Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 410
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38843833

ABSTRACT

While ultraviolet (UV) radiation damages DNA, eliciting the DNA damage response (DDR), it also damages RNA, triggering transcriptome-wide ribosomal collisions and eliciting a ribotoxic stress response (RSR). However, the relative contributions, timing, and regulation of these pathways in determining cell fate is unclear. Here we use time-resolved phosphoproteomic, chemical-genetic, single-cell imaging, and biochemical approaches to create a chronological atlas of signaling events activated in cells responding to UV damage. We discover that UV-induced apoptosis is mediated by the RSR kinase ZAK and not through the DDR. We identify two negative-feedback modules that regulate ZAK-mediated apoptosis: (1) GCN2 activation limits ribosomal collisions and attenuates ZAK-mediated RSR and (2) ZAK activity leads to phosphodegron autophosphorylation and its subsequent degradation. These events tune ZAK's activity to collision levels to establish regimes of homeostasis, tolerance, and death, revealing its key role as the cellular sentinel for nucleic acid damage.

2.
Cell ; 186(18): 3945-3967.e26, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37582358

ABSTRACT

Post-translational modifications (PTMs) play key roles in regulating cell signaling and physiology in both normal and cancer cells. Advances in mass spectrometry enable high-throughput, accurate, and sensitive measurement of PTM levels to better understand their role, prevalence, and crosstalk. Here, we analyze the largest collection of proteogenomics data from 1,110 patients with PTM profiles across 11 cancer types (10 from the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium [CPTAC]). Our study reveals pan-cancer patterns of changes in protein acetylation and phosphorylation involved in hallmark cancer processes. These patterns revealed subsets of tumors, from different cancer types, including those with dysregulated DNA repair driven by phosphorylation, altered metabolic regulation associated with immune response driven by acetylation, affected kinase specificity by crosstalk between acetylation and phosphorylation, and modified histone regulation. Overall, this resource highlights the rich biology governed by PTMs and exposes potential new therapeutic avenues.


Subject(s)
Neoplasms , Protein Processing, Post-Translational , Proteomics , Humans , Acetylation , Histones/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Proteomics/methods
3.
Cell ; 186(18): 3921-3944.e25, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37582357

ABSTRACT

Cancer driver events refer to key genetic aberrations that drive oncogenesis; however, their exact molecular mechanisms remain insufficiently understood. Here, our multi-omics pan-cancer analysis uncovers insights into the impacts of cancer drivers by identifying their significant cis-effects and distal trans-effects quantified at the RNA, protein, and phosphoprotein levels. Salient observations include the association of point mutations and copy-number alterations with the rewiring of protein interaction networks, and notably, most cancer genes converge toward similar molecular states denoted by sequence-based kinase activity profiles. A correlation between predicted neoantigen burden and measured T cell infiltration suggests potential vulnerabilities for immunotherapies. Patterns of cancer hallmarks vary by polygenic protein abundance ranging from uniform to heterogeneous. Overall, our work demonstrates the value of comprehensive proteogenomics in understanding the functional states of oncogenic drivers and their links to cancer development, surpassing the limitations of studying individual cancer types.


Subject(s)
Neoplasms , Proteogenomics , Humans , Neoplasms/genetics , Oncogenes , Cell Transformation, Neoplastic/genetics , DNA Copy Number Variations
4.
Cell ; 174(6): 1347-1360, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193109

ABSTRACT

Chromosomal instability (CIN) is a hallmark of human cancer, and it is associated with poor prognosis, metastasis, and therapeutic resistance. CIN results from errors in chromosome segregation during mitosis, leading to structural and numerical chromosomal abnormalities. In addition to generating genomic heterogeneity that acts as a substrate for natural selection, CIN promotes inflammatory signaling by introducing double-stranded DNA into the cytosol, engaging the cGAS-STING anti-viral pathway. These multipronged effects distinguish CIN as a central driver of tumor evolution and as a genomic source for the crosstalk between the tumor and its microenvironment, in the course of immune editing and evasion.


Subject(s)
Chromosomal Instability , Aneuploidy , Humans , Immunity, Innate , Interferon Type I/metabolism , Membrane Proteins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Nucleotidyltransferases/metabolism , Signal Transduction , Tumor Microenvironment
5.
Cell ; 175(6): 1546-1560.e17, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30500537

ABSTRACT

Mammalian folate metabolism is comprised of cytosolic and mitochondrial pathways with nearly identical core reactions, yet the functional advantages of such an organization are not well understood. Using genome-editing and biochemical approaches, we find that ablating folate metabolism in the mitochondria of mammalian cell lines results in folate degradation in the cytosol. Mechanistically, we show that QDPR, an enzyme in tetrahydrobiopterin metabolism, moonlights to repair oxidative damage to tetrahydrofolate (THF). This repair capacity is overwhelmed when cytosolic THF hyperaccumulates in the absence of mitochondrially produced formate, leading to THF degradation. Unexpectedly, we also find that the classic antifolate methotrexate, by inhibiting its well-known target DHFR, causes even more extensive folate degradation in nearly all tested cancer cell lines. These findings shed light on design features of folate metabolism, provide a biochemical basis for clinically observed folate deficiency in QDPR-deficient patients, and reveal a hitherto unknown and unexplored cellular effect of methotrexate.


Subject(s)
Carbon/metabolism , Cytosol/metabolism , Formates/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Tetrahydrofolates/metabolism , Cytosol/pathology , HCT116 Cells , HeLa Cells , Humans , MCF-7 Cells , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Tetrahydrofolate Dehydrogenase/metabolism
6.
Cell ; 170(4): 605-635, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28802037

ABSTRACT

Phosphoinositide 3-kinase (PI3K) activity is stimulated by diverse oncogenes and growth factor receptors, and elevated PI3K signaling is considered a hallmark of cancer. Many PI3K pathway-targeted therapies have been tested in oncology trials, resulting in regulatory approval of one isoform-selective inhibitor (idelalisib) for treatment of certain blood cancers and a variety of other agents at different stages of development. In parallel to PI3K research by cancer biologists, investigations in other fields have uncovered exciting and often unpredicted roles for PI3K catalytic and regulatory subunits in normal cell function and in disease. Many of these functions impinge upon oncology by influencing the efficacy and toxicity of PI3K-targeted therapies. Here we provide a perspective on the roles of class I PI3Ks in the regulation of cellular metabolism and in immune system functions, two topics closely intertwined with cancer biology. We also discuss recent progress developing PI3K-targeted therapies for treatment of cancer and other diseases.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Cell Physiological Phenomena , Humans , Immune System/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
7.
Cell ; 171(7): 1545-1558.e18, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29153836

ABSTRACT

mTORC1 is a signal integrator and master regulator of cellular anabolic processes linked to cell growth and survival. Here, we demonstrate that mTORC1 promotes lipid biogenesis via SRPK2, a key regulator of RNA-binding SR proteins. mTORC1-activated S6K1 phosphorylates SRPK2 at Ser494, which primes Ser497 phosphorylation by CK1. These phosphorylation events promote SRPK2 nuclear translocation and phosphorylation of SR proteins. Genome-wide transcriptome analysis reveals that lipid biosynthetic enzymes are among the downstream targets of mTORC1-SRPK2 signaling. Mechanistically, SRPK2 promotes SR protein binding to U1-70K to induce splicing of lipogenic pre-mRNAs. Inhibition of this signaling pathway leads to intron retention of lipogenic genes, which triggers nonsense-mediated mRNA decay. Genetic or pharmacological inhibition of SRPK2 blunts de novo lipid synthesis, thereby suppressing cell growth. These results thus reveal a novel role of mTORC1-SRPK2 signaling in post-transcriptional regulation of lipid metabolism and demonstrate that SRPK2 is a potential therapeutic target for mTORC1-driven metabolic disorders.


Subject(s)
Gene Expression Regulation , Lipogenesis , RNA Processing, Post-Transcriptional , Signal Transduction , Animals , Cell Nucleus/metabolism , Cholesterol/metabolism , Fatty Acids/metabolism , Female , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
9.
Cell ; 164(3): 433-46, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824656

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway regulates multiple steps in glucose metabolism and also cytoskeletal functions, such as cell movement and attachment. Here, we show that PI3K directly coordinates glycolysis with cytoskeletal dynamics in an AKT-independent manner. Growth factors or insulin stimulate the PI3K-dependent activation of Rac, leading to disruption of the actin cytoskeleton, release of filamentous actin-bound aldolase A, and an increase in aldolase activity. Consistently, PI3K inhibitors, but not AKT, SGK, or mTOR inhibitors, cause a significant decrease in glycolysis at the step catalyzed by aldolase, while activating PIK3CA mutations have the opposite effect. These results point toward a master regulatory function of PI3K that integrates an epithelial cell's metabolism and its form, shape, and function, coordinating glycolysis with the energy-intensive dynamics of actin remodeling.


Subject(s)
Fructose-Bisphosphate Aldolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cytoskeleton/metabolism , Cytosol/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Glycolysis , Humans , Insulin/metabolism , Mice , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction
10.
Nature ; 629(8014): 1174-1181, 2024 May.
Article in English | MEDLINE | ID: mdl-38720073

ABSTRACT

Phosphorylation of proteins on tyrosine (Tyr) residues evolved in metazoan organisms as a mechanism of coordinating tissue growth1. Multicellular eukaryotes typically have more than 50 distinct protein Tyr kinases that catalyse the phosphorylation of thousands of Tyr residues throughout the proteome1-3. How a given Tyr kinase can phosphorylate a specific subset of proteins at unique Tyr sites is only partially understood4-7. Here we used combinatorial peptide arrays to profile the substrate sequence specificity of all human Tyr kinases. Globally, the Tyr kinases demonstrate considerable diversity in optimal patterns of residues surrounding the site of phosphorylation, revealing the functional organization of the human Tyr kinome by substrate motif preference. Using this information, Tyr kinases that are most compatible with phosphorylating any Tyr site can be identified. Analysis of mass spectrometry phosphoproteomic datasets using this compendium of kinase specificities accurately identifies specific Tyr kinases that are dysregulated in cells after stimulation with growth factors, treatment with anti-cancer drugs or expression of oncogenic variants. Furthermore, the topology of known Tyr signalling networks naturally emerged from a comparison of the sequence specificities of the Tyr kinases and the SH2 phosphotyrosine (pTyr)-binding domains. Finally we show that the intrinsic substrate specificity of Tyr kinases has remained fundamentally unchanged from worms to humans, suggesting that the fidelity between Tyr kinases and their protein substrate sequences has been maintained across hundreds of millions of years of evolution.


Subject(s)
Phosphotyrosine , Protein-Tyrosine Kinases , Substrate Specificity , Tyrosine , Animals , Humans , Amino Acid Motifs , Evolution, Molecular , Mass Spectrometry , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/metabolism , Proteome/chemistry , Proteome/metabolism , Proteomics , Signal Transduction , src Homology Domains , Tyrosine/metabolism , Tyrosine/chemistry
11.
Cell ; 159(7): 1492-4, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525870

ABSTRACT

Cancer cells have distinctive nutrient demands to fuel growth and proliferation, including the disproportionate use of glucose, glutamine, and fatty acids. Comerford et al. and Mashimo et al. now demonstrate that several types of cancer are avid consumers of acetate, which facilitates macromolecular biosynthesis and histone modification.

12.
Cell ; 156(4): 771-85, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24529379

ABSTRACT

mTORC1 promotes cell growth in response to nutrients and growth factors. Insulin activates mTORC1 through the PI3K-Akt pathway, which inhibits the TSC1-TSC2-TBC1D7 complex (the TSC complex) to turn on Rheb, an essential activator of mTORC1. However, the mechanistic basis of how this pathway integrates with nutrient-sensing pathways is unknown. We demonstrate that insulin stimulates acute dissociation of the TSC complex from the lysosomal surface, where subpopulations of Rheb and mTORC1 reside. The TSC complex associates with the lysosome in a Rheb-dependent manner, and its dissociation in response to insulin requires Akt-mediated TSC2 phosphorylation. Loss of the PTEN tumor suppressor results in constitutive activation of mTORC1 through the Akt-dependent dissociation of the TSC complex from the lysosome. These findings provide a unifying mechanism by which independent pathways affecting the spatial recruitment of mTORC1 and the TSC complex to Rheb at the lysosomal surface serve to integrate diverse growth signals.


Subject(s)
Insulin/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acids/metabolism , Animals , Cell Line , GTP Phosphohydrolases/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
13.
Cell ; 158(6): 1309-1323, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215489

ABSTRACT

The balance between oxidative and nonoxidative glucose metabolism is essential for a number of pathophysiological processes. By deleting enzymes that affect aerobic glycolysis with different potencies, we examine how modulating glucose metabolism specifically affects hematopoietic and leukemic cell populations. We find that a deficiency in the M2 pyruvate kinase isoform (PKM2) reduces the levels of metabolic intermediates important for biosynthesis and impairs progenitor function without perturbing hematopoietic stem cells (HSCs), whereas lactate dehydrogenase A (LDHA) deletion significantly inhibits the function of both HSCs and progenitors during hematopoiesis. In contrast, leukemia initiation by transforming alleles putatively affecting either HSCs or progenitors is inhibited in the absence of either PKM2 or LDHA, indicating that the cell-state-specific responses to metabolic manipulation in hematopoiesis do not apply to the setting of leukemia. This finding suggests that fine-tuning the level of glycolysis may be explored therapeutically for treating leukemia while preserving HSC function.


Subject(s)
Glycolysis , Hematopoiesis , Leukemia/metabolism , Animals , Gene Deletion , Hematopoietic Stem Cells/metabolism , Humans , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Mice , Mice, Congenic , Mice, Inbred C57BL , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
14.
Nature ; 617(7959): 147-153, 2023 05.
Article in English | MEDLINE | ID: mdl-36949200

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is characterized by aggressive local invasion and metastatic spread, leading to high lethality. Although driver gene mutations during PDA progression are conserved, no specific mutation is correlated with the dissemination of metastases1-3. Here we analysed RNA splicing data of a large cohort of primary and metastatic PDA tumours to identify differentially spliced events that correlate with PDA progression. De novo motif analysis of these events detected enrichment of motifs with high similarity to the RBFOX2 motif. Overexpression of RBFOX2 in a patient-derived xenograft (PDX) metastatic PDA cell line drastically reduced the metastatic potential of these cells in vitro and in vivo, whereas depletion of RBFOX2 in primary pancreatic tumour cell lines increased the metastatic potential of these cells. These findings support the role of RBFOX2 as a potent metastatic suppressor in PDA. RNA-sequencing and splicing analysis of RBFOX2 target genes revealed enrichment of genes in the RHO GTPase pathways, suggesting a role of RBFOX2 splicing activity in cytoskeletal organization and focal adhesion formation. Modulation of RBFOX2-regulated splicing events, such as via myosin phosphatase RHO-interacting protein (MPRIP), is associated with PDA metastases, altered cytoskeletal organization and the induction of focal adhesion formation. Our results implicate the splicing-regulatory function of RBFOX2 as a tumour suppressor in PDA and suggest a therapeutic approach for metastatic PDA.


Subject(s)
Alternative Splicing , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Alternative Splicing/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Animals , Neoplasm Metastasis , Focal Adhesions
15.
Nature ; 613(7945): 759-766, 2023 01.
Article in English | MEDLINE | ID: mdl-36631611

ABSTRACT

Protein phosphorylation is one of the most widespread post-translational modifications in biology1,2. With advances in mass-spectrometry-based phosphoproteomics, 90,000 sites of serine and threonine phosphorylation have so far been identified, and several thousand have been associated with human diseases and biological processes3,4. For the vast majority of phosphorylation events, it is not yet known which of the more than 300 protein serine/threonine (Ser/Thr) kinases encoded in the human genome are responsible3. Here we used synthetic peptide libraries to profile the substrate sequence specificity of 303 Ser/Thr kinases, comprising more than 84% of those predicted to be active in humans. Viewed in its entirety, the substrate specificity of the kinome was substantially more diverse than expected and was driven extensively by negative selectivity. We used our kinome-wide dataset to computationally annotate and identify the kinases capable of phosphorylating every reported phosphorylation site in the human Ser/Thr phosphoproteome. For the small minority of phosphosites for which the putative protein kinases involved have been previously reported, our predictions were in excellent agreement. When this approach was applied to examine the signalling response of tissues and cell lines to hormones, growth factors, targeted inhibitors and environmental or genetic perturbations, it revealed unexpected insights into pathway complexity and compensation. Overall, these studies reveal the intrinsic substrate specificity of the human Ser/Thr kinome, illuminate cellular signalling responses and provide a resource to link phosphorylation events to biological pathways.


Subject(s)
Phosphoproteins , Protein Serine-Threonine Kinases , Proteome , Serine , Threonine , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Substrate Specificity , Threonine/metabolism , Proteome/chemistry , Proteome/metabolism , Datasets as Topic , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Cell Line , Phosphoserine/metabolism , Phosphothreonine/metabolism
16.
Cell ; 155(4): 778-92, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209617

ABSTRACT

Regeneration capacity declines with age, but why juvenile organisms show enhanced tissue repair remains unexplained. Lin28a, a highly conserved RNA-binding protein expressed during embryogenesis, plays roles in development, pluripotency, and metabolism. To determine whether Lin28a might influence tissue repair in adults, we engineered the reactivation of Lin28a expression in several models of tissue injury. Lin28a reactivation improved hair regrowth by promoting anagen in hair follicles and accelerated regrowth of cartilage, bone, and mesenchyme after ear and digit injuries. Lin28a inhibits let-7 microRNA biogenesis; however, let-7 repression was necessary but insufficient to enhance repair. Lin28a bound to and enhanced the translation of mRNAs for several metabolic enzymes, thereby increasing glycolysis and oxidative phosphorylation (OxPhos). Lin28a-mediated enhancement of tissue repair was negated by OxPhos inhibition, whereas a pharmacologically induced increase in OxPhos enhanced repair. Thus, Lin28a enhances tissue repair in some adult tissues by reprogramming cellular bioenergetics. PAPERCLIP:


Subject(s)
RNA-Binding Proteins/metabolism , Wound Healing , Animals , Embryo, Mammalian/metabolism , Energy Metabolism , Extremities/physiology , Hair Follicle/physiology , Humans , Mice , Mice, Transgenic , MicroRNAs/metabolism , Regeneration
17.
Cell ; 154(2): 311-324, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23830207

ABSTRACT

Tumor cells metastasize to distant organs through genetic and epigenetic alterations, including changes in microRNA (miR) expression. Here we find miR-22 triggers epithelial-mesenchymal transition (EMT), enhances invasiveness and promotes metastasis in mouse xenografts. In a conditional mammary gland-specific transgenic (TG) mouse model, we show that miR-22 enhances mammary gland side-branching, expands the stem cell compartment, and promotes tumor development. Critically, miR-22 promotes aggressive metastatic disease in MMTV-miR-22 TG mice, as well as compound MMTV-neu or -PyVT-miR-22 TG mice. We demonstrate that miR-22 exerts its metastatic potential by silencing antimetastatic miR-200 through direct targeting of the TET (Ten eleven translocation) family of methylcytosine dioxygenases, thereby inhibiting demethylation of the mir-200 promoter. Finally, we show that miR-22 overexpression correlates with poor clinical outcomes and silencing of the TET-miR-200 axis in patients. Taken together, our findings implicate miR-22 as a crucial epigenetic modifier and promoter of EMT and breast cancer stemness toward metastasis.


Subject(s)
Breast Neoplasms/pathology , Chromatin Assembly and Disassembly , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Breast Neoplasms/metabolism , Cytosine/analogs & derivatives , Cytosine/metabolism , Humans , Mice , Mice, Transgenic , Neoplasm Transplantation , Proto-Oncogene Proteins/metabolism , RNA Interference , Transplantation, Heterologous
18.
Cell ; 153(4): 840-54, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23663782

ABSTRACT

Proliferating mammalian cells use glutamine as a source of nitrogen and as a key anaplerotic source to provide metabolites to the tricarboxylic acid cycle (TCA) for biosynthesis. Recently, mammalian target of rapamycin complex 1 (mTORC1) activation has been correlated with increased nutrient uptake and metabolism, but no molecular connection to glutaminolysis has been reported. Here, we show that mTORC1 promotes glutamine anaplerosis by activating glutamate dehydrogenase (GDH). This regulation requires transcriptional repression of SIRT4, the mitochondrial-localized sirtuin that inhibits GDH. Mechanistically, mTORC1 represses SIRT4 by promoting the proteasome-mediated destabilization of cAMP-responsive element binding 2 (CREB2). Thus, a relationship between mTORC1, SIRT4, and cancer is suggested by our findings. Indeed, SIRT4 expression is reduced in human cancer, and its overexpression reduces cell proliferation, transformation, and tumor development. Finally, our data indicate that targeting nutrient metabolism in energy-addicted cancers with high mTORC1 signaling may be an effective therapeutic approach.


Subject(s)
Glutamine/metabolism , Mitochondrial Proteins/metabolism , Neoplasms/metabolism , Sirtuins/metabolism , Activating Transcription Factors/metabolism , Animals , Cell Proliferation , Embryo, Mammalian/cytology , Energy Metabolism , Glutamate Dehydrogenase/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Neoplasm Transplantation , Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic , Transplantation, Heterologous , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitination
19.
Cell ; 155(2): 397-409, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120138

ABSTRACT

The pyruvate kinase M2 isoform (PKM2) is expressed in cancer and plays a role in regulating anabolic metabolism. To determine whether PKM2 is required for tumor formation or growth, we generated mice with a conditional allele that abolishes PKM2 expression without disrupting PKM1 expression. PKM2 deletion accelerated mammary tumor formation in a Brca1-loss-driven model of breast cancer. PKM2 null tumors displayed heterogeneous PKM1 expression, with PKM1 found in nonproliferating tumor cells and no detectable pyruvate kinase expression in proliferating cells. This suggests that PKM2 is not necessary for tumor cell proliferation and implies that the inactive state of PKM2 is associated with the proliferating cell population within tumors, whereas nonproliferating tumor cells require active pyruvate kinase. Consistent with these findings, variable PKM2 expression and heterozygous PKM2 mutations are found in human tumors. These data suggest that regulation of PKM2 activity supports the different metabolic requirements of proliferating and nonproliferating tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Gene Deletion , Mammary Neoplasms, Experimental/metabolism , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Exons , Female , Gene Knockout Techniques , Heterografts , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutation , Neoplasm Metastasis , Neoplasm Transplantation , RNA Splicing
20.
Cell ; 155(4): 844-57, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209622

ABSTRACT

Here, we show that a subset of breast cancers express high levels of the type 2 phosphatidylinositol-5-phosphate 4-kinases α and/or ß (PI5P4Kα and ß) and provide evidence that these kinases are essential for growth in the absence of p53. Knocking down PI5P4Kα and ß in a breast cancer cell line bearing an amplification of the gene encoding PI5P4K ß and deficient for p53 impaired growth on plastic and in xenografts. This growth phenotype was accompanied by enhanced levels of reactive oxygen species (ROS) leading to senescence. Mice with homozygous deletion of both TP53 and PIP4K2B were not viable, indicating a synthetic lethality for loss of these two genes. Importantly however, PIP4K2A(-/-), PIP4K2B(+/-), and TP53(-/-) mice were viable and had a dramatic reduction in tumor formation compared to TP53(-/-) littermates. These results indicate that inhibitors of PI5P4Ks could be effective in preventing or treating cancers with mutations in TP53.


Subject(s)
Breast Neoplasms/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Cell Respiration , Cellular Senescence , Embryo, Mammalian/metabolism , Gene Knockdown Techniques , Genes, Lethal , Heterografts , Humans , Mice , Neoplasm Transplantation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL