Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nature ; 568(7751): 254-258, 2019 04.
Article in English | MEDLINE | ID: mdl-30842661

ABSTRACT

Mitochondrial metabolism is an attractive target for cancer therapy1,2. Reprogramming metabolic pathways could improve the ability of metabolic inhibitors to suppress cancers with limited treatment options, such as triple-negative breast cancer (TNBC)1,3. Here we show that BTB and CNC homology1 (BACH1)4, a haem-binding transcription factor that is increased in expression in tumours from patients with TNBC, targets mitochondrial metabolism. BACH1 decreases glucose utilization in the tricarboxylic acid cycle and negatively regulates transcription of electron transport chain (ETC) genes. BACH1 depletion by shRNA or degradation by hemin sensitizes cells to ETC inhibitors such as metformin5,6, suppressing growth of both cell line and patient-derived tumour xenografts. Expression of a haem-resistant BACH1 mutant in cells that express a short hairpin RNA for BACH1 rescues the BACH1 phenotype and restores metformin resistance in hemin-treated cells and tumours7. Finally, BACH1 gene expression inversely correlates with ETC gene expression in tumours from patients with breast cancer and in other tumour types, which highlights the clinical relevance of our findings. This study demonstrates that mitochondrial metabolism can be exploited by targeting BACH1 to sensitize breast cancer and potentially other tumour tissues to mitochondrial inhibitors.


Subject(s)
Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Hemin/therapeutic use , Metformin/therapeutic use , Mitochondria/drug effects , Mitochondria/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Citric Acid Cycle/physiology , Electron Transport/genetics , Female , Glucose/metabolism , Hemin/metabolism , Heterografts , Humans , Metformin/metabolism , Mice , Mice, Nude , Mitochondria/genetics , Proteolysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Chem Rev ; 118(14): 6893-6923, 2018 07 25.
Article in English | MEDLINE | ID: mdl-29939018

ABSTRACT

Dysregulation of cancer cell metabolism contributes to abnormal cell growth, the biological end point of cancer. We review here numerous affected oncogenes and metabolic pathways common in cancer and how they contribute to cancer pathogenesis and malignancy. This review also discusses various pharmacological manipulations that take advantage of these metabolic abnormalities and the current targeted therapies that have arisen from this research.


Subject(s)
Neoplasms/metabolism , Amino Acids/metabolism , Carbohydrate Metabolism , Fatty Acids/metabolism , Humans , Neoplasms/pathology , Neoplasms/therapy , Oncogenes , Pentose Phosphate Pathway , Tumor Suppressor Proteins/metabolism
3.
Nat Cell Biol ; 20(7): 811-822, 2018 07.
Article in English | MEDLINE | ID: mdl-29941929

ABSTRACT

G protein αs (GNAS) mediates receptor-stimulated cAMP signalling, which integrates diverse environmental cues with intracellular responses. GNAS is mutationally activated in multiple tumour types, although its oncogenic mechanisms remain elusive. We explored this question in pancreatic tumourigenesis where concurrent GNAS and KRAS mutations characterize pancreatic ductal adenocarcinomas (PDAs) arising from intraductal papillary mucinous neoplasms (IPMNs). By developing genetically engineered mouse models, we show that GnasR201C cooperates with KrasG12D to promote initiation of IPMN, which progress to invasive PDA following Tp53 loss. Mutant Gnas remains critical for tumour maintenance in vivo. This is driven by protein-kinase-A-mediated suppression of salt-inducible kinases (Sik1-3), associated with induction of lipid remodelling and fatty acid oxidation. Comparison of Kras-mutant pancreatic cancer cells with and without Gnas mutations reveals striking differences in the functions of this network. Thus, we uncover Gnas-driven oncogenic mechanisms, identify Siks as potent tumour suppressors, and demonstrate unanticipated metabolic heterogeneity among Kras-mutant pancreatic neoplasms.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cellular Reprogramming/genetics , Chromogranins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Lipid Metabolism/genetics , Mutation , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Chromogranins/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Repression , Fatty Acids/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation, Neoplastic , Genes, ras , Genetic Predisposition to Disease , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Mutant Strains , Mice, Transgenic , Oxidation-Reduction , Pancreatic Neoplasms/pathology , Phenotype , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Dev Cell ; 44(1): 97-112.e7, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29275994

ABSTRACT

Lipid droplet (LD) functions are regulated by a complement of integral and peripheral proteins that associate with the bounding LD phospholipid monolayer. Defining the composition of the LD proteome has remained a challenge due to the presence of contaminating proteins in LD-enriched buoyant fractions. To overcome this limitation, we developed a proximity labeling strategy that exploits LD-targeted APEX2 to biotinylate LD proteins in living cells. Application of this approach to two different cell types identified the vast majority of previously validated LD proteins, excluded common contaminating proteins, and revealed new LD proteins. Moreover, quantitative analysis of LD proteome dynamics uncovered a role for endoplasmic reticulum-associated degradation in controlling the composition of the LD proteome. These data provide an important resource for future LD studies and demonstrate the utility of proximity labeling to study the regulation of LD proteomes.


Subject(s)
Biomarkers/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Lipid Droplets/metabolism , Proteome/metabolism , Staining and Labeling/methods , ATPases Associated with Diverse Cellular Activities/metabolism , Carrier Proteins/metabolism , Humans , Membrane Proteins/metabolism , Membrane Transport Proteins , Proteome/analysis , Receptors, Autocrine Motility Factor/metabolism
5.
Cell Chem Biol ; 24(11): 1368-1376.e4, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28919038

ABSTRACT

Many natural products that show therapeutic activities are often difficult to synthesize or isolate and have unknown targets, hindering their development as drugs. Identifying druggable hotspots targeted by covalently acting anti-cancer natural products can enable pharmacological interrogation of these sites with more synthetically tractable compounds. Here, we used chemoproteomic platforms to discover that the anti-cancer natural product withaferin A targets C377 on the regulatory subunit PPP2R1A of the tumor-suppressor protein phosphatase 2A (PP2A) complex leading to activation of PP2A activity, inactivation of AKT, and impaired breast cancer cell proliferation. We developed a more synthetically tractable cysteine-reactive covalent ligand, JNS 1-40, that selectively targets C377 of PPP2R1A to impair breast cancer signaling, proliferation, and in vivo tumor growth. Our study highlights the utility of using chemoproteomics to map druggable hotspots targeted by complex natural products and subsequently interrogating these sites with more synthetically tractable covalent ligands for cancer therapy.


Subject(s)
Antineoplastic Agents/metabolism , Biological Products/metabolism , Protein Phosphatase 2/metabolism , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine/chemistry , Female , Humans , Ligands , MCF-7 Cells , Protein Phosphatase 2/chemistry , Proteome/drug effects , Proteome/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Withanolides/chemistry , Withanolides/pharmacology
6.
Cell Chem Biol ; 23(5): 567-578, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27185638

ABSTRACT

Breast cancers possess fundamentally altered metabolism that fuels their pathogenicity. While many metabolic drivers of breast cancers have been identified, the metabolic pathways that mediate breast cancer malignancy and poor prognosis are less well understood. Here, we used a reactivity-based chemoproteomic platform to profile metabolic enzymes that are enriched in breast cancer cell types linked to poor prognosis, including triple-negative breast cancer (TNBC) cells and breast cancer cells that have undergone an epithelial-mesenchymal transition-like state of heightened malignancy. We identified glutathione S-transferase Pi 1 (GSTP1) as a novel TNBC target that controls cancer pathogenicity by regulating glycolytic and lipid metabolism, energetics, and oncogenic signaling pathways through a protein interaction that activates glyceraldehyde-3-phosphate dehydrogenase activity. We show that genetic or pharmacological inactivation of GSTP1 impairs cell survival and tumorigenesis in TNBC cells. We put forth GSTP1 inhibitors as a novel therapeutic strategy for combatting TNBCs through impairing key cancer metabolism and signaling pathways.


Subject(s)
Glutathione S-Transferase pi/metabolism , Leucine/analogs & derivatives , Triazines/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glutathione S-Transferase pi/antagonists & inhibitors , Glutathione S-Transferase pi/genetics , Humans , Leucine/chemistry , Leucine/pharmacology , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Triazines/chemistry , Triple Negative Breast Neoplasms/drug therapy , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...