Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
2.
Nature ; 615(7953): 712-719, 2023 03.
Article in English | MEDLINE | ID: mdl-36922590

ABSTRACT

Mitochondria are critical to the governance of metabolism and bioenergetics in cancer cells1. The mitochondria form highly organized networks, in which their outer and inner membrane structures define their bioenergetic capacity2,3. However, in vivo studies delineating the relationship between the structural organization of mitochondrial networks and their bioenergetic activity have been limited. Here we present an in vivo structural and functional analysis of mitochondrial networks and bioenergetic phenotypes in non-small cell lung cancer (NSCLC) using an integrated platform consisting of positron emission tomography imaging, respirometry and three-dimensional scanning block-face electron microscopy. The diverse bioenergetic phenotypes and metabolic dependencies we identified in NSCLC tumours align with distinct structural organization of mitochondrial networks present. Further, we discovered that mitochondrial networks are organized into distinct compartments within tumour cells. In tumours with high rates of oxidative phosphorylation (OXPHOSHI) and fatty acid oxidation, we identified peri-droplet mitochondrial networks wherein mitochondria contact and surround lipid droplets. By contrast, we discovered that in tumours with low rates of OXPHOS (OXPHOSLO), high glucose flux regulated perinuclear localization of mitochondria, structural remodelling of cristae and mitochondrial respiratory capacity. Our findings suggest that in NSCLC, mitochondrial networks are compartmentalized into distinct subpopulations that govern the bioenergetic capacity of tumours.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Energy Metabolism , Lung Neoplasms , Mitochondria , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Fatty Acids/metabolism , Glucose/metabolism , Lipid Droplets/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/ultrastructure , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , Oxidative Phosphorylation , Phenotype , Positron-Emission Tomography
3.
Nat Commun ; 13(1): 1090, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35228570

ABSTRACT

LKB1 is among the most frequently altered tumor suppressors in lung adenocarcinoma. Inactivation of Lkb1 accelerates the growth and progression of oncogenic KRAS-driven lung tumors in mouse models. However, the molecular mechanisms by which LKB1 constrains lung tumorigenesis and whether the cancer state that stems from Lkb1 deficiency can be reverted remains unknown. To identify the processes governed by LKB1 in vivo, we generated an allele which enables Lkb1 inactivation at tumor initiation and subsequent Lkb1 restoration in established tumors. Restoration of Lkb1 in oncogenic KRAS-driven lung tumors suppressed proliferation and led to tumor stasis. Lkb1 restoration activated targets of C/EBP transcription factors and drove neoplastic cells from a progenitor-like state to a less proliferative alveolar type II cell-like state. We show that C/EBP transcription factors govern a subset of genes that are induced by LKB1 and depend upon NKX2-1. We also demonstrate that a defining factor of the alveolar type II lineage, C/EBPα, constrains oncogenic KRAS-driven lung tumor growth in vivo. Thus, this key tumor suppressor regulates lineage-specific transcription factors, thereby constraining lung tumor development through enforced differentiation.


Subject(s)
AMP-Activated Protein Kinases , Lung Neoplasms , Proto-Oncogene Proteins p21(ras) , AMP-Activated Protein Kinases/metabolism , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics
5.
Cell Chem Biol ; 29(3): 423-435.e10, 2022 03 17.
Article in English | MEDLINE | ID: mdl-34715056

ABSTRACT

Efforts to target glucose metabolism in cancer have been limited by the poor potency and specificity of existing anti-glycolytic agents and a poor understanding of the glucose dependence of cancer subtypes in vivo. Here, we present an extensively characterized series of potent, orally bioavailable inhibitors of the class I glucose transporters (GLUTs). The representative compound KL-11743 specifically blocks glucose metabolism, triggering an acute collapse in NADH pools and a striking accumulation of aspartate, indicating a dramatic shift toward oxidative phosphorylation in the mitochondria. Disrupting mitochondrial metabolism via chemical inhibition of electron transport, deletion of the malate-aspartate shuttle component GOT1, or endogenous mutations in tricarboxylic acid cycle enzymes, causes synthetic lethality with KL-11743. Patient-derived xenograft models of succinate dehydrogenase A (SDHA)-deficient cancers are specifically sensitive to KL-11743, providing direct evidence that TCA cycle-mutant tumors are vulnerable to GLUT inhibitors in vivo.


