Samuel Peña-Llopis
University of Texas Southwestern Medical Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Samuel Peña-Llopis.
Nature Genetics | 2012
Samuel Peña-Llopis; Silvia Vega-Rubin-de-Celis; Arnold Liao; Nan Leng; Andrea Pavia-Jimenez; Shanshan Wang; Toshinari Yamasaki; Leah Zhrebker; Sharanya Sivanand; Patrick Spence; Lisa N. Kinch; Tina Hambuch; Suneer Jain; Yair Lotan; Vitaly Margulis; Arthur I. Sagalowsky; Pia Banerji Summerour; Wareef Kabbani; S. W. Wendy Wong; Nick V. Grishin; Marc Laurent; Xian Jin Xie; Christian D. Haudenschild; Mark T. Ross; David R. Bentley; Payal Kapur; James Brugarolas
The molecular pathogenesis of renal cell carcinoma (RCC) is poorly understood. Whole-genome and exome sequencing followed by innovative tumorgraft analyses (to accurately determine mutant allele ratios) identified several putative two-hit tumor suppressor genes, including BAP1. The BAP1 protein, a nuclear deubiquitinase, is inactivated in 15% of clear cell RCCs. BAP1 cofractionates with and binds to HCF-1 in tumorgrafts. Mutations disrupting the HCF-1 binding motif impair BAP1-mediated suppression of cell proliferation but not deubiquitination of monoubiquitinated histone 2A lysine 119 (H2AK119ub1). BAP1 loss sensitizes RCC cells in vitro to genotoxic stress. Notably, mutations in BAP1 and PBRM1 anticorrelate in tumors (P = 3 × 10−5), and combined loss of BAP1 and PBRM1 in a few RCCs was associated with rhabdoid features (q = 0.0007). BAP1 and PBRM1 regulate seemingly different gene expression programs, and BAP1 loss was associated with high tumor grade (q = 0.0005). Our results establish the foundation for an integrated pathological and molecular genetic classification of RCC, paving the way for subtype-specific treatments exploiting genetic vulnerabilities.
The EMBO Journal | 2011
Samuel Peña-Llopis; Silvia Vega-Rubin-de-Celis; Jacob C. Schwartz; Nicholas C. Wolff; Tram Anh T. Tran; Lihua Zou; Xian Jin Xie; David R. Corey; James Brugarolas
Mammalian target of rapamycin (mTOR) complex 1 (mTORC1) is an important, highly conserved, regulator of cell growth. Ancient among the signals that regulate mTORC1 are nutrients. Amino acids direct mTORC1 to the surface of the late endosome/lysosome, where mTORC1 becomes receptive to other inputs. However, the interplay between endosomes and mTORC1 is poorly understood. Here, we report the discovery of a network that links mTORC1 to a critical component of the late endosome/lysosome, the V‐ATPase. In an unbiased screen, we found that mTORC1 regulated the expression of, among other lysosomal genes, the V‐ATPases. mTORC1 regulates V‐ATPase expression both in cells and in mice. V‐ATPase regulation by mTORC1 involves a transcription factor translocated in renal cancer, TFEB. TFEB is required for the expression of a large subset of mTORC1 responsive genes. mTORC1 coordinately regulates TFEB phosphorylation and nuclear localization and in a manner dependent on both TFEB and V‐ATPases, mTORC1 promotes endocytosis. These data uncover a regulatory network linking an oncogenic transcription factor that is a master regulator of lysosomal biogenesis, TFEB, to mTORC1 and endocytosis.
Lancet Oncology | 2013
Payal Kapur; Samuel Peña-Llopis; Alana Christie; Leah Zhrebker; Andrea Pavia-Jimenez; W.Kimryn Rathmell; Xian Jin Xie; James Brugarolas
BACKGROUND Clear-cell renal-cell carcinomas display divergent clinical behaviours. However, the molecular genetic events driving these behaviours are unknown. We discovered that BAP1 is mutated in about 15% of clear-cell renal-cell carcinoma, and that BAP1 and PBRM1 mutations are largely mutually exclusive. The aim of this study was to investigate the clinicopathological significance of these molecular subtypes and to determine whether patients with BAP1-mutant and PBRM1-mutant tumours had different overall survival. METHODS In this retrospective analysis, we assessed 145 patients with primary clear-cell renal-cell carcinoma and defined PBRM1 and BAP1 mutation status from the University of Texas Southwestern Medical Center (UTSW), TX, USA, between 1998 and 2011. We classified patients into those with BAP1-mutant tumours and those with tumours exclusively mutated for PBRM1 (PBRM1-mutant). We used a second independent cohort (n=327) from The Cancer Genome Atlas (TCGA) for validation. In both cohorts, more than 80% of patients had localised or locoregional disease at presentation. Overall both cohorts were similar, although the TCGA had more patients with metastatic and higher-grade disease, and more TCGA patients presented before molecularly targeted therapies became available. FINDINGS The median overall survival in the UTSW cohort was significantly shorter for patients with BAP1-mutant tumours (4·6 years; 95% CI 2·1-7·2), than for patients with PBRM1-mutant tumours (10·6 years; 9·8-11·5), corresponding to a HR of 2·7 (95% CI 0·99-7·6, p=0·044). Median overall survival in the TCGA cohort was 1·9 years (95% CI 0·6-3·3) for patients with BAP1-mutant tumours and 5·4 years (4·0-6·8) for those with PBRM1-mutant tumours. A HR similar to the UTSW cohort was noted in the TCGA cohort (2·8; 95% CI 1·4-5·9; p=0·004). Patients with mutations in both BAP1 and PBRM1, although a minority (three in UTSW cohort and four in TCGA cohort), had the worst overall survival (median 2·1 years, 95% CI 0·3-3·8, for the UTSW cohort, and 0·2 years, 0·0-1·2, for the TCGA cohort). INTERPRETATION Our findings identify mutation-defined subtypes of clear-cell renal-cell carcinoma with distinct clinical outcomes, a high-risk BAP1-mutant group and a favourable PBRM1-mutant group. These data establish the basis for a molecular genetic classification of clear-cell renal-cell carcinoma that could influence treatment decisions in the future. The existence of different molecular subtypes with disparate outcomes should be considered in the design and assessment of clinical studies. FUNDING Cancer Prevention and Research Institution of Texas and National Cancer Institute.
Nature Genetics | 2015
Steffen Durinck; Eric Stawiski; Andrea Pavia-Jimenez; Zora Modrusan; Payal Kapur; Bijay S. Jaiswal; Na Zhang; Vanina Toffessi-Tcheuyap; Thong T. Nguyen; Kanika Bajaj Pahuja; Ying Jiun Chen; Sadia Saleem; Subhra Chaudhuri; Sherry Heldens; Marlena Jackson; Samuel Peña-Llopis; Joseph Guillory; Karen Toy; Connie Ha; Corissa J. Harris; Eboni Holloman; Haley Hill; Jeremy Stinson; Celina Sanchez Rivers; Vasantharajan Janakiraman; Weiru Wang; Lisa N. Kinch; Nick V. Grishin; Peter M. Haverty; Bernard Chow
To further understand the molecular distinctions between kidney cancer subtypes, we analyzed exome, transcriptome and copy number alteration data from 167 primary human tumors that included renal oncocytomas and non–clear cell renal cell carcinomas (nccRCCs), consisting of papillary (pRCC), chromophobe (chRCC) and translocation (tRCC) subtypes. We identified ten significantly mutated genes in pRCC, including MET, NF2, SLC5A3, PNKD and CPQ. MET mutations occurred in 15% (10/65) of pRCC samples and included previously unreported recurrent activating mutations. In chRCC, we found TP53, PTEN, FAAH2, PDHB, PDXDC1 and ZNF765 to be significantly mutated. Gene expression analysis identified a five-gene set that enabled the molecular classification of chRCC, renal oncocytoma and pRCC. Using RNA sequencing, we identified previously unreported gene fusions, including ACTG1-MITF fusion. Ectopic expression of the ACTG1-MITF fusion led to cellular transformation and induced the expression of downstream target genes. Finally, we observed upregulation of the anti-apoptotic factor BIRC7 in MiTF-high RCC tumors, suggesting a potential therapeutic role for BIRC7 inhibitors.
Nature Communications | 2013
Lei Wang; Jianjun Chang; Diana Varghese; Michael T. Dellinger; Subodh Kumar; Anne M. Best; Julio C. Ruiz; Richard K. Bruick; Samuel Peña-Llopis; Junjie Xu; David J. Babinski; Doug E. Frantz; Rolf A. Brekken; Amy Quinn; Anton Simeonov; Johnny Easmon; Elisabeth D. Martinez
The pharmacological inhibition of general transcriptional regulators has the potential to block growth through targeting multiple tumorigenic signaling pathways simultaneously. Here, using an innovative cell-based screen, we identify a structurally unique small molecule (named JIB-04) which specifically inhibits the activity of the Jumonji family of histone demethylases in vitro, in cancer cells, and in tumors in vivo. Unlike known inhibitors, JIB-04 is not a competitive inhibitor of α-ketoglutarate. In cancer but not in patient-matched normal cells, JIB-04 alters a subset of transcriptional pathways and blocks viability. In mice, JIB-04 reduces tumor burden and prolongs survival. Importantly, we find that patients with breast tumors that overexpress Jumonji demethylases have significantly lower survival. Thus JIB-04, a novel inhibitor of Jumonji demethylases in vitro and in vivo, constitutes a unique potential therapeutic and research tool against cancer, and validates the use of unbiased cellular screens to discover chemical modulators with disease relevance.
Science Translational Medicine | 2012
Sharanya Sivanand; Samuel Peña-Llopis; Hong Zhao; Blanka Kucejova; Patrick Spence; Andrea Pavia-Jimenez; Toshinari Yamasaki; David McBride; Jessica Gillen; Nicholas C. Wolff; Lorraine K. Morlock; Yair Lotan; Ganesh V. Raj; Arthur I. Sagalowsky; Vitaly Margulis; Jeffrey A. Cadeddu; Mark T. Ross; David R. Bentley; Wareef Kabbani; Xian Jin Xie; Payal Kapur; Noelle S. Williams; James Brugarolas
Extensively validated tumorgraft model shows activity of investigational agent dovitinib against renal cell carcinoma. Grafting a Better Cancer Model When it comes to predicting drug responsiveness in cancer patients, the standard mouse models (xenografts) get low marks: Drugs that work in these mice are frequently ineffective in humans. Xenograft models are created by injecting human tumor cell lines—which often acquire new mutations in culture—into immunocompromised mice. The resulting tumors are generally different from the original tumor. Tumorgrafts, instead created by implanting fragments of human tumors directly into mice, are generating new excitement among some researchers. Sivanand et al. now describe and validate a tumorgraft model of renal cell carcinoma (RCC) that shows promise for preclinical drug studies. The scientists implanted small fragments of RCC tumor from 94 patients into mice, placing the fragments under the fibrous capsule that surrounds the kidney. Sixteen stable tumor lines—which could be serially passaged to new mice—were ultimately established. Examination by a clinical pathologist revealed that the tumorgrafts were quite similar histologically to the original tumors; gene expression patterns, DNA copy number changes, and most mutations in protein-coding regions were also preserved. In addition, mice bearing tumorgrafts from patients that developed elevated concentrations of serum calcium similarly developed tumor-induced hypercalcemia. Moreover, tumorgraft growth was inhibited by two drugs (sunitinib, approved for treating RCC, and sirolimus, the active metabolite of an approved RCC drug), but did not respond to a lung cancer drug that is inactive against RCC. Finally, dovitinib, an inhibitor of several growth factor receptors that is being studied in clinical trials, suppressed the tumorgrafts more potently than sunitinib or sirolimus, suggesting that dovitinib would be active against RCC in humans. These results apply more broadly: The tumorgrafts should be useful for testing other targeted drugs preclinically—an important need, given that most anticancer drugs that enter clinical trials fail to gain approval. Most anticancer drugs entering clinical trials fail to achieve approval from the U.S. Food and Drug Administration. Drug development is hampered by the lack of preclinical models with therapeutic predictive value. Herein, we report the development and validation of a tumorgraft model of renal cell carcinoma (RCC) and its application to the evaluation of an experimental drug. Tumor samples from 94 patients were implanted in the kidneys of mice without additives or disaggregation. Tumors from 35 of these patients formed tumorgrafts, and 16 stable lines were established. Samples from metastatic sites engrafted at higher frequency than those from primary tumors, and stable engraftment of primary tumors in mice correlated with decreased patient survival. Tumorgrafts retained the histology, gene expression, DNA copy number alterations, and more than 90% of the protein-coding gene mutations of the corresponding tumors. As determined by the induction of hypercalcemia in tumorgraft-bearing mice, tumorgrafts retained the ability to induce paraneoplastic syndromes. In studies simulating drug exposures in patients, RCC tumorgraft growth was inhibited by sunitinib and sirolimus (the active metabolite of temsirolimus in humans), but not by erlotinib, which was used as a control. Dovitinib, a drug in clinical development, showed greater activity than sunitinib and sirolimus. The routine incorporation of models recapitulating the molecular genetics and drug sensitivities of human tumors into preclinical programs has the potential to improve oncology drug development.
Molecular Cancer Research | 2013
Megan N. Farley; Laura S. Schmidt; Jessica Mester; Samuel Peña-Llopis; Andrea Pavia-Jimenez; Alana Christie; Cathy D. Vocke; Christopher J. Ricketts; James Peterson; Lindsay A. Middelton; Lisa N. Kinch; Nick V. Grishin; Maria J. Merino; Adam R. Metwalli; Chao Xing; Xian Jin Xie; Patricia L M Dahia; Charis Eng; W. Marston Linehan; James Brugarolas
Renal cell carcinoma (RCC) clusters in some families. Familial RCC arises from mutations in several genes, including the von Hippel-Lindau (VHL) tumor suppressor, which is also mutated in sporadic RCC. However, a significant percentage of familial RCC remains unexplained. Recently, we discovered that the BRCA1-associated protein-1 (BAP1) gene is mutated in sporadic RCC. The BAP1 gene encodes a nuclear deubiquitinase and appears to be a classic two-hit tumor suppressor gene. Somatic BAP1 mutations are associated with high-grade, clear-cell RCC (ccRCC) and poor patient outcomes. To determine whether BAP1 predisposes to familial RCC, the BAP1 gene was sequenced in 83 unrelated probands with unexplained familial RCC. Interestingly, a novel variant (c.41T>A; p.L14H) was uncovered that cosegregated with the RCC phenotype. The p.L14H variant targets a highly conserved residue in the catalytic domain, which is frequently targeted by missense mutations. The family with the novel BAP1 variant was characterized by early-onset ccRCC, occasionally of high Fuhrman grade, and lacked other features that typify VHL syndrome. These findings suggest that BAP1 is an early-onset familial RCC predisposing gene. Implications: BAP1 mutations may drive tumor development in a subset of patients with inherited renal cell cancer. Mol Cancer Res; 11(9); 1061–71. ©2013 AACR.
Molecular Cancer Research | 2011
Blanka Kucejova; Samuel Peña-Llopis; Toshinari Yamasaki; Sharanya Sivanand; Tram Anh T. Tran; Shane Alexander; Nicholas C. Wolff; Yair Lotan; Xian Jin Xie; Wareef Kabbani; Payal Kapur; James Brugarolas
mTOR complex 1 (mTORC1) is implicated in cell growth control and is extensively regulated. We previously reported that in response to hypoxia, mTORC1 is inhibited by the protein regulated in development and DNA damage response 1 (REDD1). REDD1 is upregulated by hypoxia-inducible factor (HIF)-1, and forced REDD1 expression is sufficient to inhibit mTORC1. REDD1-induced mTORC1 inhibition is dependent on a protein complex formed by the tuberous sclerosis complex (TSC)1 and 2 (TSC2) proteins. In clear-cell renal cell carcinoma (ccRCC), the von Hippel-Lindau (VHL) gene is frequently inactivated leading to constitutive activation of HIF-2 and/or HIF-1, which may be expected to upregulate REDD1 and inhibit mTORC1. However, mTORC1 is frequently activated in ccRCC, and mTORC1 inhibitors are effective against this tumor type; a paradox herein examined. REDD1 was upregulated in VHL-deficient ccRCC by in silico microarray analyses, as well as by quantitative real-time PCR, Western blot, and immunohistochemistry. Vhl disruption in a mouse model was sufficient to induce Redd1. Using ccRCC-derived cell lines, we show that REDD1 upregulation in tumors is VHL dependent and that both HIF-1 and HIF-2 are, in a cell-type-dependent manner, recruited to, and essential for, REDD1 induction. Interestingly, whereas mTORC1 is responsive to REDD1 in some tumors, strategies have evolved in others, such as mutations disrupting TSC1, to subvert mTORC1 inhibition by REDD1. Sequencing analyses of 77 ccRCCs for mutations in TSC1, TSC2, and REDD1, using PTEN as a reference, implicate the TSC1 gene, and possibly REDD1, as tumor suppressors in sporadic ccRCC. Understanding how ccRCCs become refractory to REDD1-induced mTORC1 inhibition should shed light into the development of ccRCC and may aid in patient selection for molecular-targeted therapies. Mol Cancer Res; 9(9); 1255–65. ©2011 AACR.
Nature Chemical Biology | 2011
Miguel A. Mata; Neal Satterly; Gijs A. Versteeg; Doug E. Frantz; Shuguang Wei; Noelle S. Williams; Mirco Schmolke; Samuel Peña-Llopis; James Brugarolas; Christian V. Forst; Michael A. White; Adolfo García-Sastre; Michael G. Roth; Beatriz M. A. Fontoura
A chemical genetics approach was taken to identify inhibitors of NS1, a major influenza A virus virulence factor that inhibits host gene expression. A high-throughput screen of 200,000 synthetic compounds identified small molecules that reversed NS1-mediated inhibition of host gene expression. A counterscreen for suppression of influenza virus cytotoxicity identified naphthalimides that inhibited replication of influenza virus and vesicular stomatitis virus (VSV). The mechanism of action occurs through activation of REDD1 expression and concomitant inhibition of mammalian target of rapamycin complex 1 (mTORC1) via TSC1-TSC2 complex. The antiviral activity of naphthalimides was abolished in REDD1(-/-) cells. Inhibition of REDD1 expression by viruses resulted in activation of the mTORC1 pathway. REDD1(-/-) cells prematurely upregulated viral proteins via mTORC1 activation and were permissive to virus replication. In contrast, cells conditionally expressing high concentrations of REDD1 downregulated the amount of viral protein. Thus, REDD1 is a new host defense factor, and chemical activation of REDD1 expression represents a potent antiviral intervention strategy.
Cancer Research | 2013
Samuel Peña-Llopis; Alana Christie; Xian Jin Xie; James Brugarolas
It is poorly understood how driver mutations in cancer genes work together to promote tumor development. Renal cell carcinoma (RCC) offers a unique opportunity to study complex relationships among cancer genes. The four most commonly mutated genes in RCC of clear-cell type (the most common type) are two-hit tumor suppressor genes, and they cluster in a 43-Mb region on chromosome 3p that is deleted in approximately 90% of tumors: VHL (mutated in ∼80%), PBRM1 (∼50%), BAP1 (∼15%), and SETD2 (∼15%). Meta-analyses that we conducted show that mutations in PBRM1 and SETD2 co-occur in tumors at a frequency higher than expected by chance alone, indicating that these mutations may cooperate in tumorigenesis. In contrast, consistent with our previous results, mutations in PBRM1 and BAP1 tend to be mutually exclusive. Mutation exclusivity analyses (often confounded by lack of statistical power) raise the possibility of functional redundancy. However, mutation exclusivity may indicate negative genetic interactions, as proposed herein for PBRM1 and BAP1, and mutations in these genes define RCC with different pathologic features, gene expression profiles, and outcomes. Negative genetic interactions among cancer genes point toward broader context dependencies of cancer gene action beyond tissue dependencies. An enhanced understanding of cancer gene dependencies may help to unravel vulnerabilities that can be exploited therapeutically.