX) cultures. The anti-CSC mechanism of action of this specific chemical series was found to rely on induction of lysosomal membrane permeabilization (LMP), which is likely associated with the increased lysosomal mass observed in PaCSCs. Using the well characterized LMP-inducer siramesine as a tool molecule, we show elimination of the PaCSC population in mice implanted with tumors from two PDX models. Collectively, our approach identified lysosomal disruption as a promising anti-CSC therapeutic strategy for PDAC.

3.
ESMO Open ; 1(5): e000076, 2016.
Article in English | MEDLINE | ID: mdl-27843638

ABSTRACT

Understanding the early evolution of cancer heterogeneity during the initial steps of tumorigenesis can uncover vulnerabilities of cancer cells that may be masked at later stages. We describe a comprehensive approach employing gene expression analysis in early lesions to identify novel therapeutic targets and the use of mouse models to test synthetic lethal drug combinations to treat human Kirsten rat sarcoma viral oncogene homologue (KRAS)-driven lung adenocarcinoma.

4.
Proc Natl Acad Sci U S A ; 112(26): E3402-11, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26080399

ABSTRACT

Genetic aberrations responsible for soft-tissue sarcoma formation in adults are largely unknown, with targeted therapies sorely needed for this complex and heterogeneous family of diseases. Here we report that that the Hippo pathway is deregulated in many soft-tissue sarcomas, resulting in elevated expression of the effector molecule Yes-Associated Protein (YAP). Based on data gathered from human sarcoma patients, a novel autochthonous mouse model, and mechanistic analyses, we determined that YAP-dependent expression of the transcription factor forkhead box M1 (FOXM1) is necessary for cell proliferation/tumorigenesis in a subset of soft-tissue sarcomas. Notably, FOXM1 directly interacts with the YAP transcriptional complex via TEAD1, resulting in coregulation of numerous critical pro-proliferation targets that enhance sarcoma progression. Finally, pharmacologic inhibition of FOXM1 decreases tumor size in vivo, making FOXM1 an attractive therapeutic target for the treatment of some sarcoma subtypes.


Subject(s)
Carcinogenesis , Forkhead Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Sarcoma/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Forkhead Box Protein M1 , Forkhead Transcription Factors/physiology , Hippo Signaling Pathway , Humans , Phosphoproteins/metabolism , Sarcoma/pathology , Transcription Factors , YAP-Signaling Proteins
5.
J Clin Invest ; 125(2): 824-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25574842

ABSTRACT

NOTCH signaling suppresses tumor growth and proliferation in several types of stratified epithelia. Here, we show that missense mutations in NOTCH1 and NOTCH2 found in human bladder cancers result in loss of function. In murine models, genetic ablation of the NOTCH pathway accelerated bladder tumorigenesis and promoted the formation of squamous cell carcinomas, with areas of mesenchymal features. Using bladder cancer cells, we determined that the NOTCH pathway stabilizes the epithelial phenotype through its effector HES1 and, consequently, loss of NOTCH activity favors the process of epithelial-mesenchymal transition. Evaluation of human bladder cancer samples revealed that tumors with low levels of HES1 present mesenchymal features and are more aggressive. Together, our results indicate that NOTCH serves as a tumor suppressor in the bladder and that loss of this pathway promotes mesenchymal and invasive features.


Subject(s)
Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Transcription Factor HES-1 , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
6.
Aging Cell ; 13(6): 1087-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25116423

ABSTRACT

Exceptional longevity (EL) is a rare phenotype that can cluster in families, and co-segregation of genetic variation in these families may point to candidate genes that could contribute to extended lifespan. In this study, for the first time, we have sequenced a total of seven exomes from exceptionally long-lived siblings (probands ≥ 103 years and at least one sibling ≥ 97 years) that come from three separate families. We have focused on rare functional variants (RFVs) which have ≤ 1% minor allele frequency according to databases and that are likely to alter gene product function. Based on this, we have identified one candidate longevity gene carrying RFVs in all three families, APOB. Interestingly, APOB is a component of lipoprotein particles together with APOE, and variants in the genes encoding these two proteins have been previously associated with human longevity. Analysis of nonfamilial EL cases showed a trend, without reaching statistical significance, toward enrichment of APOB RFVs. We have also identified candidate longevity genes shared between two families (5-13) or within individual families (66-156 genes). Some of these genes have been previously linked to longevity in model organisms, such as PPARGC1A, NRG1, RAD52, RAD51, NCOR1, and ADCY5 genes. This work provides an initial catalog of genes that could contribute to exceptional familial longevity.


Subject(s)
Exome , Gene Frequency , Longevity/genetics , Aged, 80 and over , Apolipoprotein B-100/genetics , Family Health , Female , Genetic Variation , Humans , Male
7.
Free Radic Biol Med ; 43(9): 1219-25, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17893032

ABSTRACT

Many organisms activate adaptive transcriptional programs to help them cope with decreased oxygen (O(2)) levels, or hypoxia, in their environment. These responses are triggered by various O(2) sensing systems in bacteria, yeast and metazoans. In metazoans, the hypoxia inducible factors (HIFs) mediate the adaptive transcriptional response to hypoxia by upregulating genes involved in maintaining bioenergetic homeostasis. The HIFs in turn are regulated by HIF-specific prolyl hydroxlase activity, which is sensitive to cellular O(2) levels and other factors such as tricarboxylic acid cycle metabolites and reactive oxygen species (ROS). Establishing a role for ROS in cellular oxygen sensing has been challenging since ROS are intrinsically unstable and difficult to measure. However, recent advances in fluorescence energy transfer resonance (FRET)-based methods for measuring ROS are alleviating some of the previous difficulties associated with dyes and luminescent chemicals. In addition, new genetic models have demonstrated that functional mitochondrial electron transport and associated ROS production during hypoxia are required for HIF stabilization in mammalian cells. Current efforts are directed at determining how ROS mediate prolyl hydroxylase activity and hypoxic HIF stabilization. Progress in understanding this process has been enhanced by the development of the FRET-based ROS probe, an vivo prolyl hydroxylase reporter and various genetic models harboring mutations in components of the mitochondrial electron transport chain.


Subject(s)
Oxygen/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Hypoxia/physiology , Homeostasis , Reactive Oxygen Species/chemistry
8.
Mol Cell ; 21(4): 521-31, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16483933

ABSTRACT

Oxygen (O2) deprivation, or hypoxia, has profound effects on cell metabolism and growth. Cells can adapt to low O2 in part through activation of hypoxia-inducible factor (HIF). We report here that hypoxia inhibits mRNA translation by suppressing multiple key regulators, including eIF2alpha, eEF2, and the mammalian target of rapamycin (mTOR) effectors 4EBP1, p70S6K, and rpS6, independent of HIF. Hypoxia results in energy starvation and activation of the AMPK/TSC2/Rheb/mTOR pathway. Hypoxic AMP-activated protein kinase (AMPK) activation also leads to eEF2 inhibition. Moreover, hypoxic effects on cellular bioenergetics and mTOR inhibition increase over time. Mutation of the TSC2 tumor suppressor gene confers a growth advantage to cells by repressing hypoxic mTOR inhibition and hypoxia-induced G1 arrest. Together, eIF2alpha, eEF2, and mTOR inhibition represent important HIF-independent mechanisms of energy conservation that promote survival under low O2 conditions.


Subject(s)
Energy Metabolism , Eukaryotic Initiation Factor-2/metabolism , Hypoxia , Peptide Elongation Factor 2/metabolism , Protein Biosynthesis , Protein Kinases/metabolism , RNA, Messenger/metabolism , AMP-Activated Protein Kinases , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , Enzyme Activation , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Multienzyme Complexes/metabolism , Neuropeptides/metabolism , Oxygen/metabolism , Peptide Elongation Factor 2/antagonists & inhibitors , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ras Homolog Enriched in Brain Protein , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , eIF-2 Kinase/metabolism
9.
Cell Metab ; 1(6): 393-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16054088

ABSTRACT

While cellular responses to low oxygen (O(2)) or hypoxia have been studied extensively, the precise identity of mammalian cellular O(2) sensors remains controversial. Using murine embryonic cells lacking cytochrome c, and therefore mitochondrial activity, we show that mitochondrial reactive oxygen species (mtROS) are essential for proper O(2) sensing and subsequent HIF-1 alpha and HIF-2 alpha stabilization at 1.5% O(2). In the absence of this signal, HIF-alpha subunits continue to be degraded. Furthermore, exogenous treatment with H(2)O(2) or severe O(2) deprivation is sufficient to stabilize HIF-alpha even in the absence of cytochrome c and functional mitochondria. These results provide genetic evidence indicating that mtROS act upstream of prolyl hydroxylases in regulating HIF-1 alpha and HIF-2 alpha in this O(2)-sensing pathway.


Subject(s)
Cytochromes c/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Cells, Cultured , DNA Replication/physiology , DNA, Mitochondrial/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Mice
10.
Oncogene ; 21(55): 8470-6, 2002 Dec 05.
Article in English | MEDLINE | ID: mdl-12466966

ABSTRACT

Tuberous sclerosis complex (TSC) is a tumor suppressor gene syndrome characterized by seizures, mental retardation, autism, and tumors of the brain, kidney, heart, retina, and skin. TSC is caused by mutations in either TSC1 or TSC2, both of which are tumor suppressor genes. Hamartin, the protein product of TSC1, was found to interact with the ezrin-radixin-moesin family of cytoskeletal proteins and to activate the small GTPase Rho. To determine whether tuberin, the TSC2 product, can also activate Rho, we stably expressed full-length human tuberin in two cell types: MDCK cells and ELT3 cells. ELT3 cells lack endogenous tuberin expression. We found that expression of human tuberin in both MDCK and ELT3 cells was associated with an increase in the amount of Rho-GTP, but not in Rac1-GTP or cdc42-GTP. Tuberin expression increased cell adhesion in both cell types, and decreased chemotactic cell migration in ELT3 cells. In MDCK cells, there was a decrease in the amount of total Focal Adhesion Kinase (FAK) and an increase in the fraction of phosphorylated FAK. These findings demonstrate for the first time that tuberin activates Rho and regulates cell adhesion and migration. Pathways involving Rho activation may have relevance to the clinical manifestations of TSC, including pulmonary lymphangioleiomyomatosis.


Subject(s)
Cell Adhesion/genetics , Genes, Tumor Suppressor , Repressor Proteins/genetics , rho GTP-Binding Proteins/metabolism , Animals , Cell Line , Cell Movement/genetics , Dogs , Gene Expression Regulation, Neoplastic , Humans , Proteins/genetics , Rats , Transfection , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...