Subject(s)
Citric Acid Cycle , Neoplasms , Aspartic Acid/metabolism , Glucose/metabolism , Humans , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism
6.
Article in English | MEDLINE | ID: mdl-34127512

ABSTRACT

Lung cancer is a heterogeneous disease that is subdivided into histopathological subtypes with distinct behaviors. Each subtype is characterized by distinct features and molecular alterations that influence tumor metabolism. Alterations in tumor metabolism can be exploited by imaging modalities that use metabolite tracers for the detection and characterization of tumors. Microenvironmental factors, including nutrient and oxygen availability and the presence of stromal cells, are a critical influence on tumor metabolism. Recent technological advances facilitate the direct evaluation of metabolic alterations in patient tumors in this complex microenvironment. In addition, molecular alterations directly influence tumor cell metabolism and metabolic dependencies that influence response to therapy. Current therapeutic approaches to target tumor metabolism are currently being developed and translated into the clinic for patient therapy.


Subject(s)
Adaptation, Physiological/genetics , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Phenotype , Tumor Microenvironment , Amino Acids/analysis , Blood Glucose/analysis , Genes, erbB-1/genetics , Genes, p53/genetics , Humans , Molecular Targeted Therapy , Nucleotides/metabolism , Positron-Emission Tomography , Proto-Oncogene Proteins p21(ras)/metabolism , Tomography, X-Ray Computed
7.
Cancer Res ; 81(12): 3295-3308, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33853830

ABSTRACT

LKB1 inactivating mutations are commonly observed in patients with KRAS-mutant non-small cell lung cancer (NSCLC). Although treatment of NSCLC with immune checkpoint inhibitors (ICI) has resulted in improved overall survival in a subset of patients, studies have revealed that co-occurring KRAS/LKB1 mutations drive primary resistance to ICIs in NSCLC. Effective therapeutic options that overcome ICI resistance in LKB1-mutant NSCLC are limited. Here, we report that loss of LKB1 results in increased secretion of the C-X-C motif (CXC) chemokines with an NH2-terminal Glu-Leu-Arg (ELR) motif in premalignant and cancerous cells, as well as in genetically engineered murine models (GEMM) of NSCLC. Heightened levels of ELR+ CXC chemokines in LKB1-deficient murine models of NSCLC positively correlated with increased abundance of granulocytic myeloid-derived suppressor cells (G-MDSC) locally within the tumor microenvironment and systemically in peripheral blood and spleen. Depletion of G-MDSCs with antibody or functional inhibition via all-trans-retinoic acid (ATRA) led to enhanced antitumor T-cell responses and sensitized LKB1-deficent murine tumors to PD-1 blockade. Combination therapy with anti-PD-1 and ATRA improved local and systemic T-cell proliferation and generated tumor-specific immunity. Our findings implicate ELR+ CXC chemokine-mediated enrichment of G-MDSCs as a potential mediator of immunosuppression in LKB1-deficient NSCLC and provide a rationale for using ATRA in combination with anti-PD-1 therapy in patients with LKB1-deficient NSCLC refractory to ICIs. SIGNIFICANCE: These findings show that accumulation of myeloid-derived suppressor cells in LKB1-deficient non-small cell lung cancer can be overcome via treatment with all-trans-retinoic acid, sensitizing tumors to immunotherapy.


Subject(s)
AMP-Activated Protein Kinase Kinases/deficiency , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Granulocytes/immunology , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Myeloid-Derived Suppressor Cells/immunology , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Nat Commun ; 12(1): 1876, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767183

ABSTRACT

Viruses hijack host cell metabolism to acquire the building blocks required for replication. Understanding how SARS-CoV-2 alters host cell metabolism may lead to potential treatments for COVID-19. Here we profile metabolic changes conferred by SARS-CoV-2 infection in kidney epithelial cells and lung air-liquid interface (ALI) cultures, and show that SARS-CoV-2 infection increases glucose carbon entry into the TCA cycle via increased pyruvate carboxylase expression. SARS-CoV-2 also reduces oxidative glutamine metabolism while maintaining reductive carboxylation. Consistent with these changes, SARS-CoV-2 infection increases the activity of mTORC1 in cell lines and lung ALI cultures. Lastly, we show evidence of mTORC1 activation in COVID-19 patient lung tissue, and that mTORC1 inhibitors reduce viral replication in kidney epithelial cells and lung ALI cultures. Our results suggest that targeting mTORC1 may be a feasible treatment strategy for COVID-19 patients, although further studies are required to determine the mechanism of inhibition and potential efficacy in patients.


Subject(s)
COVID-19/pathology , Citric Acid Cycle/physiology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Benzamides/pharmacology , Cell Line , Chlorocebus aethiops , Glucose/metabolism , Glutamine/metabolism , HEK293 Cells , Humans , Lung/metabolism , Lung/virology , Morpholines/pharmacology , Naphthyridines/pharmacology , Pyrimidines/pharmacology , Pyruvate Carboxylase/biosynthesis , SARS-CoV-2/metabolism , Vero Cells , Virus Replication/drug effects
9.
Cell Metab ; 33(5): 1013-1026.e6, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33609439

ABSTRACT

Mitochondrial respiration is critical for cell proliferation. In addition to producing ATP, respiration generates biosynthetic precursors, such as aspartate, an essential substrate for nucleotide synthesis. Here, we show that in addition to depleting intracellular aspartate, electron transport chain (ETC) inhibition depletes aspartate-derived asparagine, increases ATF4 levels, and impairs mTOR complex I (mTORC1) activity. Exogenous asparagine restores proliferation, ATF4 and mTORC1 activities, and mTORC1-dependent nucleotide synthesis in the context of ETC inhibition, suggesting that asparagine communicates active respiration to ATF4 and mTORC1. Finally, we show that combination of the ETC inhibitor metformin, which limits tumor asparagine synthesis, and either asparaginase or dietary asparagine restriction, which limit tumor asparagine consumption, effectively impairs tumor growth in multiple mouse models of cancer. Because environmental asparagine is sufficient to restore tumor growth in the context of respiration impairment, our findings suggest that asparagine synthesis is a fundamental purpose of tumor mitochondrial respiration, which can be harnessed for therapeutic benefit to cancer patients.


Subject(s)
Activating Transcription Factor 4/metabolism , Asparagine/metabolism , Mitochondria/metabolism , Animals , Asparagine/pharmacology , Aspartic Acid/deficiency , Aspartic Acid/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Diet/veterinary , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mice , Mice, Inbred NOD , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/mortality , Neoplasms/pathology , Nucleotides/metabolism , Survival Rate
10.
Cancer Immunol Immunother ; 70(8): 2389-2400, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33507343

ABSTRACT

Conditional genetically engineered mouse models (GEMMs) of non-small cell lung cancer (NSCLC) harbor common oncogenic driver mutations of the disease, but in contrast to human NSCLC these models possess low tumor mutational burden (TMB). As a result, these models often lack tumor antigens that can elicit host adaptive immune responses, which limits their utility in immunotherapy studies. Here, we establish Kras-mutant murine models of NSCLC bearing the common driver mutations associated with the disease and increased TMB, by in vitro exposure of cell lines derived from GEMMs of NSCLC [KrasG12D (K), KrasG12DTp53-/-(KP), KrasG12DTp53+/-Lkb1-/- (KPL)] to the alkylating agent N-methyl-N-nitrosourea (MNU). Increasing the TMB enhanced host anti-tumor T cell responses and improved anti-PD-1 efficacy in syngeneic models across all genetic backgrounds. However, limited anti-PD-1 efficacy was observed in the KPL cell lines with increased TMB, which possessed a distinct immunosuppressed tumor microenvironment (TME) primarily composed of granulocytic myeloid-derived suppressor cells (G-MDSCs). This KPL phenotype is consistent with findings in human KRAS-mutant NSCLC where LKB1 loss is a driver of primary resistance to PD-1 blockade. In summary, these novel Kras-mutant NSCLC murine models with known driver mutations and increased TMB have distinct TMEs and recapitulate the therapeutic vulnerabilities of human NSCLC. We anticipate that these immunogenic models will facilitate the development of innovative immunotherapies in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Disease Models, Animal , Mice , Protein Serine-Threonine Kinases/genetics , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics
11.
Cancer Discov ; 11(3): 714-735, 2021 03.
Article in English | MEDLINE | ID: mdl-33318037

ABSTRACT

MAPK targeting in cancer often fails due to MAPK reactivation. MEK inhibitor (MEKi) monotherapy provides limited clinical benefits but may serve as a foundation for combination therapies. Here, we showed that combining a type II RAF inhibitor (RAFi) with an allosteric MEKi durably prevents and overcomes acquired resistance among cancers with KRAS, NRAS, NF1, BRAF non-V600, and BRAF V600 mutations. Tumor cell-intrinsically, type II RAFi plus MEKi sequester MEK in RAF complexes, reduce MEK/MEK dimerization, and uncouple MEK from ERK in acquired-resistant tumor subpopulations. Immunologically, this combination expands memory and activated/exhausted CD8+ T cells, and durable tumor regression elicited by this combination requires CD8+ T cells, which can be reinvigorated by anti-PD-L1 therapy. Whereas MEKi reduces dominant intratumoral T-cell clones, type II RAFi cotreatment reverses this effect and promotes T-cell clonotypic expansion. These findings rationalize the clinical development of type II RAFi plus MEKi and their further combination with PD-1/L1-targeted therapy. SIGNIFICANCE: Type I RAFi + MEKi are indicated only in certain BRAF V600MUT cancers. In contrast, type II RAFi + MEKi are durably active against acquired MEKi resistance across broad cancer indications, which reveals exquisite MAPK addiction. Allosteric modulation of MAPK protein/protein interactions and temporal preservation of intratumoral CD8+ T cells are mechanisms that may be further exploited.This article is highlighted in the In This Issue feature, p. 521.


Subject(s)
Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Immunity, Cellular/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Protein Stability , Treatment Outcome , Xenograft Model Antitumor Assays
13.
Life Sci Alliance ; 3(7)2020 07.
Article in English | MEDLINE | ID: mdl-32499316

ABSTRACT

Recent breakthroughs in live-cell imaging have enabled visualization of cristae, making it feasible to investigate the structure-function relationship of cristae in real time. However, quantifying live-cell images of cristae in an unbiased way remains challenging. Here, we present a novel, semi-automated approach to quantify cristae, using the machine-learning Trainable Weka Segmentation tool. Compared with standard techniques, our approach not only avoids the bias associated with manual thresholding but more efficiently segments cristae from Airyscan and structured illumination microscopy images. Using a cardiolipin-deficient cell line, as well as FCCP, we show that our approach is sufficiently sensitive to detect perturbations in cristae density, size, and shape. This approach, moreover, reveals that cristae are not uniformly distributed within the mitochondrion, and sites of mitochondrial fission are localized to areas of decreased cristae density. After a fusion event, individual cristae from the two mitochondria, at the site of fusion, merge into one object with distinct architectural values. Overall, our study shows that machine learning represents a compelling new strategy for quantifying cristae in living cells.


Subject(s)
Mitochondria/physiology , Mitochondria/ultrastructure , Mitochondrial Dynamics , Cell Line , Humans , Image Processing, Computer-Assisted , Microscopy, Fluorescence/methods , Mitochondrial Membranes/physiology , Mitochondrial Membranes/ultrastructure , Optical Imaging/methods
15.
Nature ; 575(7782): 380-384, 2019 11.
Article in English | MEDLINE | ID: mdl-31666695

ABSTRACT

Mitochondria are essential regulators of cellular energy and metabolism, and have a crucial role in sustaining the growth and survival of cancer cells. A central function of mitochondria is the synthesis of ATP by oxidative phosphorylation, known as mitochondrial bioenergetics. Mitochondria maintain oxidative phosphorylation by creating a membrane potential gradient that is generated by the electron transport chain to drive the synthesis of ATP1. Mitochondria are essential for tumour initiation and maintaining tumour cell growth in cell culture and xenografts2,3. However, our understanding of oxidative mitochondrial metabolism in cancer is limited because most studies have been performed in vitro in cell culture models. This highlights a need for in vivo studies to better understand how oxidative metabolism supports tumour growth. Here we measure mitochondrial membrane potential in non-small-cell lung cancer in vivo using a voltage-sensitive, positron emission tomography (PET) radiotracer known as 4-[18F]fluorobenzyl-triphenylphosphonium (18F-BnTP)4. By using PET imaging of 18F-BnTP, we profile mitochondrial membrane potential in autochthonous mouse models of lung cancer, and find distinct functional mitochondrial heterogeneity within subtypes of lung tumours. The use of 18F-BnTP PET imaging enabled us to functionally profile mitochondrial membrane potential in live tumours.


Subject(s)
Carcinoma, Non-Small-Cell Lung/physiopathology , Lung Neoplasms/physiopathology , Membrane Potential, Mitochondrial , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Humans , Lung Neoplasms/diagnostic imaging , Mice , Mice, Transgenic , Organophosphorus Compounds , Positron-Emission Tomography
16.
EMBO J ; 38(22): e101056, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31609012

ABSTRACT

The mitochondrial membrane potential (ΔΨm ) is the main driver of oxidative phosphorylation (OXPHOS). The inner mitochondrial membrane (IMM), consisting of cristae and inner boundary membranes (IBM), is considered to carry a uniform ΔΨm . However, sequestration of OXPHOS components in cristae membranes necessitates a re-examination of the equipotential representation of the IMM. We developed an approach to monitor ΔΨm at the resolution of individual cristae. We found that the IMM was divided into segments with distinct ΔΨm , corresponding to cristae and IBM. ΔΨm was higher at cristae compared to IBM. Treatment with oligomycin increased, whereas FCCP decreased, ΔΨm heterogeneity along the IMM. Impairment of cristae structure through deletion of MICOS-complex components or Opa1 diminished this intramitochondrial heterogeneity of ΔΨm . Lastly, we determined that different cristae within the individual mitochondrion can have disparate membrane potentials and that interventions causing acute depolarization may affect some cristae while sparing others. Altogether, our data support a new model in which cristae within the same mitochondrion behave as independent bioenergetic units, preventing the failure of specific cristae from spreading dysfunction to the rest.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial , Mitochondria/physiology , Mitochondrial Membranes/metabolism , Myoblasts/metabolism , Adenosine Triphosphate/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cells, Cultured , Female , HeLa Cells , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Myoblasts/cytology , Oxidative Phosphorylation
17.
Cell Rep ; 28(7): 1860-1878.e9, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412252

ABSTRACT

Squamous cell carcinoma (SCC), a malignancy arising across multiple anatomical sites, is responsible for significant cancer mortality due to insufficient therapeutic options. Here, we identify exceptional glucose reliance among SCCs dictated by hyperactive GLUT1-mediated glucose influx. Mechanistically, squamous lineage transcription factors p63 and SOX2 transactivate the intronic enhancer cluster of SLC2A1. Elevated glucose influx fuels generation of NADPH and GSH, thereby heightening the anti-oxidative capacity in SCC tumors. Systemic glucose restriction by ketogenic diet and inhibiting renal glucose reabsorption with SGLT2 inhibitor precipitate intratumoral oxidative stress and tumor growth inhibition. Furthermore, reduction of blood glucose lowers blood insulin levels, which suppresses PI3K/AKT signaling in SCC cells. Clinically, we demonstrate a robust correlation between blood glucose concentration and worse survival among SCC patients. Collectively, this study identifies the exceptional glucose reliance of SCC and suggests its candidacy as a highly vulnerable cancer type to be targeted by systemic glucose restriction.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 1/physiology , Glucose/metabolism , Membrane Proteins/metabolism , SOXB1 Transcription Factors/metabolism , AMP-Activated Protein Kinases , Animals , Apoptosis , Carcinoma, Squamous Cell/genetics , Cell Proliferation , Female , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , SOXB1 Transcription Factors/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Cancer Cell ; 35(5): 709-711, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31085172

ABSTRACT

In this issue of Cancer Cell, Elgendy et al. describe the use of intermittent fasting as a strategy to reduce tumor glucose levels and sensitize otherwise resistant tumor cells to metformin. The authors demonstrate that intermittent fasting before metformin treatment sensitized tumors to metformin and significantly reduced tumor growth.


Subject(s)
Hypoglycemia , Metformin , Fasting , Glycogen Synthase Kinase 3 beta , Humans , Myeloid Cell Leukemia Sequence 1 Protein
19.
Cell Metab ; 29(2): 285-302.e7, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30415923

ABSTRACT

AMPK, a conserved sensor of low cellular energy, can either repress or promote tumor growth depending on the context. However, no studies have examined AMPK function in autochthonous genetic mouse models of epithelial cancer. Here, we examine the role of AMPK in murine KrasG12D-mediated non-small-cell lung cancer (NSCLC), a cancer type in humans that harbors frequent inactivating mutations in the LKB1 tumor suppressor-the predominant upstream activating kinase of AMPK and 12 related kinases. Unlike LKB1 deletion, AMPK deletion in KrasG12D lung tumors did not accelerate lung tumor growth. Moreover, deletion of AMPK in KrasG12D p53f/f tumors reduced lung tumor burden. We identified a critical role for AMPK in regulating lysosomal gene expression through the Tfe3 transcription factor, which was required to support NSCLC growth. Thus, AMPK supports the growth of KrasG12D-dependent lung cancer through the induction of lysosomes, highlighting an unrecognized liability of NSCLC.


Subject(s)
AMP-Activated Protein Kinases/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line, Tumor , Loss of Function Mutation , Mice
20.
Sci Transl Med ; 10(467)2018 11 14.
Article in English | MEDLINE | ID: mdl-30429355

ABSTRACT

The diagnostic definition of indeterminate lung nodules as malignant or benign poses a major challenge for clinicians. We discovered a potential marker, the sodium-dependent glucose transporter 2 (SGLT2), whose activity identified metabolically active lung premalignancy and early-stage lung adenocarcinoma (LADC). We found that SGLT2 is expressed early in lung tumorigenesis and is found specifically in premalignant lesions and well-differentiated adenocarcinomas. SGLT2 activity could be detected in vivo by positron emission tomography (PET) with the tracer methyl 4-deoxy-4-[18F] fluoro-alpha-d-glucopyranoside (Me4FDG), which specifically detects SGLT activity. Using a combination of immunohistochemistry and Me4FDG PET, we identified high expression and functional activity of SGLT2 in lung premalignancy and early-stage/low-grade LADC. Furthermore, selective targeting of SGLT2 with FDA-approved small-molecule inhibitors, the gliflozins, greatly reduced tumor growth and prolonged survival in autochthonous mouse models and patient-derived xenografts of LADC. Targeting SGLT2 in lung tumors may intercept lung cancer progression at early stages of development by pairing Me4FDG PET imaging with therapy using SGLT2 inhibitors.


Subject(s)
Adenocarcinoma of Lung/diagnosis , Molecular Targeted Therapy , Sodium-Glucose Transporter 2/metabolism , Adenocarcinoma of Lung/diagnostic imaging , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Aged , Aged, 80 and over , Animals , Biological Transport/drug effects , Cell Differentiation , Cell Proliferation/drug effects , Female , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Humans , Male , Mice, SCID , Mice, Transgenic , Middle Aged , Neoplasm Staging , Positron-Emission Tomography , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Survival Analysis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...