Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38552609

ABSTRACT

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Subject(s)
Neoplasms , Humans , Carcinogenesis , Microbiota , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Obesity/complications , Quality of Life
2.
Cell ; 186(9): 1824-1845, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37116469

ABSTRACT

Cachexia, a systemic wasting condition, is considered a late consequence of diseases, including cancer, organ failure, or infections, and contributes to significant morbidity and mortality. The induction process and mechanistic progression of cachexia are incompletely understood. Refocusing academic efforts away from advanced cachexia to the etiology of cachexia may enable discoveries of new therapeutic approaches. Here, we review drivers, mechanisms, organismal predispositions, evidence for multi-organ interaction, model systems, clinical research, trials, and care provision from early onset to late cachexia. Evidence is emerging that distinct inflammatory, metabolic, and neuro-modulatory drivers can initiate processes that ultimately converge on advanced cachexia.


Subject(s)
Cachexia , Humans , Cachexia/drug therapy , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Muscle, Skeletal/metabolism , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/pathology , Infections/complications , Infections/pathology , Multiple Organ Failure/complications , Multiple Organ Failure/pathology
3.
Cell ; 155(6): 1216-9, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24315093

ABSTRACT

Mouse models for cancer are revealing novel cancer-promoting roles for autophagy. Autophagy promotes tumor growth by suppressing the p53 response, maintaining mitochondrial function, sustaining metabolic homeostasis and survival in stress, and preventing diversion of tumor progression to benign oncocytomas.


Subject(s)
Autophagy , Carcinogenesis , Neoplasms/metabolism , Animals , Humans , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism
4.
Genes Dev ; 34(9-10): 688-700, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32193353

ABSTRACT

Autophagy captures intracellular components and delivers them to lysosomes for degradation and recycling. Conditional autophagy deficiency in adult mice causes liver damage, shortens life span to 3 mo due to neurodegeneration, and is lethal upon fasting. As autophagy deficiency causes p53 induction and cell death in neurons, we sought to test whether p53 mediates the lethal consequences of autophagy deficiency. Here, we conditionally deleted Trp53 (p53 hereafter) and/or the essential autophagy gene Atg7 throughout adult mice. Compared with Atg7Δ/Δ mice, the life span of Atg7Δ/Δp53Δ/Δ mice was extended due to delayed neurodegeneration and resistance to death upon fasting. Atg7 also suppressed apoptosis induced by p53 activator Nutlin-3, suggesting that autophagy inhibited p53 activation. To test whether increased oxidative stress in Atg7Δ/Δ mice was responsible for p53 activation, Atg7 was deleted in the presence or absence of the master regulator of antioxidant defense nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2-/-Atg7Δ/Δ mice died rapidly due to small intestine damage, which was not rescued by p53 codeletion. Thus, Atg7 limits p53 activation and p53-mediated neurodegeneration. In turn, NRF2 mitigates lethal intestine degeneration upon autophagy loss. These findings illustrate the tissue-specific roles for autophagy and functional dependencies on the p53 and NRF2 stress response mechanisms.


Subject(s)
Autophagy/genetics , Longevity/genetics , Oxidative Stress/genetics , Tumor Suppressor Protein p53/genetics , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Gene Deletion , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/metabolism
5.
Genes Dev ; 33(11-12): 610-619, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31160394

ABSTRACT

Macroautophagy (referred to here as autophagy) degrades and recycles cytoplasmic constituents to sustain cellular and mammalian metabolism and survival during starvation. Deregulation of autophagy is involved in numerous diseases, such as cancer. Cancers up-regulate autophagy and depend on it for survival, growth, and malignancy in a tumor cell-autonomous fashion. Recently, it has become apparent that autophagy in host tissues as well as the tumor cells themselves contribute to tumor growth. Understanding how autophagy regulates metabolism and tumor growth has revealed new essential tumor nutrients, where they come from, and how they are supplied and used, which can now be targeted for cancer therapy.


Subject(s)
Autophagy , Neoplasms/metabolism , Animals , Autophagy/genetics , Humans , Neoplasms/physiopathology , Neoplasms/therapy
6.
Mol Cell ; 70(3): 502-515.e8, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29727620

ABSTRACT

Nutrients are not only organic compounds fueling bioenergetics and biosynthesis, but also key chemical signals controlling growth and metabolism. Nutrients enormously impact the production of reactive oxygen species (ROS), which play essential roles in normal physiology and diseases. How nutrient signaling is integrated with redox regulation is an interesting, but not fully understood, question. Herein, we report that superoxide dismutase 1 (SOD1) is a conserved component of the mechanistic target of rapamycin complex 1 (mTORC1) nutrient signaling. mTORC1 regulates SOD1 activity through reversible phosphorylation at S39 in yeast and T40 in humans in response to nutrients, which moderates ROS level and prevents oxidative DNA damage. We further show that SOD1 activation enhances cancer cell survival and tumor formation in the ischemic tumor microenvironment and protects against the chemotherapeutic agent cisplatin. Collectively, these findings identify a conserved mechanism by which eukaryotes dynamically regulate redox homeostasis in response to changing nutrient conditions.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Nutrients/metabolism , Phosphorylation/physiology , Superoxide Dismutase-1/metabolism , Animals , Cell Line , Cell Line, Tumor , DNA Damage/physiology , Energy Metabolism/physiology , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism
8.
Nature ; 565(7737): E3, 2019 01.
Article in English | MEDLINE | ID: mdl-30523330

ABSTRACT

In this Letter, 'released' should have been 'regulated' in the sentence starting: 'Deletion of Atg5 in the host similarly regulated circulating arginine and suppressed tumorigenesis...' This has been corrected online.

9.
Proc Natl Acad Sci U S A ; 119(21): e2202016119, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35537042

ABSTRACT

Autophagy defects are a risk factor for inflammatory bowel diseases (IBDs) through unknown mechanisms. Whole-body conditional deletion of autophagy-related gene (Atg) Atg7 in adult mice (Atg7Δ/Δ) causes tissue damage and death within 3 mo due to neurodegeneration without substantial effect on intestine. In contrast, we report here that whole-body conditional deletion of other essential Atg genes Atg5 or Fip200/Atg17 in adult mice (Atg5Δ/Δ or Fip200Δ/Δ) caused death within 5 d due to rapid autophagy inhibition, elimination of ileum stem cells, and loss of barrier function. Atg5Δ/Δ mice lost PDGFRα+ mesenchymal cells (PMCs) and Wnt signaling essential for stem cell renewal, which were partially rescued by exogenous Wnt. Matrix-assisted laser desorption ionization coupled to mass spectrometry imaging (MALDI-MSI) of Atg5Δ/Δ ileum revealed depletion of aspartate and nucleotides, consistent with metabolic insufficiency underlying PMC loss. The difference in the autophagy gene knockout phenotypes is likely due to distinct kinetics of autophagy loss, as deletion of Atg5 more gradually extended lifespan phenocopying deletion of Atg7 or Atg12. Thus, autophagy is required for PMC metabolism and ileum stem cell and mammalian survival. Failure to maintain PMCs through autophagy may therefore contribute to IBD.


Subject(s)
Autophagy , Intestines , Receptor, Platelet-Derived Growth Factor alpha , Stem Cells , Animals , Autophagy/genetics , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Cell Survival , Mice , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stem Cells/metabolism
10.
PLoS Genet ; 18(4): e1010138, 2022 04.
Article in English | MEDLINE | ID: mdl-35404932

ABSTRACT

The PALB2 tumor suppressor plays key roles in DNA repair and has been implicated in redox homeostasis. Autophagy maintains mitochondrial quality, mitigates oxidative stress and suppresses neurodegeneration. Here we show that Palb2 deletion in the mouse brain leads to mild motor deficits and that co-deletion of Palb2 with the essential autophagy gene Atg7 accelerates and exacerbates neurodegeneration induced by ATG7 loss. Palb2 deletion leads to elevated DNA damage, oxidative stress and mitochondrial markers, especially in Purkinje cells, and co-deletion of Palb2 and Atg7 results in accelerated Purkinje cell loss. Further analyses suggest that the accelerated Purkinje cell loss and severe neurodegeneration in the double deletion mice are due to excessive oxidative stress and mitochondrial dysfunction, rather than DNA damage, and partially dependent on p53 activity. Our studies uncover a role of PALB2 in mitochondrial homeostasis and a cooperation between PALB2 and ATG7/autophagy in maintaining redox and mitochondrial homeostasis essential for neuronal survival.


Subject(s)
Autophagy , Mitochondria , Animals , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Brain/metabolism , Fanconi Anemia Complementation Group N Protein , Homeostasis/genetics , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oxidation-Reduction
11.
Cell ; 137(6): 1062-75, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524509

ABSTRACT

Allelic loss of the essential autophagy gene beclin1 occurs in human cancers and renders mice tumor-prone suggesting that autophagy is a tumor-suppression mechanism. While tumor cells utilize autophagy to survive metabolic stress, autophagy also mitigates the resulting cellular damage that may limit tumorigenesis. In response to stress, autophagy-defective tumor cells preferentially accumulated p62/SQSTM1 (p62), endoplasmic reticulum (ER) chaperones, damaged mitochondria, reactive oxygen species (ROS), and genome damage. Moreover, suppressing ROS or p62 accumulation prevented damage resulting from autophagy defects indicating that failure to regulate p62 caused oxidative stress. Importantly, sustained p62 expression resulting from autophagy defects was sufficient to alter NF-kappaB regulation and gene expression and to promote tumorigenesis. Thus, defective autophagy is a mechanism for p62 upregulation commonly observed in human tumors that contributes directly to tumorigenesis likely by perturbing the signal transduction adaptor function of p62-controlling pathways critical for oncogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Neoplasms/metabolism , Aneuploidy , Animals , Apoptosis , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Mice , Mitochondria/metabolism , Molecular Chaperones/metabolism , NF-kappa B/metabolism , Neoplasms/genetics , Oxidative Stress , Protein Disulfide-Isomerases/metabolism , Sequestosome-1 Protein , Transcription Factor TFIIH , Transcription Factors
12.
Nature ; 563(7732): 569-573, 2018 11.
Article in English | MEDLINE | ID: mdl-30429607

ABSTRACT

Autophagy captures intracellular components and delivers them to lysosomes, where they are degraded and recycled to sustain metabolism and to enable survival during starvation1-5. Acute, whole-body deletion of the essential autophagy gene Atg7 in adult mice causes a systemic metabolic defect that manifests as starvation intolerance and gradual loss of white adipose tissue, liver glycogen and muscle mass1. Cancer cells also benefit from autophagy. Deletion of essential autophagy genes impairs the metabolism, proliferation, survival and malignancy of spontaneous tumours in models of autochthonous cancer6,7. Acute, systemic deletion of Atg7 or acute, systemic expression of a dominant-negative ATG4b in mice induces greater regression of KRAS-driven cancers than does tumour-specific autophagy deletion, which suggests that host autophagy promotes tumour growth1,8. Here we show that host-specific deletion of Atg7 impairs the growth of multiple allografted tumours, although not all tumour lines were sensitive to host autophagy status. Loss of autophagy in the host was associated with a reduction in circulating arginine, and the sensitive tumour cell lines were arginine auxotrophs owing to the lack of expression of the enzyme argininosuccinate synthase 1. Serum proteomic analysis identified the arginine-degrading enzyme arginase I (ARG1) in the circulation of Atg7-deficient hosts, and in vivo arginine metabolic tracing demonstrated that serum arginine was degraded to ornithine. ARG1 is predominantly expressed in the liver and can be released from hepatocytes into the circulation. Liver-specific deletion of Atg7 produced circulating ARG1, and reduced both serum arginine and tumour growth. Deletion of Atg5 in the host similarly regulated [corrected] circulating arginine and suppressed tumorigenesis, which demonstrates that this phenotype is specific to autophagy function rather than to deletion of Atg7. Dietary supplementation of Atg7-deficient hosts with arginine partially restored levels of circulating arginine and tumour growth. Thus, defective autophagy in the host leads to the release of ARG1 from the liver and the degradation of circulating arginine, which is essential for tumour growth; this identifies a metabolic vulnerability of cancer.


Subject(s)
Arginine/blood , Autophagy , Neoplasms/blood , Neoplasms/pathology , Allografts , Animals , Arginase/blood , Arginase/metabolism , Arginine/administration & dosage , Arginine/pharmacology , Autophagy/genetics , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 7/deficiency , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dietary Supplements , Hepatocytes/enzymology , Hepatocytes/metabolism , Liver/enzymology , Male , Mice , Neoplasm Transplantation , Neoplasms/genetics , Ornithine/metabolism
13.
Mol Cell ; 61(5): 667-676, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26942671

ABSTRACT

Decades ago, Otto Warburg observed that cancers ferment glucose in the presence of oxygen, suggesting that defects in mitochondrial respiration may be the underlying cause of cancer. We now know that the genetic events that drive aberrant cancer cell proliferation also alter biochemical metabolism, including promoting aerobic glycolysis, but do not typically impair mitochondrial function. Mitochondria supply energy; provide building blocks for new cells; and control redox homeostasis, oncogenic signaling, innate immunity, and apoptosis. Indeed, mitochondrial biogenesis and quality control are often upregulated in cancers. While some cancers have mutations in nuclear-encoded mitochondrial tricarboxylic acid (TCA) cycle enzymes that produce oncogenic metabolites, there is negative selection for pathogenic mitochondrial genome mutations. Eliminating mtDNA limits tumorigenesis, and rare human tumors with mutant mitochondrial genomes are relatively benign. Thus, mitochondria play a central and multifunctional role in malignant tumor progression, and targeting mitochondria provides therapeutic opportunities.


Subject(s)
Energy Metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Genetic Predisposition to Disease , Humans , Mitochondria/drug effects , Mitochondria/pathology , Molecular Targeted Therapy , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction
14.
Genes Dev ; 30(17): 1913-30, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27664235

ABSTRACT

Macroautophagy (referred to here as autophagy) is induced by starvation to capture and degrade intracellular proteins and organelles in lysosomes, which recycles intracellular components to sustain metabolism and survival. Autophagy also plays a major homeostatic role in controlling protein and organelle quality and quantity. Dysfunctional autophagy contributes to many diseases. In cancer, autophagy can be neutral, tumor-suppressive, or tumor-promoting in different contexts. Large-scale genomic analysis of human cancers indicates that the loss or mutation of core autophagy genes is uncommon, whereas oncogenic events that activate autophagy and lysosomal biogenesis have been identified. Autophagic flux, however, is difficult to measure in human tumor samples, making functional assessment of autophagy problematic in a clinical setting. Autophagy impacts cellular metabolism, the proteome, and organelle numbers and quality, which alter cell functions in diverse ways. Moreover, autophagy influences the interaction between the tumor and the host by promoting stress adaptation and suppressing activation of innate and adaptive immune responses. Additionally, autophagy can promote a cross-talk between the tumor and the stroma, which can support tumor growth, particularly in a nutrient-limited microenvironment. Thus, the role of autophagy in cancer is determined by nutrient availability, microenvironment stress, and the presence of an immune system. Here we discuss recent developments in the role of autophagy in cancer, in particular how autophagy can promote cancer through suppressing p53 and preventing energy crisis, cell death, senescence, and an anti-tumor immune response.


Subject(s)
Autophagy/physiology , Gene Expression Regulation, Neoplastic , Neoplasms/physiopathology , Animals , Autophagy/genetics , Disease Models, Animal , Drug Delivery Systems , Humans , Neoplasms/genetics
15.
Genes Dev ; 30(4): 399-407, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26883359

ABSTRACT

Understanding new therapeutic paradigms for both castrate-sensitive and more aggressive castrate-resistant prostate cancer is essential to improve clinical outcomes. As a critically important cellular process, autophagy promotes stress tolerance by recycling intracellular components to sustain metabolism important for tumor survival. To assess the importance of autophagy in prostate cancer, we generated a new autochthonous genetically engineered mouse model (GEMM) with inducible prostate-specific deficiency in the Pten tumor suppressor and autophagy-related-7 (Atg7) genes. Atg7 deficiency produced an autophagy-deficient phenotype and delayed Pten-deficient prostate tumor progression in both castrate-naïve and castrate-resistant cancers. Atg7-deficient tumors display evidence of endoplasmic reticulum (ER) stress, suggesting that autophagy may promote prostate tumorigenesis through management of protein homeostasis. Taken together, these data support the importance of autophagy for both castrate-naïve and castrate-resistant growth in a newly developed GEMM, suggesting a new paradigm and model to study approaches to inhibit autophagy in combination with known and new therapies for advanced prostate cancer.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Microtubule-Associated Proteins/metabolism , Prostatic Neoplasms/physiopathology , Animals , Animals, Genetically Modified , Autophagy/genetics , Autophagy-Related Protein 7 , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Gene Deletion , Male , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/physiopathology , Signal Transduction/genetics
16.
Genes Dev ; 30(15): 1704-17, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27516533

ABSTRACT

Autophagy degrades and is thought to recycle proteins, other macromolecules, and organelles. In genetically engineered mouse models (GEMMs) for Kras-driven lung cancer, autophagy prevents the accumulation of defective mitochondria and promotes malignancy. Autophagy-deficient tumor-derived cell lines are respiration-impaired and starvation-sensitive. However, to what extent their sensitivity to starvation arises from defective mitochondria or an impaired supply of metabolic substrates remains unclear. Here, we sequenced the mitochondrial genomes of wild-type or autophagy-deficient (Atg7(-/-)) Kras-driven lung tumors. Although Atg7 deletion resulted in increased mitochondrial mutations, there were too few nonsynonymous mutations to cause generalized mitochondrial dysfunction. In contrast, pulse-chase studies with isotope-labeled nutrients revealed impaired mitochondrial substrate supply during starvation of the autophagy-deficient cells. This was associated with increased reactive oxygen species (ROS), lower energy charge, and a dramatic drop in total nucleotide pools. While starvation survival of the autophagy-deficient cells was not rescued by the general antioxidant N-acetyl-cysteine, it was fully rescued by glutamine or glutamate (both amino acids that feed the TCA cycle and nucleotide synthesis) or nucleosides. Thus, maintenance of nucleotide pools is a critical challenge for starving Kras-driven tumor cells. By providing bioenergetic and biosynthetic substrates, autophagy supports nucleotide pools and thereby starvation survival.


Subject(s)
Autophagy , Lung Neoplasms/metabolism , Nucleotides/metabolism , ras Proteins/metabolism , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Cell Line, Tumor , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Deletion , Genetic Variation , Genome, Mitochondrial/genetics , Glutamine/pharmacology , Lung Neoplasms/physiopathology , Mice , Mitochondria/metabolism , Nucleosides/pharmacology , Oxidation-Reduction
17.
Nature ; 551(7678): 115-118, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29045397

ABSTRACT

Mammalian tissues are fuelled by circulating nutrients, including glucose, amino acids, and various intermediary metabolites. Under aerobic conditions, glucose is generally assumed to be burned fully by tissues via the tricarboxylic acid cycle (TCA cycle) to carbon dioxide. Alternatively, glucose can be catabolized anaerobically via glycolysis to lactate, which is itself also a potential nutrient for tissues and tumours. The quantitative relevance of circulating lactate or other metabolic intermediates as fuels remains unclear. Here we systematically examine the fluxes of circulating metabolites in mice, and find that lactate can be a primary source of carbon for the TCA cycle and thus of energy. Intravenous infusions of 13C-labelled nutrients reveal that, on a molar basis, the circulatory turnover flux of lactate is the highest of all metabolites and exceeds that of glucose by 1.1-fold in fed mice and 2.5-fold in fasting mice; lactate is made primarily from glucose but also from other sources. In both fed and fasted mice, 13C-lactate extensively labels TCA cycle intermediates in all tissues. Quantitative analysis reveals that during the fasted state, the contribution of glucose to tissue TCA metabolism is primarily indirect (via circulating lactate) in all tissues except the brain. In genetically engineered lung and pancreatic cancer tumours in fasted mice, the contribution of circulating lactate to TCA cycle intermediates exceeds that of glucose, with glutamine making a larger contribution than lactate in pancreatic cancer. Thus, glycolysis and the TCA cycle are uncoupled at the level of lactate, which is a primary circulating TCA substrate in most tissues and tumours.


Subject(s)
Citric Acid Cycle , Glucose/metabolism , Lactic Acid/blood , Lactic Acid/metabolism , Animals , Blood Glucose/metabolism , Brain/metabolism , Carbon/blood , Carbon/metabolism , Fasting/blood , Fasting/metabolism , Glutamine/blood , Glutamine/metabolism , Glycolysis , Mice , Muscles/metabolism , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/metabolism
18.
Genes Dev ; 29(19): 2010-21, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26404941

ABSTRACT

An unbalanced karyotype, a condition known as aneuploidy, has a profound impact on cellular physiology and is a hallmark of cancer. Aneuploid cells experience a number of stresses that are caused by aneuploidy-induced proteomic changes. How the aneuploidy-associated stresses affect cells and whether cells respond to them are only beginning to be understood. Here we show that autophagosomal cargo such as protein aggregates accumulate within lysosomes in aneuploid cells. This causes a lysosomal stress response. Aneuploid cells activate the transcription factor TFEB, a master regulator of autophagic and lysosomal gene expression, thereby increasing the expression of genes needed for autophagy-mediated protein degradation. Accumulation of autophagic cargo within the lysosome and activation of TFEB-responsive genes are also observed in cells in which proteasome function is inhibited, suggesting that proteotoxic stress causes TFEB activation. Our results reveal a TFEB-mediated lysosomal stress response as a universal feature of the aneuploid state.


Subject(s)
Aneuploidy , Autophagy/genetics , Lysosomes/pathology , Stress, Physiological/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line , Gene Expression Regulation , Humans , Proteasome Endopeptidase Complex , Protein Aggregates/physiology , Protein Folding , Proteolysis
19.
Mol Cell ; 55(6): 916-930, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25175026

ABSTRACT

Ras-driven cancer cells upregulate basal autophagy that degrades and recycles intracellular proteins and organelles. Autophagy-mediated proteome degradation provides free amino acids to support metabolism and macromolecular synthesis, which confers a survival advantage in starvation and promotes tumorigenesis. While the degradation of isolated protein substrates by autophagy has been implicated in controlling cellular function, the extent and specificity by which autophagy remodels the cellular proteome and the underlying functional consequences were unknown. Here we compared the global proteome of autophagy-functional and -deficient Ras-driven cancer cells, finding that autophagy affects the majority of the proteome yet is highly selective. While levels of vesicle trafficking proteins important for autophagy are preserved during starvation-induced autophagy, deleterious inflammatory response pathway components are eliminated even under basal conditions, preventing cytokine-induced paracrine cell death. This reveals the global, functional impact of autophagy-mediated proteome remodeling on cell survival and identifies critical autophagy substrates that mediate this process.


Subject(s)
Autophagy , Immunity, Innate , Proteome/physiology , ras Proteins/genetics , Animals , Cell Line, Tumor , Cell Survival , Humans , Mice , Protein Transport , Transport Vesicles
20.
Genes Dev ; 27(19): 2065-71, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24115766

ABSTRACT

Oncogenic Ras promotes glucose fermentation and glutamine use to supply central carbon metabolism, but how and why have only emerged recently. Ras-mediated metabolic reprogramming generates building blocks for growth and promotes antioxidant defense. To fuel metabolic pathways, Ras scavenges extracellular proteins and lipids. To bolster metabolism and mitigate stress, Ras activates cellular self-cannibalization and recycling of proteins and organelles by autophagy. Targeting these distinct features of Ras-driven cancers provides novel approaches to cancer therapy.


Subject(s)
Metabolic Networks and Pathways , Neoplasms/metabolism , ras Proteins/metabolism , Animals , Autophagy , Glucose/metabolism , Glutamine/metabolism , Humans , Lipid Metabolism , Neoplasms/genetics , Neoplasms/therapy , Proteins/metabolism , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL