Try a new search

Format these results:

Searched for:

in-biosketch:yes

person:papagt01

Total Results:

63


Targeting Metabolic Bottlenecks in Lung Cancer

Sayin, Volkan I; LeBoeuf, Sarah E; Papagiannakopoulos, Thales
Lung cancer remains one of the most genetically complex, aggressive, and lethal solid malignancies. Understanding how distinct lung cancer mutations give rise to altered nutrient requirements and promote immune evasion in the context of a heterogeneous lung tumor microenvironment is vital for the development of novel personalized therapeutic strategies.
PMID: 31421901
ISSN: 2405-8025
CID: 4046492

Nrf2 Activation Promotes Lung Cancer Metastasis by Inhibiting the Degradation of Bach1

Lignitto, Luca; LeBoeuf, Sarah E; Homer, Harrison; Jiang, Shaowen; Askenazi, Manor; Karakousi, Triantafyllia R; Pass, Harvey I; Bhutkar, Arjun J; Tsirigos, Aristotelis; Ueberheide, Beatrix; Sayin, Volkan I; Papagiannakopoulos, Thales; Pagano, Michele
Approximately 30% of human lung cancers acquire mutations in either Keap1 or Nfe2l2, resulting in the stabilization of Nrf2, the Nfe2l2 gene product, which controls oxidative homeostasis. Here, we show that heme triggers the degradation of Bach1, a pro-metastatic transcription factor, by promoting its interaction with the ubiquitin ligase Fbxo22. Nrf2 accumulation in lung cancers causes the stabilization of Bach1 by inducing Ho1, the enzyme catabolizing heme. In mouse models of lung cancers, loss of Keap1 or Fbxo22 induces metastasis in a Bach1-dependent manner. Pharmacological inhibition of Ho1 suppresses metastasis in a Fbxo22-dependent manner. Human metastatic lung cancer display high levels of Ho1 and Bach1. Bach1 transcriptional signature is associated with poor survival and metastasis in lung cancer patients. We propose that Nrf2 activates a metastatic program by inhibiting the heme- and Fbxo22-mediated degradation of Bach1, and that Ho1 inhibitors represent an effective therapeutic strategy to prevent lung cancer metastasis.
PMID: 31257023
ISSN: 1097-4172
CID: 3967782

TrxR1, Gsr, and oxidative stress determine hepatocellular carcinoma malignancy

McLoughlin, Michael R; Orlicky, David J; Prigge, Justin R; Krishna, Pushya; Talago, Emily A; Cavigli, Ian R; Eriksson, Sofi; Miller, Colin G; Kundert, Jean A; Sayin, Volkan I; Sabol, Rachel A; Heinemann, Joshua; Brandenberger, Luke O; Iverson, Sonya V; Bothner, Brian; Papagiannakopoulos, Thales; Shearn, Colin T; Arnér, Elias S J; Schmidt, Edward E
Thioredoxin reductase-1 (TrxR1)-, glutathione reductase (Gsr)-, and Nrf2 transcription factor-driven antioxidant systems form an integrated network that combats potentially carcinogenic oxidative damage yet also protects cancer cells from oxidative death. Here we show that although unchallenged wild-type (WT), TrxR1-null, or Gsr-null mouse livers exhibited similarly low DNA damage indices, these were 100-fold higher in unchallenged TrxR1/Gsr-double-null livers. Notwithstanding, spontaneous cancer rates remained surprisingly low in TrxR1/Gsr-null livers. All genotypes, including TrxR1/Gsr-null, were susceptible to N-diethylnitrosamine (DEN)-induced liver cancer, indicating that loss of these antioxidant systems did not prevent cancer cell survival. Interestingly, however, following DEN treatment, TrxR1-null livers developed threefold fewer tumors compared with WT livers. Disruption of TrxR1 in a marked subset of DEN-initiated cancer cells had no effect on their subsequent contributions to tumors, suggesting that TrxR1-disruption does not affect cancer progression under normal care, but does decrease the frequency of DEN-induced cancer initiation. Consistent with this idea, TrxR1-null livers showed altered basal and DEN-exposed metabolomic profiles compared with WT livers. To examine how oxidative stress influenced cancer progression, we compared DEN-induced cancer malignancy under chronically low oxidative stress (TrxR1-null, standard care) vs. elevated oxidative stress (TrxR1/Gsr-null livers, standard care or phenobarbital-exposed TrxR1-null livers). In both cases, elevated oxidative stress was correlated with significantly increased malignancy. Finally, although TrxR1-null and TrxR1/Gsr-null livers showed strong Nrf2 activity in noncancerous hepatocytes, there was no correlation between malignancy and Nrf2 expression within tumors across genotypes. We conclude that TrxR1, Gsr, Nrf2, and oxidative stress are major determinants of liver cancer but in a complex, context-dependent manner.
PMCID:6561278
PMID: 31097586
ISSN: 1091-6490
CID: 4000002

Optofluidic real-time cell sorter for longitudinal CTC studies in mouse models of cancer

Hamza, Bashar; Ng, Sheng Rong; Prakadan, Sanjay M; Delgado, Francisco Feijó; Chin, Christopher R; King, Emily M; Yang, Lucy F; Davidson, Shawn M; DeGouveia, Kelsey L; Cermak, Nathan; Navia, Andrew W; Winter, Peter S; Drake, Riley S; Tammela, Tuomas; Li, Carman Man-Chung; Papagiannakopoulos, Thales; Gupta, Alejandro J; Shaw Bagnall, Josephine; Knudsen, Scott M; Vander Heiden, Matthew G; Wasserman, Steven C; Jacks, Tyler; Shalek, Alex K; Manalis, Scott R
Circulating tumor cells (CTCs) play a fundamental role in cancer progression. However, in mice, limited blood volume and the rarity of CTCs in the bloodstream preclude longitudinal, in-depth studies of these cells using existing liquid biopsy techniques. Here, we present an optofluidic system that continuously collects fluorescently labeled CTCs from a genetically engineered mouse model (GEMM) for several hours per day over multiple days or weeks. The system is based on a microfluidic cell sorting chip connected serially to an unanesthetized mouse via an implanted arteriovenous shunt. Pneumatically controlled microfluidic valves capture CTCs as they flow through the device, and CTC-depleted blood is returned back to the mouse via the shunt. To demonstrate the utility of our system, we profile CTCs isolated longitudinally from animals over 4 days of treatment with the BET inhibitor JQ1 using single-cell RNA sequencing (scRNA-Seq) and show that our approach eliminates potential biases driven by intermouse heterogeneity that can occur when CTCs are collected across different mice. The CTC isolation and sorting technology presented here provides a research tool to help reveal details of how CTCs evolve over time, allowing studies to credential changes in CTCs as biomarkers of drug response and facilitating future studies to understand the role of CTCs in metastasis.
PMCID:6369805
PMID: 30674677
ISSN: 1091-6490
CID: 5507582

Recurrent homozygous deletion of DROSHA and microduplication of PDE4DIP in pineoblastoma

Snuderl, Matija; Kannan, Kasthuri; Pfaff, Elke; Wang, Shiyang; Stafford, James M; Serrano, Jonathan; Heguy, Adriana; Ray, Karina; Faustin, Arline; Aminova, Olga; Dolgalev, Igor; Stapleton, Stacie L; Zagzag, David; Chiriboga, Luis; Gardner, Sharon L; Wisoff, Jeffrey H; Golfinos, John G; Capper, David; Hovestadt, Volker; Rosenblum, Marc K; Placantonakis, Dimitris G; LeBoeuf, Sarah E; Papagiannakopoulos, Thales Y; Chavez, Lukas; Ahsan, Sama; Eberhart, Charles G; Pfister, Stefan M; Jones, David T W; Karajannis, Matthias A
Pineoblastoma is a rare and highly aggressive brain cancer of childhood, histologically belonging to the spectrum of primitive neuroectodermal tumors. Patients with germline mutations in DICER1, a ribonuclease involved in microRNA processing, have increased risk of pineoblastoma, but genetic drivers of sporadic pineoblastoma remain unknown. Here, we analyzed pediatric and adult pineoblastoma samples (n = 23) using a combination of genome-wide DNA methylation profiling and whole-exome sequencing or whole-genome sequencing. Pediatric and adult pineoblastomas showed distinct methylation profiles, the latter clustering with lower-grade pineal tumors and normal pineal gland. Recurrent variants were found in genes involved in PKA- and NF-κB signaling, as well as in chromatin remodeling genes. We identified recurrent homozygous deletions of DROSHA, acting upstream of DICER1 in microRNA processing, and a novel microduplication involving chromosomal region 1q21 containing PDE4DIP (myomegalin), comprising the ancient DUF1220 protein domain. Expresion of PDE4DIP and DUF1220 proteins was present exclusively in pineoblastoma with PDE4DIP gain.
PMCID:6054684
PMID: 30030436
ISSN: 2041-1723
CID: 3202352

Aspartate is a limiting metabolite for cancer cell proliferation under hypoxia and in tumours

Garcia-Bermudez, Javier; Baudrier, Lou; La, Konnor; Zhu, Xiphias Ge; Fidelin, Justine; Sviderskiy, Vladislav O; Papagiannakopoulos, Thales; Molina, Henrik; Snuderl, Matija; Lewis, Caroline A; Possemato, Richard L; Birsoy, Kıvanç
As oxygen is essential for many metabolic pathways, tumour hypoxia may impair cancer cell proliferation1-4. However, the limiting metabolites for proliferation under hypoxia and in tumours are unknown. Here, we assessed proliferation of a collection of cancer cells following inhibition of the mitochondrial electron transport chain (ETC), a major metabolic pathway requiring molecular oxygen 5 . Sensitivity to ETC inhibition varied across cell lines, and subsequent metabolomic analysis uncovered aspartate availability as a major determinant of sensitivity. Cell lines least sensitive to ETC inhibition maintain aspartate levels by importing it through an aspartate/glutamate transporter, SLC1A3. Genetic or pharmacologic modulation of SLC1A3 activity markedly altered cancer cell sensitivity to ETC inhibitors. Interestingly, aspartate levels also decrease under low oxygen, and increasing aspartate import by SLC1A3 provides a competitive advantage to cancer cells at low oxygen levels and in tumour xenografts. Finally, aspartate levels in primary human tumours negatively correlate with the expression of hypoxia markers, suggesting that tumour hypoxia is sufficient to inhibit ETC and, consequently, aspartate synthesis in vivo. Therefore, aspartate may be a limiting metabolite for tumour growth, and aspartate availability could be targeted for cancer therapy.
PMCID:6030478
PMID: 29941933
ISSN: 1476-4679
CID: 3161882

Recurrent homozygous deletion of DROSHA and microduplication of PDE4DIP containing the ancestral DUF1220 domain in pineoblastoma [Meeting Abstract]

Snuderl, M; Kannan, K; Pfaff, E; Wang, S; Stafford, J; Serrano, J; Heguy, A; Ray, K; Faustin, A; Aminova, O; Dolgalev, I; Stapleton, S; Zagzag, D; Chiriboga, L; Gardner, S; Wisoff, J; Golfinos, J; Capper, D; Hovestadt, V; Rosenblum, M; Placantonakis, D; LeBoeuf, S; Papagiannakopoulos, T; Chavez, L; Ahsan, S; Eberhart, C; Pfister, S; Jones, D; Karajannis, M
BACKGROUND: Pineoblastoma is a rare and highly aggressive brain cancer of childhood, histologically belonging to the spectrum of primitive neuroectodermal tumors. Patients with germline mutations in DICER1, a ribonuclease involved in microRNA processing, have increased risk of pineoblastoma, but genetic drivers of sporadic pineoblastoma remain unknown. METHODS: We analyzed pediatric and adult pineoblastoma samples (n=23) using integrated genomic studies, including genome-wide DNA methylation profiling, whole-exome or whole-genome sequencing, and whole-transcriptome analysis. RESULTS: Pediatric and adult pineoblastomas showed distinct methylation profiles, the latter clustering with lower grade pineal tumors and normal pineal gland. Recurrent somatic mutations were found in genes involved in PKA-and NF-kappaB signaling, as well as in chromatin remodeling genes. We identified recurrent homozygous deletions of DROSHA, acting upstream of DICER1 in microRNA processing, and a novel microduplication involving chromosomal region 1q21 containing PDE4DIP (myomegalin), comprising the ancient DUF1220 protein domain. Expression of PDE4DIP and DUF1220 proteins was present exclusively in pineoblastoma with PDE4DIP gain. Whole-transcriptome analysis showed that homozygous loss of DROSHA led to distinct changes in RNA expression profile. Disruption of the DROSHA locus in human neural stem cells using the CRISPR/Cas9 system, led to decrease of the DROSHA protein, and massive loss of miRNAs. CONCLUSION: We identified recurrent homozygous deletions of DROSHA in pineoblastoma, suggesting that different mechanisms disrupting miRNA processing are involved in the pathogenesis of familial versus sporadic pineoblastoma. Furthermore, a novel microduplication of PDE4DIP leading to upregulation of DUF1220 protein suggests DUF1220 as a novel oncogenic driver in pineoblastoma
EMBASE:623098707
ISSN: 1523-5866
CID: 3211282

Loss of Keap1 promotes KRAS-driven lung cancer and results in genotype-specific vulnerabilities. [Meeting Abstract]

Romero, Rodrigo; Sayin, Volkan I.; Shawn, Davidson M.; Bauer, Matthew; Singh, Simranjit X.; LeBoeuf, Sarah; Karakousi, Triantafyllia R.; Ellis, Donald C.; Bhutkar, Arjun; Sanchez-Rivera, Francisco; Subbaraj, Lakshmipriya; Martinez, Britney; Bronson, Roderick T.; Prigge, Justin R.; Schmidt, Edward E.; Thomas, Craig J.; Davies, Angela; Dolgalev, Igor; Heguy, Adriana; Allaj, Viola; Piorier, John T.; Moreira, Andre L.; Rudin, Charles M.; Pass, Harvey I.; Heiden, Matthew G. Vander; Jacks, Tyler; Papagiannakopoulos, Thales
ISI:000432307300068
ISSN: 0008-5472
CID: 3132562

Serine Catabolism by SHMT2 Is Required for Proper Mitochondrial Translation Initiation and Maintenance of Formylmethionyl-tRNAs

Minton, Denise R; Nam, Minwoo; McLaughlin, Daniel J; Shin, Jong; Bayraktar, Erol C; Alvarez, Samantha W; Sviderskiy, Vladislav O; Papagiannakopoulos, Thales; Sabatini, David M; Birsoy, Kıvanç; Possemato, Richard
Upon glucose restriction, eukaryotic cells upregulate oxidative metabolism to maintain homeostasis. Using genetic screens, we find that the mitochondrial serine hydroxymethyltransferase (SHMT2) is required for robust mitochondrial oxygen consumption and low glucose proliferation. SHMT2 catalyzes the first step in mitochondrial one-carbon metabolism, which, particularly in proliferating cells, produces tetrahydrofolate (THF)-conjugated one-carbon units used in cytoplasmic reactions despite the presence of a parallel cytoplasmic pathway. Impairing cytoplasmic one-carbon metabolism or blocking efflux of one-carbon units from mitochondria does not phenocopy SHMT2 loss, indicating that a mitochondrial THF cofactor is responsible for the observed phenotype. The enzyme MTFMT utilizes one such cofactor, 10-formyl THF, producing formylmethionyl-tRNAs, specialized initiator tRNAs necessary for proper translation of mitochondrially encoded proteins. Accordingly, SHMT2 null cells specifically fail to maintain formylmethionyl-tRNA pools and mitochondrially encoded proteins, phenotypes similar to those observed in MTFMT-deficient patients. These findings provide a rationale for maintaining a compartmentalized one-carbon pathway in mitochondria.
PMCID:5819360
PMID: 29452640
ISSN: 1097-4164
CID: 2958432

Elevated Nrf-2 responses are insufficient to mitigate protein carbonylation in hepatospecific PTEN deletion mice

Petersen, Dennis R; Saba, Laura M; Sayin, Volkan I; Papagiannakopoulos, Thales; Schmidt, Edward E; Merrill, Gary F; Orlicky, David J; Shearn, Colin T
OBJECTIVE:In the liver, a contributing factor in the pathogenesis of non-alcoholic fatty liver disease (NASH) is oxidative stress, which leads to the accumulation of highly reactive electrophilic α/β unsaturated aldehydes. The objective of this study was to determine the impact of NASH on protein carbonylation and antioxidant responses in a murine model. METHODS:Liver-specific phosphatase and tensin homolog (PTEN)-deletion mice (PTENLKO) or control littermates were fed a standard chow diet for 45-55 weeks followed by analysis for liver injury, oxidative stress and inflammation. RESULTS:Histology and Picrosirius red-staining of collagen deposition within the extracellular matrix revealed extensive steatosis and fibrosis in the PTENLKO mice but no steatosis or fibrosis in controls. Increased steatosis and fibrosis corresponded with significant increases in inflammation. PTEN-deficient livers showed significantly increased cell-specific oxidative damage, as detected by 4-hydroxy-2-nonenal (4-HNE) and acrolein staining. Elevated staining correlated with an increase in nuclear DNA repair foci (γH2A.X) and cellular proliferation index (Ki67) within zones 1 and 3, indicating oxidative damage was zonally restricted and was associated with increased DNA damage and cell proliferation. Immunoblots showed that total levels of antioxidant response proteins induced by nuclear factor erythroid-2-like-2 (Nrf2), including GSTμ, GSTπ and CBR1/3, but not HO-1, were elevated in PTENLKO as compared to controls, and IHC showed this response also occurred only in zones 1 and 3. Furthermore, an analysis of autophagy markers revealed significant elevation of p62 and LC3II expression. Mass spectrometric (MS) analysis identified significantly more carbonylated proteins in whole cell extracts prepared from PTENLKO mice (966) as compared to controls (809). Pathway analyses of identified proteins did not uncover specific pathways that were preferentially carbonylated in PTENLKO livers but, did reveal specific strongly increased carbonylation of thioredoxin reductase and of glutathione-S-transferases (GST) M6, O1, and O2. CONCLUSIONS:Results show that disruption of PTEN resulted in steatohepatitis, fibrosis and caused hepatic induction of the Nrf2-dependent antioxidant system at least in part due to elevation of p62. This response was both cell-type and zone specific. However, these responses were insufficient to mitigate the accumulation of products of lipid peroxidation.
PMCID:5969769
PMID: 29799837
ISSN: 1932-6203
CID: 3136192