11 research outputs found

    HIF drives lipid deposition and cancer in ccRCC via repression of fatty acid metabolism

    Get PDF
    Clear cell renal cell carcinoma (ccRCC) is histologically defined by its lipid and glycogen-rich cytoplasmic deposits. Alterations in the VHL tumor suppressor stabilizing the hypoxiainducible factors (HIFs) are the most prevalent molecular features of clear cell tumors. The significance of lipid deposition remains undefined. We describe the mechanism of lipid deposition in ccRCC by identifying the rate-limiting component of mitochondrial fatty acid transport, carnitine palmitoyltransferase 1A (CPT1A), as a direct HIF target gene. CPT1A is repressed by HIF1 and HIF2, reducing fatty acid transport into the mitochondria, and forcing fatty acids to lipid droplets for storage. Droplet formation occurs independent of lipid source, but only when CPT1A is repressed. Functionally, repression of CPT1A is critical for tumor formation, as elevated CPT1A expression limits tumor growth. In human tumors, CPT1A expression and activity are decreased versus normal kidney; and poor patient outcome associates with lower expression of CPT1A in tumors in TCGA. Together, our studies identify HIF control of fatty acid metabolism as essential for ccRCC tumorigenesis

    Factor XII and Upar Upregulate Neutrophil Functions to Influence Wound Healing

    Get PDF
    Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12–/–) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor–mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of αMβ2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12–/– mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12–/– hosts was sufficient to correct the neutrophil migration defect in F12–/– mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated

    The L 98-59 System: Three Transiting, Terrestrial-size Planets Orbiting a Nearby M Dwarf

    Get PDF
    We report the Transiting Exoplanet Survey Satellite (TESS) discovery of three terrestrial-size planets transiting L 98-59 (TOI-175, TIC 307210830)—a bright M dwarf at a distance of 10.6 pc. Using the Gaia-measured distance and broadband photometry, we find that the host star is an M3 dwarf. Combined with the TESS transits from three sectors, the corresponding stellar parameters yield planet radii ranging from 0.8 R ⊕ to 1.6 R ⊕. All three planets have short orbital periods, ranging from 2.25 to 7.45 days with the outer pair just wide of a 2:1 period resonance. Diagnostic tests produced by the TESS Data Validation Report and the vetting package DAVE rule out common false-positive sources. These analyses, along with dedicated follow-up and the multiplicity of the system, lend confidence that the observed signals are caused by planets transiting L 98-59 and are not associated with other sources in the field. The L 98-59 system is interesting for a number of reasons: the host star is bright (V = 11.7 mag, K = 7.1 mag) and the planets are prime targets for further follow-up observations including precision radial-velocity mass measurements and future transit spectroscopy with the James Webb Space Telescope; the near-resonant configuration makes the system a laboratory to study planetary system dynamical evolution; and three planets of relatively similar size in the same system present an opportunity to study terrestrial planets where other variables (age, metallicity, etc.) can be held constant. L 98-59 will be observed in four more TESS sectors, which will provide a wealth of information on the three currently known planets and have the potential to reveal additional planets in the system

    SAT1 and glioblastoma multiforme: Disarming the resistance

    No full text
    Glioblastoma multiforme is the most common and most detrimental form of brain tumor, with a current survival time of as little as 14 months. We have recently identified a novel mechanism of therapeutic resistance based on overexpression of the polyamine catabolic enzyme spermidine/spermine N1-acetyltransferase, which promotes DNA repair via chromatin modification

    Abstract 3954: SAT1 (Spermidine/spermine-N1-acetyltrasferase 1) promotes radiation resistance in glioblastoma multiforme

    No full text
    Abstract Glioblastoma multiforme (GBM), is the most common and severe form of brain cancer. The median survival time is approximately 14 months due to poor responses to surgery, radiation and chemotherapy. GBM cells have been shown to exhibit high resistance to radiation. In order to understand the mechanisms involved in radioresistance, we conducted a genetic screen using a shRNA library on GBM cell lines to identify genes whose inhibition would sensitize cells to radiation. The results were cross-referred with the Oncomine and Rembrandt databases to focus on genes that are highly expressed in GBM tumors, and are associated with poor patient outcomes. Spermidine/spermine-N1-acetyltransferase 1 (SAT1), an enzyme involved in polyamine catabolism, was identified as a gene that promotes resistance to ionizing radiation (IR), is overexpressed in brain tumors and its high expression correlates with poor outcomes. The objective of this study is to explore the role of SAT1 in radioresistance with the purpose of targeting SAT1 as a means to sensitize tumors. The knockdown of SAT1 using, shRNA and siRNA approaches, in multiple cell lines and neurosphere lines results in sensitization of GBM cells to radiation in colony formation assays. This was seen specifically in G2/M and S phases, leading to the hypothesis that SAT1 plays a role in homologous recombination (HR). By measuring HR in the DR-GFP reporter system, we confirmed that SAT1 promotes HR, since depletion of SAT1 results in decrease in HR. To test whether SAT1 depletion sensitizes cells to S-phase poisons, SAT1 knockdown GBM cells were then exposed to camptothecin, and were found to be sensitized. To explore the mechanisms of interaction of SAT1 with the DNA damage pathway, chromatin immunoprecipitation (ChIP) was performed under the theory that polyamine-DNA interactions may be altered during DNA repair. The results show that polyamines are decreased in the chromatin of the HR site following induction of double-strand breaks. Interestingly, western blot analyses show that SAT1 knockdown decreases the levels of acetylated histone 3 (H3) proteins, suggesting a new role of SAT1 in chromatin remodeling. Overall the results suggest that SAT1 is involved in radioresistance in GBM through regulation of HR and alterations in chromatin remodeling at sites of damage. Our findings suggest that the biological significance of SAT1 expression in GBM lies in its contribution to cell radioresistance and SAT1 may potentially be a therapeutic target to sensitize GBM to cancer therapies. Citation Format: Adina Brett-Morris, Scott M. Welford, Eli Bar, Raffaella Spina, Bradley Wright, Junran Zhang, Jun Lu, Yuji Seo. SAT1 (Spermidine/spermine-N1-acetyltrasferase 1) promotes radiation resistance in glioblastoma multiforme. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3954. doi:10.1158/1538-7445.AM2014-3954</jats:p

    The polyamine catabolic enzyme SAT1 modulates tumorigenesis and radiation response in GBM

    Get PDF
    Glioblastoma multiforme (GBM) is the most common and severe form of brain cancer. The median survival time of patients is approximately 12 months due to poor responses to surgery and chemoradiation. To understand the mechanisms involved in radioresistance, we conducted a genetic screen using an shRNA library to identify genes in which inhibition would sensitize cells to radiation. The results were cross-referenced with the Oncomine and Rembrandt databases to focus on genes that are highly expressed in GBM tumors and associated with poor patient outcomes. Spermidine/spermine-N1-acetyltransferase 1 (SAT1), an enzyme involved in polyamine catabolism, was identified as a gene that promotes resistance to ionizing radiation (IR), is overexpressed in brain tumors, and correlates with poor outcomes. Knockdown of SAT1 using shRNA and siRNA approaches in multiple cell and neurosphere lines resulted in sensitization of GBM cells to radiation in colony formation assays and tumors, and decreased tumorigenesis in vivo. Radiosensitization occurred specifically in G2-M and S phases, suggesting a role for SAT1 in homologous recombination (HR) that was confirmed in a DR-GFP reporter system. Mechanistically, we found that SAT1 promotes acetylation of histone H3, suggesting a new role of SAT1 in chromatin remodeling and regulation of gene expression. In particular, SAT1 depletion led to a dramatic reduction in BRCA1 expression, explaining decreased HR capacity. Our findings suggest that the biologic significance of elevated SAT1 expression in GBM lies in its contribution to cell radioresistance and that SAT1 may potentially be a therapeutic target to sensitize GBM to cancer therapies

    HIF drives lipid deposition and cancer in ccRCC via repression of fatty acid metabolism

    No full text
    Clear cell renal cell carcinoma (ccRCC) is histologically defined by its lipid and glycogen-rich cytoplasmic deposits. Alterations in the VHL tumor suppressor stabilizing the hypoxiainducible factors (HIFs) are the most prevalent molecular features of clear cell tumors. The significance of lipid deposition remains undefined. We describe the mechanism of lipid deposition in ccRCC by identifying the rate-limiting component of mitochondrial fatty acid transport, carnitine palmitoyltransferase 1A (CPT1A), as a direct HIF target gene. CPT1A is repressed by HIF1 and HIF2, reducing fatty acid transport into the mitochondria, and forcing fatty acids to lipid droplets for storage. Droplet formation occurs independent of lipid source, but only when CPT1A is repressed. Functionally, repression of CPT1A is critical for tumor formation, as elevated CPT1A expression limits tumor growth. In human tumors, CPT1A expression and activity are decreased versus normal kidney; and poor patient outcome associates with lower expression of CPT1A in tumors in TCGA. Together, our studies identify HIF control of fatty acid metabolism as essential for ccRCC tumorigenesis

    Spermidine/spermine N1-acetyltransferase 1 is a gene-specific transcriptional regulator that drives brain tumor aggressiveness

    No full text
    Spermidine/spermine N1-acetyltransferase 1 (SAT1), the rate-limiting enzyme in polyamine catabolism, has broad regulatory roles due to near ubiquitous polyamine binding. We describe a novel function of SAT1 as a gene-specific transcriptional regulator through local polyamine acetylation. SAT1 expression is elevated in aggressive brain tumors and promotes resistance to radiotherapy. Expression profiling in glioma cells identified SAT1 target genes that distinguish high- and low-grade tumors, in support of the prognostic utility of SAT1 expression. We further discovered mechanisms of SAT1-driven tumor aggressiveness through promotion of expression of both DNA damage response pathways as well as cell cycle regulatory genes. Mechanistically, SAT1 associates specifically with the promoter of the MELK gene, which functionally controls other SAT1 targets, and leads biologically to maintenance of neurosphere stemness in conjunction with FOXM1 and EZH2. CRISPR knockin mutants demonstrate the essentiality of the polyamine acetyltransferase activity of SAT1 for its function as a transcriptional regulator. Together, the data demonstrate that gene-specific polyamine removal is a major transcriptional regulatory mechanism active in high-grade gliomas that drives poor outcomes
    corecore