Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Miguel Rivera is active.

Publication


Featured researches published by Miguel Rivera.


Nature Genetics | 2004

Detection of large-scale variation in the human genome

A. John Iafrate; Lars Feuk; Miguel Rivera; Marc Listewnik; Patricia K. Donahoe; Ying Qi; Stephen W. Scherer; Charles Lee

We identified 255 loci across the human genome that contain genomic imbalances among unrelated individuals. Twenty-four variants are present in > 10% of the individuals that we examined. Half of these regions overlap with genes, and many coincide with segmental duplications or gaps in the human genome assembly. This previously unappreciated heterogeneity may underlie certain human phenotypic variation and susceptibility to disease and argues for a more dynamic human genome structure.


Journal of Experimental Medicine | 2002

Efficient Targeting of Protein Antigen to the Dendritic Cell Receptor DEC-205 in the Steady State Leads to Antigen Presentation on Major Histocompatibility Complex Class I Products and Peripheral CD8 T Cell Tolerance

Laura Bonifaz; David P. Bonnyay; Karsten Mahnke; Miguel Rivera; Michel C. Nussenzweig; Ralph M. Steinman

To identify endocytic receptors that allow dendritic cells (DCs) to capture and present antigens on major histocompatibility complex (MHC) class I products in vivo, we evaluated DEC-205, which is abundant on DCs in lymphoid tissues. Ovalbumin (OVA) protein, when chemically coupled to monoclonal αDEC-205 antibody, was presented by CD11c+ lymph node DCs, but not by CD11c− cells, to OVA-specific, CD4+ and CD8+ T cells. Receptor-mediated presentation was at least 400 times more efficient than unconjugated OVA and, for MHC class I, the DCs had to express transporter of antigenic peptides (TAP) transporters. When αDEC-205:OVA was injected subcutaneously, OVA protein was identified over a 4–48 h period in DCs, primarily in the lymph nodes draining the injection site. In vivo, the OVA protein was selectively presented by DCs to TCR transgenic CD8+ cells, again at least 400 times more effectively than soluble OVA and in a TAP-dependent fashion. Targeting of αDEC-205:OVA to DCs in the steady state initially induced 4–7 cycles of T cell division, but the T cells were then deleted and the mice became specifically unresponsive to rechallenge with OVA in complete Freunds adjuvant. In contrast, simultaneous delivery of a DC maturation stimulus via CD40, together with αDEC-205:OVA, induced strong immunity. The CD8+ T cells responding in the presence of agonistic αCD40 antibody produced large amounts of interleukin 2 and interferon γ, acquired cytolytic function in vivo, emigrated in large numbers to the lung, and responded vigorously to OVA rechallenge. Therefore, DEC-205 provides an efficient receptor-based mechanism for DCs to process proteins for MHC class I presentation in vivo, leading to tolerance in the steady state and immunity after DC maturation.


Science | 2011

Aberrant Overexpression of Satellite Repeats in Pancreatic and Other Epithelial Cancers

David T. Ting; Doron Lipson; Suchismita Paul; Brian W. Brannigan; Sara Akhavanfard; Erik J. Coffman; Gianmarco Contino; Vikram Deshpande; A. John Iafrate; Stan Letovsky; Miguel Rivera; Nabeel Bardeesy; Shyamala Maheswaran; Daniel A. Haber

Noncoding RNAs transcribed from DNA repeats in heterochromatin are expressed at surprisingly high levels in tumors. Satellite repeats in heterochromatin are transcribed into noncoding RNAs that have been linked to gene silencing and maintenance of chromosomal integrity. Using digital gene expression analysis, we showed that these transcripts are greatly overexpressed in mouse and human epithelial cancers. In 8 of 10 mouse pancreatic ductal adenocarcinomas (PDACs), pericentromeric satellites accounted for a mean 12% (range 1 to 50%) of all cellular transcripts, a mean 40-fold increase over that in normal tissue. In 15 of 15 human PDACs, alpha satellite transcripts were most abundant and HSATII transcripts were highly specific for cancer. Similar patterns were observed in cancers of the lung, kidney, ovary, colon, and prostate. Derepression of satellite transcripts correlated with overexpression of the long interspersed nuclear element 1 (LINE-1) retrotransposon and with aberrant expression of neuroendocrine-associated genes proximal to LINE-1 insertions. The overexpression of satellite transcripts in cancer may reflect global alterations in heterochromatin silencing and could potentially be useful as a biomarker for cancer detection.


Cancer Discovery | 2011

BIM expression in treatment-naïve cancers predicts responsiveness to kinase inhibitors

Anthony C. Faber; Ryan B. Corcoran; Hiromichi Ebi; Lecia V. Sequist; Belinda A. Waltman; Euiheon Chung; Joao Incio; Subba R. Digumarthy; Sarah F. Pollack; Youngchul Song; Alona Muzikansky; Eugene Lifshits; Sylvie Roberge; Erik J. Coffman; Cyril H. Benes; Henry Gomez; José Baselga; Carlos L. Arteaga; Miguel Rivera; Dora Dias-Santagata; Rakesh K. Jain; Jeffrey A. Engelman

Cancers with specific genetic mutations are susceptible to selective kinase inhibitors. However, there is a wide spectrum of benefit among cancers harboring the same sensitizing genetic mutations. Herein, we measured apoptotic rates among cell lines sharing the same driver oncogene following treatment with the corresponding kinase inhibitor. There was a wide range of kinase inhibitor-induced apoptosis despite comparable inhibition of the target and associated downstream signaling pathways. Surprisingly, pretreatment RNA levels of the BH3-only pro-apoptotic BIM strongly predicted the capacity of EGFR, HER2, and PI3K inhibitors to induce apoptosis in EGFR-mutant, HER2-amplified, and PIK3CA-mutant cancers, respectively, but BIM levels did not predict responsiveness to standard chemotherapies. Furthermore, BIM RNA levels in EGFR-mutant lung cancer specimens predicted response and duration of clinical benefit from EGFR inhibitors. These findings suggest assessment of BIM levels in treatment-naïve tumor biopsies may indicate the degree of benefit from single-agent kinase inhibitors in multiple oncogene-addiction paradigms.


Cell Reports | 2014

Single-Cell RNA Sequencing Identifies Extracellular Matrix Gene Expression by Pancreatic Circulating Tumor Cells

David T. Ting; Ben S. Wittner; Matteo Ligorio; Nicole Vincent Jordan; Ajay M. Shah; David T. Miyamoto; Nicola Aceto; Francesca Bersani; Brian W. Brannigan; Kristina Xega; Jordan C. Ciciliano; Huili Zhu; Olivia C. MacKenzie; Julie Trautwein; Kshitij S. Arora; Mohammad Shahid; Haley Ellis; Na Qu; Nabeel Bardeesy; Miguel Rivera; Vikram Deshpande; Cristina R. Ferrone; Ravi Kapur; Sridhar Ramaswamy; Toshi Shioda; Mehmet Toner; Shyamala Maheswaran; Daniel A. Haber

SUMMARY Circulating tumor cells (CTCs) are shed from primary tumors into the bloodstream, mediating the hematogenous spread of cancer to distant organs. To define their composition, we compared genome-wide expression profiles of CTCs with matched primary tumors in a mouse model of pancreatic cancer, isolating individual CTCs using epitope-independent microfluidic capture, followed by single-cell RNA sequencing. CTCs clustered separately from primary tumors and tumor-derived cell lines, showing low-proliferative signatures, enrichment for the stem-cell-associated gene Aldh1a2, biphenotypic expression of epithelial and mesenchymal markers, and expression of Igfbp5, a gene transcript enriched at the epithelial-stromal interface. Mouse as well as human pancreatic CTCs exhibit a very high expression of stromal-derived extracellular matrix (ECM) proteins, including SPARC, whose knockdown in cancer cells suppresses cell migration and invasiveness. The aberrant expression by CTCs of stromal ECM genes points to their contribution of microenvironmental signals for the spread of cancer to distant organs.


Cancer Research | 2007

Oncogenic activity of epidermal growth factor receptor kinase mutant alleles is enhanced by the T790M drug resistance mutation.

Nadia Godin-Heymann; Ianthe Bryant; Miguel Rivera; Lindsey Ulkus; Daphne W. Bell; David J. Riese; Jeffrey Settleman; Daniel A. Haber

Activating mutations in the epidermal growth factor receptor (EGFR) characterize a subset of non-small cell lung cancers (NSCLC) with extraordinary sensitivity to targeted tyrosine kinase inhibitors (TKI). A single secondary EGFR mutation, T790M, arising in cis with the primary activating mutation, confers acquired resistance to these drugs. However, the T790M mutation is also detected in the absence of drug selection, suggesting that it may provide a growth advantage. We show here that although T790M alone has only a modest effect on EGFR function, when combined with the characteristic activating mutations L858R or del746-750, it results in a dramatic enhancement of EGFR activity. The double mutants show potent ligand-independent receptor autophosphorylation associated with altered cellular phenotypes, soft agar colony formation, and tumorigenesis in nude mice. The significant gain-of-function properties of these double mutants may explain their initial presence before drug selection and their rapid selection as the single drug resistance mutation during therapy with gefitinib/erlotinib, and suggests that they may contribute to the adverse clinical course of TKI-resistant NSCLC.


Nature | 2016

Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma

Itay Tirosh; Andrew S. Venteicher; Christine Hebert; Leah E. Escalante; Anoop P. Patel; Keren Yizhak; Jonathan M. Fisher; Christopher Rodman; Christopher Mount; Mariella G. Filbin; Cyril Neftel; Niyati Desai; Jackson Nyman; Benjamin Izar; Christina C. Luo; Joshua M. Francis; Aanand A. Patel; Maristela L. Onozato; Nicolo Riggi; Kenneth J. Livak; Dave Gennert; Rahul Satija; Brian V. Nahed; William T. Curry; Robert L. Martuza; Ravindra Mylvaganam; A. John Iafrate; Matthew P. Frosch; Todd R. Golub; Miguel Rivera

Although human tumours are shaped by the genetic evolution of cancer cells, evidence also suggests that they display hierarchies related to developmental pathways and epigenetic programs in which cancer stem cells (CSCs) can drive tumour growth and give rise to differentiated progeny. Yet, unbiased evidence for CSCs in solid human malignancies remains elusive. Here we profile 4,347 single cells from six IDH1 or IDH2 mutant human oligodendrogliomas by RNA sequencing (RNA-seq) and reconstruct their developmental programs from genome-wide expression signatures. We infer that most cancer cells are differentiated along two specialized glial programs, whereas a rare subpopulation of cells is undifferentiated and associated with a neural stem cell expression program. Cells with expression signatures for proliferation are highly enriched in this rare subpopulation, consistent with a model in which CSCs are primarily responsible for fuelling the growth of oligodendroglioma in humans. Analysis of copy number variation (CNV) shows that distinct CNV sub-clones within tumours display similar cellular hierarchies, suggesting that the architecture of oligodendroglioma is primarily dictated by developmental programs. Subclonal point mutation analysis supports a similar model, although a full phylogenetic tree would be required to definitively determine the effect of genetic evolution on the inferred hierarchies. Our single-cell analyses provide insight into the cellular architecture of oligodendrogliomas at single-cell resolution and support the cancer stem cell model, with substantial implications for disease management.


Cancer Discovery | 2014

The Genomic Landscape of Pediatric Ewing Sarcoma

Brian D. Crompton; Chip Stewart; Amaro Taylor-Weiner; Gabriela Alexe; Kurek Kc; Monica L. Calicchio; Adam Kiezun; Scott L. Carter; Sachet A. Shukla; Swapnil Mehta; Aaron R. Thorner; de Torres C; Cinzia Lavarino; Mariona Suñol; Aaron McKenna; Andrey Sivachenko; Kristian Cibulskis; Michael S. Lawrence; Petar Stojanov; Mara Rosenberg; Lauren Ambrogio; Daniel Auclair; Sara Seepo; Brendan Blumenstiel; Matthew DeFelice; Ivan Imaz-Rosshandler; Miguel Rivera; Carlos Rodriguez-Galindo; Fleming; Todd R. Golub

UNLABELLED Pediatric Ewing sarcoma is characterized by the expression of chimeric fusions of EWS and ETS family transcription factors, representing a paradigm for studying cancers driven by transcription factor rearrangements. In this study, we describe the somatic landscape of pediatric Ewing sarcoma. These tumors are among the most genetically normal cancers characterized to date, with only EWS-ETS rearrangements identified in the majority of tumors. STAG2 loss, however, is present in more than 15% of Ewing sarcoma tumors; occurs by point mutation, rearrangement, and likely nongenetic mechanisms; and is associated with disease dissemination. Perhaps the most striking finding is the paucity of mutations in immediately targetable signal transduction pathways, highlighting the need for new therapeutic approaches to target EWS-ETS fusions in this disease. SIGNIFICANCE We performed next-generation sequencing of Ewing sarcoma, a pediatric cancer involving bone, characterized by expression of EWS-ETS fusions. We found remarkably few mutations. However, we discovered that loss of STAG2 expression occurs in 15% of tumors and is associated with metastatic disease, suggesting a potential genetic vulnerability in Ewing sarcoma.


Cancer Cell | 2014

EWS-FLI1 Utilizes Divergent Chromatin Remodeling Mechanisms to Directly Activate or Repress Enhancer Elements in Ewing Sarcoma

Nicolo Riggi; Birgit Knoechel; Shawn M. Gillespie; Esther Rheinbay; Gaylor Boulay; Mario L. Suvà; Nikki Rossetti; Wannaporn E. Boonseng; Ozgur Oksuz; Edward B. Cook; Aurélie Formey; Anoop P. Patel; Melissa Gymrek; Vishal Thapar; Vikram Deshpande; David T. Ting; Francis J. Hornicek; G. Petur Nielsen; Ivan Stamenkovic; Martin J. Aryee; Bradley E. Bernstein; Miguel Rivera

The aberrant transcription factor EWS-FLI1 drives Ewing sarcoma, but its molecular function is not completely understood. We find that EWS-FLI1 reprograms gene regulatory circuits in Ewing sarcoma by directly inducing or repressing enhancers. At GGAA repeat elements, which lack evolutionary conservation and regulatory potential in other cell types, EWS-FLI1 multimers induce chromatin opening and create de novo enhancers that physically interact with target promoters. Conversely, EWS-FLI1 inactivates conserved enhancers containing canonical ETS motifs by displacing wild-type ETS transcription factors. These divergent chromatin-remodeling patterns repress tumor suppressors and mesenchymal lineage regulators while activating oncogenes and potential therapeutic targets, such as the kinase VRK1. Our findings demonstrate how EWS-FLI1 establishes an oncogenic regulatory program governing both tumor survival and differentiation.


Science | 2017

Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq.

Andrew S. Venteicher; Itay Tirosh; Christine Hebert; Keren Yizhak; Cyril Neftel; Mariella G. Filbin; Volker Hovestadt; Leah E. Escalante; McKenzie L. Shaw; Christopher Rodman; Shawn M. Gillespie; Danielle Dionne; Christina C. Luo; Hiranmayi Ravichandran; Ravindra Mylvaganam; Christopher Mount; Maristela L. Onozato; Brian V. Nahed; Hiroaki Wakimoto; William T. Curry; A. John Iafrate; Miguel Rivera; Matthew P. Frosch; Todd R. Golub; Priscilla K. Brastianos; Gad Getz; Anoop P. Patel; Michelle Monje; Daniel P. Cahill; Orit Rozenblatt-Rosen

Single-cell RNA sequencing identifies a common origin for specific types of human glioma brain tumors. Effects of the tumor microenvironment Glioma brain tumors that carry mutant copies of the IDH gene can be subdivided into two major classes. However, the development of and differences between these two classes are not well characterized. Venteicher et al. coupled bulk sequencing and publicly available data with single-cell RNA sequencing data on glioma patient tissue samples. They identified a common lineage program that is shared between glioma subtypes. This suggests that the observed differences between the two glioma classes originate from lineage-specific genetic changes and the tumor microenvironment. Science, this issue p. eaai8478 INTRODUCTION Tumor fitness, evolution, and resistance to therapy are governed by selection of malignant cells with specific genotypes, by expression programs related to cellular phenotypes, and by influences of the tumor microenvironment (TME). Although bulk tumor analysis can interrogate the genetic state of tumor cells with high precision, bulk expression profiles average the diverse cells within each tumor, thereby masking critical differences and providing limited insight into cancer cell programs and TME influences. Single-cell RNA sequencing (scRNA-seq) can help to address those challenges but incurs financial and logistic considerations, including the time required to accrue large cohorts of fresh tumor specimen for single-cell analysis. RATIONALE We reasoned that scRNA-seq of a limited number of representative tumors could be combined with bulk data from large cohorts to decipher differences between tumor subclasses. In this approach, bulk samples collected for large cohorts, such as from The Cancer Genome Atlas (TCGA), are first used to define the combined effects of differences in cancer cell genotypes, phenotypes, and the composition of the TME. Single-cell analysis of a limited set of representative tumors is then used to distinguish those effects. We applied this approach to understand the differences between two types of isocitrate dehydrogenase (IDH)–mutant gliomas: astrocytoma (IDH-A) and oligodendroglioma (IDH-O). IDH-A and IDH-O are distinguished by co-occurring signature genetic events and by histopathology and are thought to recapitulate distinct glial lineages. By combining 9879 scRNA-seq profiles from 10 IDH-A tumors, 4347 scRNA-seq profiles from six IDH-O tumors, and 165 TCGA bulk RNA profiles, we could decipher differences between these two tumor types at single-cell resolution. RESULTS We find that differences in bulk expression profiles between IDH-A and IDH-O are primarily explained by the impact of signature genetic events and TME composition, but not by distinct expression programs of glial lineages in the malignant cells. We infer that both IDH-A and IDH-O share the same developmental hierarchy, consisting in each case of three subpopulations of malignant cells: nonproliferating cells differentiated along the astrocytic and oligodendrocytic lineages, and proliferative undifferentiated cells that resemble neural stem/progenitor cells. By analyzing tumors of different clinical grades, we observe that higher-grade tumors present enhanced proliferation, larger pools of undifferentiated glioma cells, and an increase in macrophage over microglia programs in the TME. CONCLUSION Our approach provides a general framework to decipher differences between classes of human tumors by decoupling cancer cell genotypes, phenotypes, and the composition of the TME. The shared glial lineages and developmental hierarchies observed in IDH-A and IDH-O suggest a common progenitor for all IDH-mutant gliomas, shedding light on a long-standing debate in gliomagenesis. In contrast to the similarity in glial lineages, IDH-A and IDH-O differ significantly in their TME, and in particular in the abundance of microglia/macrophage cells. Microglia and macrophages also differ between IDH-A tumors of different grades. Our study redefines the cellular composition of human IDH-mutant gliomas, with important implications for disease management. Single-cell RNA-seq of IDH-mutant gliomas reveals tumor architecture. (Top) Human samples were dissociated and analyzed by scRNA-seq. (Bottom) IDH-O and IDH-A differ in genetics and TME but are both primarily composed of three main types of malignant cells: cycling stem-like cells and noncycling astrocyte-like and oligodendrocyte-like cells. Tumor progression is associated with increased proliferation, decreased differentiation, and increase in macrophages over microglia in the TME. Tumor subclasses differ according to the genotypes and phenotypes of malignant cells as well as the composition of the tumor microenvironment (TME). We dissected these influences in isocitrate dehydrogenase (IDH)–mutant gliomas by combining 14,226 single-cell RNA sequencing (RNA-seq) profiles from 16 patient samples with bulk RNA-seq profiles from 165 patient samples. Differences in bulk profiles between IDH-mutant astrocytoma and oligodendroglioma can be primarily explained by distinct TME and signature genetic events, whereas both tumor types share similar developmental hierarchies and lineages of glial differentiation. As tumor grade increases, we find enhanced proliferation of malignant cells, larger pools of undifferentiated glioma cells, and an increase in macrophage over microglia expression programs in TME. Our work provides a unifying model for IDH-mutant gliomas and a general framework for dissecting the differences among human tumor subclasses.

Collaboration


Dive into the Miguel Rivera's collaboration.

Top Co-Authors

Avatar

M. Portolés

Instituto Politécnico Nacional

View shared research outputs
Top Co-Authors

Avatar

E. Roselló-Lletí

Instituto Politécnico Nacional

View shared research outputs
Top Co-Authors

Avatar

José Ramón González-Juanatey

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

Luis Martínez-Dolz

Instituto Politécnico Nacional

View shared research outputs
Top Co-Authors

Avatar

R. Cortés

Instituto Politécnico Nacional

View shared research outputs
Top Co-Authors

Avatar

Antonio Salvador

Instituto Politécnico Nacional

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vicente Miró

Instituto Politécnico Nacional

View shared research outputs
Researchain Logo
Decentralizing Knowledge