Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 183(5): 1202-1218.e25, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33142117

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) tumors have a nutrient-poor, desmoplastic, and highly innervated tumor microenvironment. Although neurons can release stimulatory factors to accelerate PDAC tumorigenesis, the metabolic contribution of peripheral axons has not been explored. We found that peripheral axons release serine (Ser) to support the growth of exogenous Ser (exSer)-dependent PDAC cells during Ser/Gly (glycine) deprivation. Ser deprivation resulted in ribosomal stalling on two of the six Ser codons, TCC and TCT, and allowed the selective translation and secretion of nerve growth factor (NGF) by PDAC cells to promote tumor innervation. Consistent with this, exSer-dependent PDAC tumors grew slower and displayed enhanced innervation in mice on a Ser/Gly-free diet. Blockade of compensatory neuronal innervation using LOXO-101, a Trk-NGF inhibitor, further decreased PDAC tumor growth. Our data indicate that axonal-cancer metabolic crosstalk is a critical adaptation to support PDAC growth in nutrient poor environments.


Subject(s)
Neurons/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Protein Biosynthesis , Serine/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Animals , Axons/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Codon/genetics , Female , Glycine/metabolism , Humans , Male , Mice , Middle Aged , Mitochondria/metabolism , Nerve Tissue/pathology , Oxygen Consumption , Pancreatic Neoplasms/pathology , Pyrazoles , Pyrimidines , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/genetics , Rats
2.
Cell ; 182(3): 641-654.e20, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32615085

ABSTRACT

Targeting glycolysis has been considered therapeutically intractable owing to its essential housekeeping role. However, the context-dependent requirement for individual glycolytic steps has not been fully explored. We show that CRISPR-mediated targeting of glycolysis in T cells in mice results in global loss of Th17 cells, whereas deficiency of the glycolytic enzyme glucose phosphate isomerase (Gpi1) selectively eliminates inflammatory encephalitogenic and colitogenic Th17 cells, without substantially affecting homeostatic microbiota-specific Th17 cells. In homeostatic Th17 cells, partial blockade of glycolysis upon Gpi1 inactivation was compensated by pentose phosphate pathway flux and increased mitochondrial respiration. In contrast, inflammatory Th17 cells experience a hypoxic microenvironment known to limit mitochondrial respiration, which is incompatible with loss of Gpi1. Our study suggests that inhibiting glycolysis by targeting Gpi1 could be an effective therapeutic strategy with minimum toxicity for Th17-mediated autoimmune diseases, and, more generally, that metabolic redundancies can be exploited for selective targeting of disease processes.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Glucose-6-Phosphate Isomerase/metabolism , Glycolysis/genetics , Oxidative Phosphorylation , Pentose Phosphate Pathway/physiology , Th17 Cells/metabolism , Animals , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Chimera/genetics , Chromatography, Gas , Chromatography, Liquid , Clostridium Infections/immunology , Cytokines/deficiency , Cytokines/genetics , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis/immunology , Homeostasis/genetics , Homeostasis/immunology , Inflammation/genetics , Inflammation/immunology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Pentose Phosphate Pathway/genetics , Pentose Phosphate Pathway/immunology , RNA-Seq , Single-Cell Analysis , Th17 Cells/immunology , Th17 Cells/pathology
3.
Cell ; 158(5): 1094-1109, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171410

ABSTRACT

It is increasingly appreciated that oncogenic transformation alters cellular metabolism to facilitate cell proliferation, but less is known about the metabolic changes that promote cancer cell aggressiveness. Here, we analyzed metabolic gene expression in cancer cell lines and found that a set of high-grade carcinoma lines expressing mesenchymal markers share a unique 44 gene signature, designated the "mesenchymal metabolic signature" (MMS). A FACS-based shRNA screen identified several MMS genes as essential for the epithelial-mesenchymal transition (EMT), but not for cell proliferation. Dihydropyrimidine dehydrogenase (DPYD), a pyrimidine-degrading enzyme, was highly expressed upon EMT induction and was necessary for cells to acquire mesenchymal characteristics in vitro and for tumorigenic cells to extravasate into the mouse lung. This role of DPYD was mediated through its catalytic activity and enzymatic products, the dihydropyrimidines. Thus, we identify metabolic processes essential for the EMT, a program associated with the acquisition of metastatic and aggressive cancer cell traits.


Subject(s)
Epithelial-Mesenchymal Transition , Pyrimidines/metabolism , Animals , Carcinoma/metabolism , Cell Line, Tumor , Dihydrouracil Dehydrogenase (NADP)/genetics , Flow Cytometry , Gene Expression Profiling , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , RNA, Small Interfering/metabolism
4.
Nature ; 597(7876): 420-425, 2021 09.
Article in English | MEDLINE | ID: mdl-34471290

ABSTRACT

Oxygen is critical for a multitude of metabolic processes that are essential for human life. Biological processes can be identified by treating cells with 18O2 or other isotopically labelled gases and systematically identifying biomolecules incorporating labeled atoms. Here we labelled cell lines of distinct tissue origins with 18O2 to identify the polar oxy-metabolome, defined as polar metabolites labelled with 18O under different physiological O2 tensions. The most highly 18O-labelled feature was 4-hydroxymandelate (4-HMA). We demonstrate that 4-HMA is produced by hydroxyphenylpyruvate dioxygenase-like (HPDL), a protein of previously unknown function in human cells. We identify 4-HMA as an intermediate involved in the biosynthesis of the coenzyme Q10 (CoQ10) headgroup in human cells. The connection of HPDL to CoQ10 biosynthesis provides crucial insights into the mechanisms underlying recently described neurological diseases related to HPDL deficiencies1-4 and cancers with HPDL overexpression5.


Subject(s)
4-Hydroxyphenylpyruvate Dioxygenase/metabolism , Mandelic Acids/metabolism , Metabolome , Ubiquinone/analogs & derivatives , Animals , Cell Line , Female , Humans , Mandelic Acids/analysis , Mice , Mice, Nude , Tyrosine/metabolism , Ubiquinone/biosynthesis
5.
Nature ; 520(7547): 363-7, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25855294

ABSTRACT

Cancer cells adapt their metabolic processes to support rapid proliferation, but less is known about how cancer cells alter metabolism to promote cell survival in a poorly vascularized tumour microenvironment. Here we identify a key role for serine and glycine metabolism in the survival of brain cancer cells within the ischaemic zones of gliomas. In human glioblastoma multiforme, mitochondrial serine hydroxymethyltransferase (SHMT2) and glycine decarboxylase (GLDC) are highly expressed in the pseudopalisading cells that surround necrotic foci. We find that SHMT2 activity limits that of pyruvate kinase (PKM2) and reduces oxygen consumption, eliciting a metabolic state that confers a profound survival advantage to cells in poorly vascularized tumour regions. GLDC inhibition impairs cells with high SHMT2 levels as the excess glycine not metabolized by GLDC can be converted to the toxic molecules aminoacetone and methylglyoxal. Thus, SHMT2 is required for cancer cells to adapt to the tumour environment, but also renders these cells sensitive to glycine cleavage system inhibition.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Glycine Hydroxymethyltransferase/metabolism , Glycine/metabolism , Ischemia/metabolism , Acetone/analogs & derivatives , Acetone/metabolism , Acetone/toxicity , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/enzymology , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Female , Glioblastoma/blood supply , Glioblastoma/enzymology , Glycine Dehydrogenase (Decarboxylating)/antagonists & inhibitors , Glycine Dehydrogenase (Decarboxylating)/metabolism , Humans , Ischemia/enzymology , Ischemia/pathology , Mice , Necrosis , Oxygen Consumption , Pyruvaldehyde/metabolism , Pyruvaldehyde/toxicity , Pyruvate Kinase/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Proc Natl Acad Sci U S A ; 114(17): E3434-E3443, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28396387

ABSTRACT

Oncogenic PIK3CA mutations are found in a significant fraction of human cancers, but therapeutic inhibition of PI3K has only shown limited success in clinical trials. To understand how mutant PIK3CA contributes to cancer cell proliferation, we used genome scale loss-of-function screening in a large number of genomically annotated cancer cell lines. As expected, we found that PIK3CA mutant cancer cells require PIK3CA but also require the expression of the TCA cycle enzyme 2-oxoglutarate dehydrogenase (OGDH). To understand the relationship between oncogenic PIK3CA and OGDH function, we interrogated metabolic requirements and found an increased reliance on glucose metabolism to sustain PIK3CA mutant cell proliferation. Functional metabolic studies revealed that OGDH suppression increased levels of the metabolite 2-oxoglutarate (2OG). We found that this increase in 2OG levels, either by OGDH suppression or exogenous 2OG treatment, resulted in aspartate depletion that was specifically manifested as auxotrophy within PIK3CA mutant cells. Reduced levels of aspartate deregulated the malate-aspartate shuttle, which is important for cytoplasmic NAD+ regeneration that sustains rapid glucose breakdown through glycolysis. Consequently, because PIK3CA mutant cells exhibit a profound reliance on glucose metabolism, malate-aspartate shuttle deregulation leads to a specific proliferative block due to the inability to maintain NAD+/NADH homeostasis. Together these observations define a precise metabolic vulnerability imposed by a recurrently mutated oncogene.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Ketoglutarate Dehydrogenase Complex , Mutation , Neoplasm Proteins , Neoplasms , Animals , Cell Line, Tumor , Citric Acid Cycle/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Glycolysis/genetics , Humans , Ketoglutarate Dehydrogenase Complex/biosynthesis , Ketoglutarate Dehydrogenase Complex/genetics , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology
7.
Nature ; 498(7453): 246-50, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23728299

ABSTRACT

DNA damage activates a signalling network that blocks cell-cycle progression, recruits DNA repair factors and/or triggers senescence or programmed cell death. Alterations in chromatin structure are implicated in the initiation and propagation of the DNA damage response. Here we further investigate the role of chromatin structure in the DNA damage response by monitoring ionizing-radiation-induced signalling and response events with a high-content multiplex RNA-mediated interference screen of chromatin-modifying and -interacting genes. We discover that an isoform of Brd4, a bromodomain and extra-terminal (BET) family member, functions as an endogenous inhibitor of DNA damage response signalling by recruiting the condensin II chromatin remodelling complex to acetylated histones through bromodomain interactions. Loss of this isoform results in relaxed chromatin structure, rapid cell-cycle checkpoint recovery and enhanced survival after irradiation, whereas functional gain of this isoform compacted chromatin, attenuated DNA damage response signalling and enhanced radiation-induced lethality. These data implicate Brd4, previously known for its role in transcriptional control, as an insulator of chromatin that can modulate the signalling response to DNA damage.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA Damage , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Acetylation , Adenosine Triphosphatases/metabolism , Cell Cycle Checkpoints/radiation effects , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/radiation effects , Chromatin/chemistry , Chromatin/radiation effects , Chromatin Assembly and Disassembly/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , Histones/chemistry , Histones/metabolism , Humans , Lysine/chemistry , Lysine/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphorylation/radiation effects , Positive Transcriptional Elongation Factor B/metabolism , Protein Isoforms/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/genetics
8.
Nat Chem Biol ; 12(6): 452-8, 2016 06.
Article in English | MEDLINE | ID: mdl-27110680

ABSTRACT

Serine is both a proteinogenic amino acid and the source of one-carbon units essential for de novo purine and deoxythymidine synthesis. In the canonical pathway of glucose-derived serine synthesis, Homo sapiens phosphoglycerate dehydrogenase (PHGDH) catalyzes the first, rate-limiting step. Genetic loss of PHGDH is toxic toward PHGDH-overexpressing breast cancer cell lines even in the presence of exogenous serine. Here, we used a quantitative high-throughput screen to identify small-molecule PHGDH inhibitors. These compounds reduce the production of glucose-derived serine in cells and suppress the growth of PHGDH-dependent cancer cells in culture and in orthotopic xenograft tumors. Surprisingly, PHGDH inhibition reduced the incorporation into nucleotides of one-carbon units from glucose-derived and exogenous serine. We conclude that glycolytic serine synthesis coordinates the use of one-carbon units from endogenous and exogenous serine in nucleotide synthesis, and we suggest that one-carbon unit wasting thus may contribute to the efficacy of PHGDH inhibitors in vitro and in vivo.


Subject(s)
Carbon/metabolism , Enzyme Inhibitors/pharmacology , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Serine/biosynthesis , Small Molecule Libraries/pharmacology , Animals , Carbon/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Female , Glycolysis/drug effects , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Molecular Structure , Phosphoglycerate Dehydrogenase/metabolism , Purines/biosynthesis , Serine/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Thymidine/biosynthesis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Bioorg Med Chem ; 26(8): 1727-1739, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29555419

ABSTRACT

Proliferating cells, including cancer cells, obtain serine both exogenously and via the metabolism of glucose. By catalyzing the first, rate-limiting step in the synthesis of serine from glucose, phosphoglycerate dehydrogenase (PHGDH) controls flux through the biosynthetic pathway for this important amino acid and represents a putative target in oncology. To discover inhibitors of PHGDH, a coupled biochemical assay was developed and optimized to enable high-throughput screening for inhibitors of human PHGDH. Feedback inhibition was minimized by coupling PHGDH activity to two downstream enzymes (PSAT1 and PSPH), providing a marked improvement in enzymatic turnover. Further coupling of NADH to a diaphorase/resazurin system enabled a red-shifted detection readout, minimizing interference due to compound autofluorescence. With this protocol, over 400,000 small molecules were screened for PHGDH inhibition, and following hit validation and triage work, a piperazine-1-thiourea was identified. Following rounds of medicinal chemistry and SAR exploration, two probes (NCT-502 and NCT-503) were identified. These molecules demonstrated improved target activity and encouraging ADME properties, enabling in vitro assessment of the biological importance of PHGDH, and its role in the fate of serine in PHGDH-dependent cancer cells. This manuscript reports the assay development and medicinal chemistry leading to the development of NCT-502 and -503 reported in Pacold et al. (2016).


Subject(s)
Enzyme Inhibitors/pharmacology , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Piperazines/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Dose-Response Relationship, Drug , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Humans , Molecular Structure , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Piperazines/chemical synthesis , Piperazines/chemistry , Structure-Activity Relationship , Thiourea/chemical synthesis , Thiourea/chemistry
10.
Nat Chem Biol ; 16(7): 713-714, 2020 07.
Article in English | MEDLINE | ID: mdl-32572257
11.
Nature ; 476(7360): 346-50, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21760589

ABSTRACT

Cancer cells adapt their metabolic processes to drive macromolecular biosynthesis for rapid cell growth and proliferation. RNA interference (RNAi)-based loss-of-function screening has proven powerful for the identification of new and interesting cancer targets, and recent studies have used this technology in vivo to identify novel tumour suppressor genes. Here we developed a method for identifying novel cancer targets via negative-selection RNAi screening using a human breast cancer xenograft model at an orthotopic site in the mouse. Using this method, we screened a set of metabolic genes associated with aggressive breast cancer and stemness to identify those required for in vivo tumorigenesis. Among the genes identified, phosphoglycerate dehydrogenase (PHGDH) is in a genomic region of recurrent copy number gain in breast cancer and PHGDH protein levels are elevated in 70% of oestrogen receptor (ER)-negative breast cancers. PHGDH catalyses the first step in the serine biosynthesis pathway, and breast cancer cells with high PHGDH expression have increased serine synthesis flux. Suppression of PHGDH in cell lines with elevated PHGDH expression, but not in those without, causes a strong decrease in cell proliferation and a reduction in serine synthesis. We find that PHGDH suppression does not affect intracellular serine levels, but causes a drop in the levels of α-ketoglutarate, another output of the pathway and a tricarboxylic acid (TCA) cycle intermediate. In cells with high PHGDH expression, the serine synthesis pathway contributes approximately 50% of the total anaplerotic flux of glutamine into the TCA cycle. These results reveal that certain breast cancers are dependent upon increased serine pathway flux caused by PHGDH overexpression and demonstrate the utility of in vivo negative-selection RNAi screens for finding potential anticancer targets.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Genomics , Serine/biosynthesis , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle/physiology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glutamic Acid/metabolism , Humans , Ketoglutaric Acids/metabolism , Melanoma/enzymology , Melanoma/genetics , Mice , Neoplasm Transplantation , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , RNA Interference
12.
Proc Natl Acad Sci U S A ; 109(21): 8230-5, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22547809

ABSTRACT

One of the promises of nanoparticle (NP) carriers is the reformulation of promising therapeutics that have failed clinical development due to pharmacologic challenges. However, current nanomedicine research has been focused on the delivery of established and novel therapeutics. Here we demonstrate proof of the principle of using NPs to revive the clinical potential of abandoned compounds using wortmannin (Wtmn) as a model drug. Wtmn is a potent inhibitor of phosphatidylinositol 3' kinase-related kinases but failed clinical translation due to drug-delivery challenges. We engineered a NP formulation of Wtmn and demonstrated that NP Wtmn has higher solubility and lower toxicity compared with Wtmn. To establish the clinical translation potential of NP Wtmn, we evaluated the therapeutic as a radiosensitizer in vitro and in vivo. NP Wtmn was found to be a potent radiosensitizer and was significantly more effective than the commonly used radiosensitizer cisplatin in vitro in three cancer cell lines. The mechanism of action of NP Wtmn radiosensitization was found to be through the inhibition of DNA-dependent protein kinase phosphorylation. Finally, NP Wtmn was shown to be an effective radiosensitizer in vivo using two murine xenograft models of cancer. Our results demonstrate that NP drug-delivery systems can promote the readoption of abandoned drugs such as Wtmn by overcoming drug-delivery challenges.


Subject(s)
Androstadienes/pharmacokinetics , Drug Delivery Systems/methods , Nanoparticles , Neoplasms/therapy , Protein Kinase Inhibitors/pharmacokinetics , Radiation-Sensitizing Agents/pharmacokinetics , Androstadienes/toxicity , Animals , Cell Survival/drug effects , Chemoradiotherapy/methods , HT29 Cells , Humans , KB Cells , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Phosphorylation/drug effects , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Radiation-Sensitizing Agents/toxicity , Wortmannin , Xenograft Model Antitumor Assays
13.
Angew Chem Int Ed Engl ; 53(1): 199-204, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24259466

ABSTRACT

We report the synthesis of a GDP analogue, SML-8-73-1, and a prodrug derivative, SML-10-70-1, which are selective, direct-acting covalent inhibitors of the K-Ras G12C mutant relative to wild-type Ras. Biochemical and biophysical measurements suggest that modification of K-Ras with SML-8-73-1 renders the protein in an inactive state. These first-in-class covalent K-Ras inhibitors demonstrate that irreversible targeting of the K-Ras guanine-nucleotide binding site is potentially a viable therapeutic strategy for inhibition of Ras signaling.


Subject(s)
Catalytic Domain/genetics , ras Proteins/chemistry , ras Proteins/genetics , Drug Design , Signal Transduction , ras Proteins/metabolism
14.
bioRxiv ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38529505

ABSTRACT

Mitochondrial dysfunction causes devastating disorders, including mitochondrial myopathy. Here, we identified that diverse mitochondrial myopathy models elicit a protective mitochondrial integrated stress response (mt-ISR), mediated by OMA1-DELE1 signaling. The response was similar following disruptions in mtDNA maintenance, from knockout of Tfam, and mitochondrial protein unfolding, from disease-causing mutations in CHCHD10 (G58R and S59L). The preponderance of the response was directed at upregulating pathways for aminoacyl-tRNA biosynthesis, the intermediates for protein synthesis, and was similar in heart and skeletal muscle but more limited in brown adipose challenged with cold stress. Strikingly, models with early DELE1 mt-ISR activation failed to grow and survive to adulthood in the absence of Dele1, accounting for some but not all of OMA1's protection. Notably, the DELE1 mt-ISR did not slow net protein synthesis in stressed striated muscle, but instead prevented loss of translation-associated proteostasis in muscle fibers. Together our findings identify that the DELE1 mt-ISR mediates a stereotyped response to diverse forms of mitochondrial stress and is particularly critical for maintaining growth and survival in early-onset mitochondrial myopathy.

16.
Acta Neuropathol Commun ; 10(1): 150, 2022 10 23.
Article in English | MEDLINE | ID: mdl-36274161

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is an aggressive incurable brainstem tumor that targets young children. Complete resection is not possible, and chemotherapy and radiotherapy are currently only palliative. This study aimed to identify potential therapeutic agents using a computational pipeline to perform an in silico screen for novel drugs. We then tested the identified drugs against a panel of patient-derived DIPG cell lines. Using a systematic computational approach with publicly available databases of gene signature in DIPG patients and cancer cell lines treated with a library of clinically available drugs, we identified drug hits with the ability to reverse a DIPG gene signature to one that matches normal tissue background. The biological and molecular effects of drug treatment was analyzed by cell viability assay and RNA sequence. In vivo DIPG mouse model survival studies were also conducted. As a result, two of three identified drugs showed potency against the DIPG cell lines Triptolide and mycophenolate mofetil (MMF) demonstrated significant inhibition of cell viability in DIPG cell lines. Guanosine rescued reduced cell viability induced by MMF. In vivo, MMF treatment significantly inhibited tumor growth in subcutaneous xenograft mice models. In conclusion, we identified clinically available drugs with the ability to reverse DIPG gene signatures and anti-DIPG activity in vitro and in vivo. This novel approach can repurpose drugs and significantly decrease the cost and time normally required in drug discovery.


Subject(s)
Astrocytoma , Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Glioma , Humans , Mice , Animals , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/genetics , Mycophenolic Acid/therapeutic use , Glioma/drug therapy , Glioma/genetics , Glioma/metabolism , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Gene Expression , Guanosine/therapeutic use
17.
Elife ; 112022 09 27.
Article in English | MEDLINE | ID: mdl-36165439

ABSTRACT

Major genomic deletions in independent eukaryotic lineages have led to repeated ancestral loss of biosynthesis pathways for nine of the twenty canonical amino acids. While the evolutionary forces driving these polyphyletic deletion events are not well understood, the consequence is that extant metazoans are unable to produce nine essential amino acids (EAAs). Previous studies have highlighted that EAA biosynthesis tends to be more energetically costly, raising the possibility that these pathways were lost from organisms with access to abundant EAAs. It is unclear whether present-day metazoans can reaccept these pathways to resurrect biosynthetic capabilities that were lost long ago or whether evolution has rendered EAA pathways incompatible with metazoan metabolism. Here, we report progress on a large-scale synthetic genomics effort to reestablish EAA biosynthetic functionality in mammalian cells. We designed codon-optimized biosynthesis pathways based on genes mined from Escherichia coli. These pathways were de novo synthesized in 3 kilobase chunks, assembled in yeasto and genomically integrated into a Chinese hamster ovary (CHO) cell line. One synthetic pathway produced valine at a sufficient level for cell viability and proliferation. 13C-tracing verified de novo biosynthesis of valine and further revealed build-up of pathway intermediate 2,3-dihydroxy-3-isovalerate. Increasing the dosage of downstream ilvD boosted pathway performance and allowed for long-term propagation of second-generation cells in valine-free medium at 3.2 days per doubling. This work demonstrates that mammalian metabolism is amenable to restoration of ancient core pathways, paving a path for genome-scale efforts to synthetically restore metabolic functions to the metazoan lineage.


Subject(s)
Amino Acids, Essential , Genome , Amino Acids/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Escherichia coli/genetics , Mammals , Valine
18.
Pigment Cell Melanoma Res ; 35(6): 554-572, 2022 11.
Article in English | MEDLINE | ID: mdl-35912544

ABSTRACT

Brain metastases are the most common brain malignancy. This review discusses the studies presented at the third annual meeting of the Melanoma Research Foundation in the context of other recent reports on the biology and treatment of melanoma brain metastases (MBM). Although symptomatic MBM patients were historically excluded from immunotherapy trials, efforts from clinicians and patient advocates have resulted in more inclusive and even dedicated clinical trials for MBM patients. The results of checkpoint inhibitor trials were discussed in conversation with current standards of care for MBM patients, including steroids, radiotherapy, and targeted therapy. Advances in the basic scientific understanding of MBM, including the role of astrocytes and metabolic adaptations to the brain microenvironment, are exposing new vulnerabilities which could be exploited for therapeutic purposes. Technical advances including single-cell omics and multiplex imaging are expanding our understanding of the MBM ecosystem and its response to therapy. This unprecedented level of spatial and temporal resolution is expected to dramatically advance the field in the coming years and render novel treatment approaches that might improve MBM patient outcomes.


Subject(s)
Brain Neoplasms , Melanoma , Neoplasms, Second Primary , Humans , Ecosystem , Melanoma/pathology , Brain Neoplasms/therapy , Brain Neoplasms/secondary , Immunotherapy/methods , Neoplasms, Second Primary/pathology , Brain , Tumor Microenvironment
19.
Elife ; 92020 10 12.
Article in English | MEDLINE | ID: mdl-33043885

ABSTRACT

Neurons communicate by the activity-dependent release of small-molecule neurotransmitters packaged into synaptic vesicles (SVs). Although many molecules have been identified as neurotransmitters, technical limitations have precluded a full metabolomic analysis of SV content. Here, we present a workflow to rapidly isolate SVs and to interrogate their metabolic contents at high-resolution using mass spectrometry. We validated the enrichment of glutamate in SVs of primary cortical neurons using targeted polar metabolomics. Unbiased and extensive global profiling of SVs isolated from these neurons revealed that the only detectable polar metabolites they contain are the established neurotransmitters glutamate and GABA. In addition, we adapted the approach to enable quick capture of SVs directly from brain tissue and determined the neurotransmitter profiles of diverse brain regions in a cell-type-specific manner. The speed, robustness, and precision of this method to interrogate SV contents will facilitate novel insights into the chemical basis of neurotransmission.


Subject(s)
Brain/metabolism , Mass Spectrometry/methods , Metabolome , Metabolomics/methods , Synaptic Vesicles/metabolism , Animals , Female , Glutamic Acid/metabolism , Male , Mice
20.
Cancer Discov ; 10(9): 1352-1373, 2020 09.
Article in English | MEDLINE | ID: mdl-32571778

ABSTRACT

A hallmark of metastasis is the adaptation of tumor cells to new environments. Metabolic constraints imposed by the serine and glycine-limited brain environment restrict metastatic tumor growth. How brain metastases overcome these growth-prohibitive conditions is poorly understood. Here, we demonstrate that 3-phosphoglycerate dehydrogenase (PHGDH), which catalyzes the rate-limiting step of glucose-derived serine synthesis, is a major determinant of brain metastasis in multiple human cancer types and preclinical models. Enhanced serine synthesis proved important for nucleotide production and cell proliferation in highly aggressive brain metastatic cells. In vivo, genetic suppression and pharmacologic inhibition of PHGDH attenuated brain metastasis, but not extracranial tumor growth, and improved overall survival in mice. These results reveal that extracellular amino acid availability determines serine synthesis pathway dependence, and suggest that PHGDH inhibitors may be useful in the treatment of brain metastasis. SIGNIFICANCE: Using proteomics, metabolomics, and multiple brain metastasis models, we demonstrate that the nutrient-limited environment of the brain potentiates brain metastasis susceptibility to serine synthesis inhibition. These findings underscore the importance of studying cancer metabolism in physiologically relevant contexts, and provide a rationale for using PHGDH inhibitors to treat brain metastasis.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain/pathology , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Brain/metabolism , Brain Neoplasms/secondary , Cell Line, Tumor , Datasets as Topic , Drug Resistance, Neoplasm , Female , Gene Knockdown Techniques , Glycine/analysis , Glycine/metabolism , Humans , Metabolomics , Mice , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Proteomics , RNA-Seq , Serine/analysis , Serine/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL