Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
2.
J Lipid Res ; 63(10): 100281, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36115594

ABSTRACT

Serine palmitoyltransferase (SPT) predominantly incorporates serine and fatty acyl-CoAs into diverse sphingolipids (SLs) that serve as structural components of membranes and signaling molecules within or amongst cells. However, SPT also uses alanine as a substrate in the contexts of low serine availability, alanine accumulation, or disease-causing mutations in hereditary sensory neuropathy type I, resulting in the synthesis and accumulation of 1-deoxysphingolipids (deoxySLs). These species promote cytotoxicity in neurons and impact diverse cellular phenotypes, including suppression of anchorage-independent cancer cell growth. While altered serine and alanine levels can promote 1-deoxySL synthesis, they impact numerous other metabolic pathways important for cancer cells. Here, we combined isotope tracing, quantitative metabolomics, and functional studies to better understand the mechanistic drivers of 1-deoxySL toxicity in cancer cells. We determined that both alanine treatment and SPTLC1C133W expression induce 1-deoxy(dihydro)ceramide synthesis and accumulation but fail to broadly impact intermediary metabolism, abundances of other lipids, or growth of adherent cells. However, we found that spheroid culture and soft agar colony formation were compromised when endogenous 1-deoxySL synthesis was induced via SPTLC1C133W expression. Consistent with these impacts on anchorage-independent cell growth, we observed that 1-deoxySL synthesis reduced plasma membrane endocytosis. These results highlight a potential role for SPT promiscuity in linking altered amino acid metabolism to plasma membrane endocytosis.


Subject(s)
Neoplasms , Serine C-Palmitoyltransferase , Serine C-Palmitoyltransferase/metabolism , Agar/metabolism , Sphingolipids/metabolism , Serine/chemistry , Ceramides/metabolism , Alanine/metabolism , Cell Membrane/metabolism , Metabolic Networks and Pathways , Endocytosis , Neoplasms/metabolism
3.
Cancer Metab ; 9(1): 40, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34861885

ABSTRACT

BACKGROUND: Kidney cancer is a common adult malignancy in the USA. Clear cell renal cell carcinoma (ccRCC), the predominant subtype of kidney cancer, is characterized by widespread metabolic changes. Urea metabolism is one such altered pathway in ccRCC. The aim of this study was to elucidate the contributions of urea cycle enzymes, argininosuccinate synthase 1 (ASS1), and argininosuccinate lyase (ASL) towards ccRCC progression. METHODS: We employed a combination of computational, genetic, and metabolomic tools along with in vivo animal models to establish a tumor-suppressive role for ASS1 and ASL in ccRCC. RESULTS: We show that the mRNA and protein expression of urea cycle enzymes ASS1 and ASL are reduced in ccRCC tumors when compared to the normal kidney. Furthermore, the loss of ASL in HK-2 cells (immortalized renal epithelial cells) promotes growth in 2D and 3D growth assays, while combined re-expression of ASS1 and ASL in ccRCC cell lines suppresses growth in 2D, 3D, and in vivo xenograft models. We establish that this suppression is dependent on their enzymatic activity. Finally, we demonstrate that conservation of cellular aspartate, regulation of nitric oxide synthesis, and pyrimidine production play pivotal roles in ASS1+ASL-mediated growth suppression in ccRCC. CONCLUSIONS: ccRCC tumors downregulate the components of the urea cycle including the enzymes argininosuccinate synthase 1 (ASS1) and argininosuccinate lyase (ASL). These cytosolic enzymes lie at a critical metabolic hub in the cell and are involved in aspartate catabolism and arginine and nitric oxide biosynthesis. Loss of ASS1 and ASL helps cells redirect aspartate towards pyrimidine synthesis and support enhanced proliferation. Additionally, reduced levels of ASS1 and ASL might help regulate nitric oxide (NO) generation and mitigate its cytotoxic effects. Overall, our work adds to the understanding of urea cycle enzymes in a context-independent of ureagenesis, their role in ccRCC progression, and uncovers novel potential metabolic vulnerabilities in ccRCC.

4.
Nat Commun ; 11(1): 498, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980651

ABSTRACT

Tumour cells frequently utilize glutamine to meet bioenergetic and biosynthetic demands of rapid cell growth. However, glutamine dependence can be highly variable between in vitro and in vivo settings, based on surrounding microenvironments and complex adaptive responses to glutamine deprivation. Soft tissue sarcomas (STSs) are mesenchymal tumours where cytotoxic chemotherapy remains the primary approach for metastatic or unresectable disease. Therefore, it is critical to identify alternate therapies to improve patient outcomes. Using autochthonous STS murine models and unbiased metabolomics, we demonstrate that glutamine metabolism supports sarcomagenesis. STS subtypes expressing elevated glutaminase (GLS) levels are highly sensitive to glutamine starvation. In contrast to previous studies, treatment of autochthonous tumour-bearing animals with Telaglenastat (CB-839), an orally bioavailable GLS inhibitor, successfully inhibits undifferentiated pleomorphic sarcoma (UPS) tumour growth. We reveal glutamine metabolism as critical for sarcomagenesis, with CB-839 exhibiting potent therapeutic potential.


Subject(s)
Glutamine/metabolism , Sarcoma/metabolism , Sarcoma/pathology , Allografts/drug effects , Allografts/metabolism , Animals , Benzeneacetamides/pharmacology , Benzeneacetamides/therapeutic use , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Glutaminase/metabolism , Mice , Nucleosides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcoma/diagnostic imaging , Sarcoma/drug therapy , Thiadiazoles/pharmacology , Thiadiazoles/therapeutic use , Tomography, X-Ray Computed
5.
Ann N Y Acad Sci ; 1462(1): 5-13, 2020 02.
Article in English | MEDLINE | ID: mdl-31792987

ABSTRACT

Tumor cells have devised unique metabolic strategies to garner enough nutrients to sustain continuous growth and cell division. Oncogenic mutations may alter metabolic pathways to unlock new sources of energy, and cells take the advantage of various scavenging pathways to ingest material from their environment. These changes in metabolism result in a metabolic profile that, in addition to providing the building blocks for macromolecules, can also influence cell signaling pathways to promote tumor initiation and progression. Understanding what pathways tumor cells use to synthesize the materials necessary to support metabolic growth can pave the way for new cancer therapeutics. Potential strategies include depriving tumors of the materials needed to grow or targeting pathways involved in dependencies that arise by virtue of their altered metabolis.


Subject(s)
Congresses as Topic/trends , Energy Metabolism/physiology , Neoplasms/metabolism , Research Report/trends , Animals , Cell Transformation, Neoplastic/metabolism , Humans , Metabolic Networks and Pathways/physiology , New York City
6.
Cell Metab ; 27(6): 1263-1280.e6, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29754953

ABSTRACT

Kidney cancer, one of the ten most prevalent malignancies in the world, has exhibited increased incidence over the last decade. The most common subtype is "clear cell" renal cell carcinoma (ccRCC), which features consistent metabolic abnormalities, such as highly elevated glycogen and lipid deposition. By integrating metabolomics, genomic, and transcriptomic data, we determined that enzymes in multiple metabolic pathways are universally depleted in human ccRCC tumors, which are otherwise genetically heterogeneous. Notably, the expression of key urea cycle enzymes, including arginase 2 (ARG2) and argininosuccinate synthase 1 (ASS1), is strongly repressed in ccRCC. Reduced ARG2 activity promotes ccRCC tumor growth through at least two distinct mechanisms: conserving the critical biosynthetic cofactor pyridoxal phosphate and avoiding toxic polyamine accumulation. Pharmacological approaches to restore urea cycle enzyme expression would greatly expand treatment strategies for ccRCC patients, where current therapies only benefit a subset of those afflicted with renal cancer.


Subject(s)
Arginase/metabolism , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Polyamines/metabolism , Animals , Arginase/genetics , Argininosuccinate Synthase/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/enzymology , Cell Line, Tumor , Gene Expression Profiling , Heterografts , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/enzymology , Mice , Mice, Nude , Pyridoxal Phosphate/metabolism , Urea/metabolism
7.
Clin Cancer Res ; 22(2): 374-82, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26307133

ABSTRACT

PURPOSE: Copy number alterations have been shown to be involved in melanoma pathogenesis. The randomized phase III clinical trial E2603: carboplatin, paclitaxel, ± sorafenib (CP vs. CPS) offers a large collection of tumor samples to evaluate association of somatic mutations, genomic alterations, and clinical outcomes, prior to current FDA-approved therapies. EXPERIMENTAL DESIGN: Copy number and mutational analysis on 119 pretreatment samples was performed. RESULTS: CPS therapy was associated with improved progression-free survival (PFS) compared with CP in patients with tumors with RAF1 (cRAF) gene copy gains (HR, 0.372; P = 0.025) or CCND1 gene copy gains (HR, 0.45; P = 0.035). CPS therapy was associated with improved overall survival (OS) compared with CP in patients with tumors with KRAS gene copy gains (HR, 0.25; P = 0.035). BRAF gene copy gain and MET amplification were more common in samples with V600K versus V600E mutations (P < 0.001), which was validated in The Cancer Genome Atlas (TCGA) dataset. CONCLUSIONS: We observed improved treatment response with CPS in patients with melanoma whose tumors have RAF1 (cRAF), KRAS, or CCND1 amplification, all of which can be attributed to sorafenib targeting CRAF. These genomic alterations should be incorporated in future studies for evaluation as biomarkers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , DNA Copy Number Variations/drug effects , Melanoma/drug therapy , Melanoma/genetics , Niacinamide/analogs & derivatives , Paclitaxel/therapeutic use , Phenylurea Compounds/therapeutic use , DNA Copy Number Variations/genetics , DNA Mutational Analysis/methods , Disease-Free Survival , Double-Blind Method , Genes, ras/genetics , Humans , Mutation/drug effects , Mutation/genetics , Neoplasm Staging/methods , Niacinamide/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met/genetics , Sorafenib , Treatment Outcome
8.
Nat Commun ; 6: 6140, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25608029

ABSTRACT

Pheochromocytomas and paragangliomas (PCC/PGL) are the solid tumour type most commonly associated with an inherited susceptibility syndrome. However, very little is known about the somatic genetic changes leading to tumorigenesis or malignant transformation. Here we perform whole-exome sequencing on a discovery set of 21 PCC/PGL and identify somatic ATRX mutations in two SDHB-associated tumours. Targeted sequencing of a separate validation set of 103 PCC/PGL identifies somatic ATRX mutations in 12.6% of PCC/PGL. PCC/PGL with somatic ATRX mutations are associated with alternative lengthening of telomeres and clinically aggressive behaviour. This finding suggests that loss of ATRX, an SWI/SNF chromatin remodelling protein, is important in the development of clinically aggressive PCC/PGL.


Subject(s)
Adrenal Gland Neoplasms/genetics , DNA Helicases/genetics , Exome , Head and Neck Neoplasms/genetics , Nuclear Proteins/genetics , Paraganglioma/genetics , Pheochromocytoma/genetics , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Chromatin/chemistry , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Gene Expression , Genetic Predisposition to Disease , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , High-Throughput Nucleotide Sequencing , Humans , Mutation , Nuclear Proteins/metabolism , Paraganglioma/metabolism , Paraganglioma/pathology , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Telomere/ultrastructure , Telomere Homeostasis , X-linked Nuclear Protein
9.
Clin Cancer Res ; 21(7): 1652-64, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25617424

ABSTRACT

PURPOSE: To investigate the roles of melanoma-associated macrophages in melanoma resistance to BRAF inhibitors (BRAFi). EXPERIMENTAL DESIGN: An in vitro macrophage and melanoma cell coculture system was used to investigate whether macrophages play a role in melanoma resistance to BRAFi. The effects of macrophages in tumor resistance were examined by proliferation assay, cell death assay, and Western blot analyses. Furthermore, two mouse preclinical models were used to validate whether targeting macrophages can increase the antitumor activity of BRAFi. Finally, the number of macrophages in melanoma tissues was examined by immunohistochemistry. RESULTS: We demonstrate that in BRAF-mutant melanomas, BRAFi paradoxically activate the mitogen-activated protein kinase (MAPK) pathway in macrophages to produce VEGF, which reactivates the MAPK pathway and stimulates cell growth in melanoma cells. Blocking the MAPK pathway or VEGF signaling then reverses macrophage-mediated resistance. Targeting macrophages increases the antitumor activity of BRAFi in mouse and human tumor models. The presence of macrophages in melanomas predicts early relapse after therapy. CONCLUSIONS: Our findings demonstrate that macrophages play a critical role in melanoma resistance to BRAFi, suggesting that targeting macrophages will benefit patients with BRAF-mutant melanoma.


Subject(s)
Drug Resistance, Neoplasm/immunology , Macrophages/metabolism , Melanoma/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Blotting, Western , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Macrophages/drug effects , Macrophages/immunology , Melanoma/immunology , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy/adverse effects , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering , Transfection , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL