Network


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

Hotspot


Dive into the research topics where Joshua M. Francis is active.

Publication


Featured researches published by Joshua M. Francis.


Nature | 2012

Sequence analysis of mutations and translocations across breast cancer subtypes

Shantanu Banerji; Kristian Cibulskis; Claudia Rangel-Escareño; Kristin K. Brown; Scott L. Carter; Abbie M. Frederick; Michael S. Lawrence; Andrey Sivachenko; Carrie Sougnez; Lihua Zou; Maria L. Cortes; Juan Carlos Fernández-López; Shouyong Peng; Kristin Ardlie; Daniel Auclair; Verónica Bautista-Piña; Fujiko Duke; Joshua M. Francis; Joonil Jung; Antonio Maffuz-Aziz; Robert C. Onofrio; Melissa Parkin; Nam H. Pho; Valeria Quintanar-Jurado; Alex H. Ramos; Rosa Rebollar-Vega; Sergio Rodriguez-Cuevas; Sandra Romero-Cordoba; Steven E. Schumacher; Nicolas Stransky

Breast carcinoma is the leading cause of cancer-related mortality in women worldwide, with an estimated 1.38 million new cases and 458,000 deaths in 2008 alone. This malignancy represents a heterogeneous group of tumours with characteristic molecular features, prognosis and responses to available therapy. Recurrent somatic alterations in breast cancer have been described, including mutations and copy number alterations, notably ERBB2 amplifications, the first successful therapy target defined by a genomic aberration. Previous DNA sequencing studies of breast cancer genomes have revealed additional candidate mutations and gene rearrangements. Here we report the whole-exome sequences of DNA from 103 human breast cancers of diverse subtypes from patients in Mexico and Vietnam compared to matched-normal DNA, together with whole-genome sequences of 22 breast cancer/normal pairs. Beyond confirming recurrent somatic mutations in PIK3CA, TP53, AKT1, GATA3 and MAP3K1, we discovered recurrent mutations in the CBFB transcription factor gene and deletions of its partner RUNX1. Furthermore, we have identified a recurrent MAGI3–AKT3 fusion enriched in triple-negative breast cancer lacking oestrogen and progesterone receptors and ERBB2 expression. The MAGI3–AKT3 fusion leads to constitutive activation of AKT kinase, which is abolished by treatment with an ATP-competitive AKT small-molecule inhibitor.


Nature | 2015

Chromothripsis from DNA damage in micronuclei.

Cheng-Zhong Zhang; Alexander Spektor; Hauke Cornils; Joshua M. Francis; Emily Jackson; Shiwei Liu; Matthew Meyerson; David Pellman

Genome sequencing has uncovered a new mutational phenomenon in cancer and congenital disorders called chromothripsis. Chromothripsis is characterized by extensive genomic rearrangements and an oscillating pattern of DNA copy number levels, all curiously restricted to one or a few chromosomes. The mechanism for chromothripsis is unknown, but we previously proposed that it could occur through the physical isolation of chromosomes in aberrant nuclear structures called micronuclei. Here, using a combination of live cell imaging and single-cell genome sequencing, we demonstrate that micronucleus formation can indeed generate a spectrum of genomic rearrangements, some of which recapitulate all known features of chromothripsis. These events are restricted to the mis-segregated chromosome and occur within one cell division. We demonstrate that the mechanism for chromothripsis can involve the fragmentation and subsequent reassembly of a single chromatid from a micronucleus. Collectively, these experiments establish a new mutational process of which chromothripsis is one extreme outcome.


Nature Biotechnology | 2014

Whole exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer

Jens Lohr; Viktor A. Adalsteinsson; Kristian Cibulskis; Atish D. Choudhury; Mara Rosenberg; Peter Cruz-Gordillo; Joshua M. Francis; Cheng-Zhong Zhang; Alex K. Shalek; Rahul Satija; John J. Trombetta; Diana Lu; Naren Tallapragada; Narmin Tahirova; Sora Kim; Brendan Blumenstiel; Carrie Sougnez; Alarice Lowe; Bang Wong; Daniel Auclair; Eliezer M. Van Allen; Mari Nakabayashi; Rosina T. Lis; Gwo-Shu Mary Lee; Tiantian Li; Matthew S. Chabot; Amy Ly; Mary-Ellen Taplin; Thomas E. Clancy; Massimo Loda

Comprehensive analyses of cancer genomes promise to inform prognoses and precise cancer treatments. A major barrier, however, is inaccessibility of metastatic tissue. A potential solution is to characterize circulating tumor cells (CTCs), but this requires overcoming the challenges of isolating rare cells and sequencing low-input material. Here we report an integrated process to isolate, qualify and sequence whole exomes of CTCs with high fidelity using a census-based sequencing strategy. Power calculations suggest that mapping of >99.995% of the standard exome is possible in CTCs. We validated our process in two patients with prostate cancer, including one for whom we sequenced CTCs, a lymph node metastasis and nine cores of the primary tumor. Fifty-one of 73 CTC mutations (70%) were present in matched tissue. Moreover, we identified 10 early trunk and 56 metastatic trunk mutations in the non-CTC tumor samples and found 90% and 73% of these mutations, respectively, in CTC exomes. This study establishes a foundation for CTC genomics in the clinic.


Nature Genetics | 2013

Somatic mutation of CDKN1B in small intestine neuroendocrine tumors

Joshua M. Francis; Adam Kiezun; Alex H. Ramos; Stefano Serra; Chandra Sekhar Pedamallu; Zhi Rong Qian; Michaela S. Banck; Rahul Kanwar; Amit A. Kulkarni; Anna Karpathakis; Veronica E. Manzo; Tanupriya Contractor; Juliet Philips; Elizabeth Nickerson; Nam H. Pho; Susanne M. Hooshmand; Lauren K. Brais; Michael S. Lawrence; Trevor J. Pugh; Aaron McKenna; Andrey Sivachenko; Kristian Cibulskis; Scott L. Carter; Akinyemi I. Ojesina; Samuel S. Freeman; Robert T. Jones; Douglas Voet; Gordon Saksena; Daniel Auclair; Robert C. Onofrio

The diagnosed incidence of small intestine neuroendocrine tumors (SI-NETs) is increasing, and the underlying genomic mechanisms have not yet been defined. Using exome- and genome-sequence analysis of SI-NETs, we identified recurrent somatic mutations and deletions in CDKN1B, the cyclin-dependent kinase inhibitor gene, which encodes p27. We observed frameshift mutations of CDKN1B in 14 of 180 SI-NETs, and we detected hemizygous deletions encompassing CDKN1B in 7 out of 50 SI-NETs, nominating p27 as a tumor suppressor and implicating cell cycle dysregulation in the etiology of SI-NETs.


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

EGFR variant heterogeneity in glioblastoma resolved through single-nucleus sequencing

Joshua M. Francis; Cheng-Zhong Zhang; Cecile L. Maire; Joonil Jung; Veronica E. Manzo; Viktor A. Adalsteinsson; Heather Homer; Samer Haidar; Brendan Blumenstiel; Chandra Sekhar Pedamallu; Azra H. Ligon; John C Love; Matthew Meyerson; Keith L. Ligon

UNLABELLED Glioblastomas (GBM) with EGFR amplification represent approximately 50% of newly diagnosed cases, and recent studies have revealed frequent coexistence of multiple EGFR aberrations within the same tumor, which has implications for mutation cooperation and treatment resistance. However, bulk tumor sequencing studies cannot resolve the patterns of how the multiple EGFR aberrations coexist with other mutations within single tumor cells. Here, we applied a population-based single-cell whole-genome sequencing methodology to characterize genomic heterogeneity in EGFR-amplified glioblastomas. Our analysis effectively identified clonal events, including a novel translocation of a super enhancer to the TERT promoter, as well as subclonal LOH and multiple EGFR mutational variants within tumors. Correlating the EGFR mutations onto the cellular hierarchy revealed that EGFR truncation variants (EGFRvII and EGFR carboxyl-terminal deletions) identified in the bulk tumor segregate into nonoverlapping subclonal populations. In vitro and in vivo functional studies show that EGFRvII is oncogenic and sensitive to EGFR inhibitors currently in clinical trials. Thus, the association between diverse activating mutations in EGFR and other subclonal mutations within a single tumor supports an intrinsic mechanism for proliferative and clonal diversification with broad implications in resistance to treatment. SIGNIFICANCE We developed a novel single-cell sequencing methodology capable of identifying unique, nonoverlapping subclonal alterations from archived frozen clinical specimens. Using GBM as an example, we validated our method to successfully define tumor cell subpopulations containing distinct genetic and treatment resistance profiles and potentially mutually cooperative combinations of alterations in EGFR and other genes.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Loss of the retinoblastoma binding protein 2 (RBP2) histone demethylase suppresses tumorigenesis in mice lacking Rb1 or Men1

Wenchu Lin; Jian Cao; Jiayun Liu; Michael L. Beshiri; Yuko Fujiwara; Joshua M. Francis; Andrew D. Cherniack; Christoph Geisen; Lauren P. Blair; Mike R. Zou; Xiaohua Shen; Dan Kawamori; Zongzhi Liu; Chiara Grisanzio; Hideo Watanabe; Yoji Andrew Minamishima; Qing Zhang; Rohit N. Kulkarni; Sabina Signoretti; Scott J. Rodig; Roderick T. Bronson; Stuart H. Orkin; David Tuck; Elizaveta V. Benevolenskaya; Matthew Meyerson; William G. Kaelin; Qin Yan

Aberrations in epigenetic processes, such as histone methylation, can cause cancer. Retinoblastoma binding protein 2 (RBP2; also called JARID1A or KDM5A) can demethylate tri- and dimethylated lysine 4 in histone H3, which are epigenetic marks for transcriptionally active chromatin, whereas the multiple endocrine neoplasia type 1 (MEN1) tumor suppressor promotes H3K4 methylation. Previous studies suggested that inhibition of RBP2 contributed to tumor suppression by the retinoblastoma protein (pRB). Here, we show that genetic ablation of Rbp2 decreases tumor formation and prolongs survival in Rb1+/− mice and Men1-defective mice. These studies link RBP2 histone demethylase activity to tumorigenesis and nominate RBP2 as a potential target for cancer therapy.


Nature Genetics | 2016

Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers

Xiaoyang Zhang; Peter S. Choi; Joshua M. Francis; Marcin Imielinski; Hideo Watanabe; Andrew D. Cherniack; Matthew Meyerson

Whole-genome analysis approaches are identifying recurrent cancer-associated somatic alterations in noncoding DNA regions. We combined somatic copy number analysis of 12 tumor types with tissue-specific epigenetic profiling to identify significant regions of focal amplification harboring super-enhancers. Copy number gains of noncoding regions harboring super-enhancers near KLF5, USP12, PARD6B and MYC are associated with overexpression of these cancer-related genes. We show that two distinct focal amplifications of super-enhancers 3′ to MYC in lung adenocarcinoma (MYC-LASE) and endometrial carcinoma (MYC-ECSE) are physically associated with the MYC promoter and correlate with MYC overexpression. CRISPR/Cas9-mediated repression or deletion of a constituent enhancer within the MYC-LASE region led to significant reductions in the expression of MYC and its target genes and to the impairment of anchorage-independent and clonogenic growth, consistent with an oncogenic function. Our results suggest that genomic amplification of super-enhancers represents a common mechanism to activate cancer driver genes in multiple cancer types.


Proceedings of the National Academy of Sciences of the United States of America | 2009

The tumor suppressor Cdc73 functionally associates with CPSF and CstF 3′ mRNA processing factors

Orit Rozenblatt-Rosen; Takashi Nagaike; Joshua M. Francis; Syuzo Kaneko; Karen A. Glatt; Christina M. Hughes; Thomas LaFramboise; James L. Manley; Matthew Meyerson

The CDC73 tumor suppressor gene is mutationally inactivated in hereditary and sporadic parathyroid tumors. Its product, the Cdc73 protein, is a component of the RNA polymerase II and chromatin-associated human Paf1 complex (Paf1C). Here, we show that Cdc73 physically associates with the cleavage and polyadenylation specificity factor (CPSF) and cleavage stimulation factor (CstF) complexes that are required for the maturation of mRNA 3′ ends in the cell nucleus. Immunodepletion experiments indicate that the Cdc73–CPSF–CstF complex is necessary for 3′ mRNA processing in vitro. Microarray analysis of CDC73 siRNA-treated cells revealed INTS6, a gene encoding a subunit of the Integrator complex, as an in vivo Cdc73 target. Cdc73 depletion by siRNA resulted in decreased INTS6 mRNA abundance, and decreased association of CPSF and CstF subunits with the INTS6 locus. Our results suggest that Cdc73 facilitates association of 3′ mRNA processing factors with actively-transcribed chromatin and support the importance of links between tumor suppression and mRNA maturation.


Journal of Clinical Investigation | 2014

SOX2 and p63 colocalize at genetic loci in squamous cell carcinomas

Hideo Watanabe; Qiuping Ma; Shouyong Peng; Guillaume Adelmant; Danielle Swain; Wenyu Song; Cameron Fox; Joshua M. Francis; Chandra Sekhar Pedamallu; David S. DeLuca; Angela N. Brooks; Su Wang; Jianwen Que; Anil K. Rustgi; Kwok-Kin Wong; Keith L. Ligon; X. Shirley Liu; Jarrod A. Marto; Matthew Meyerson; Adam J. Bass

The transcription factor SOX2 is an essential regulator of pluripotent stem cells and promotes development and maintenance of squamous epithelia. We previously reported that SOX2 is an oncogene and subject to highly recurrent genomic amplification in squamous cell carcinomas (SCCs). Here, we have further characterized the function of SOX2 in SCC. Using ChIP-seq analysis, we compared SOX2-regulated gene profiles in multiple SCC cell lines to ES cell profiles and determined that SOX2 binds to distinct genomic loci in SCCs. In SCCs, SOX2 preferentially interacts with the transcription factor p63, as opposed to the transcription factor OCT4, which is the preferred SOX2 binding partner in ES cells. SOX2 and p63 exhibited overlapping genomic occupancy at a large number of loci in SCCs; however, coordinate binding of SOX2 and p63 was absent in ES cells. We further demonstrated that SOX2 and p63 jointly regulate gene expression, including the oncogene ETV4, which was essential for SOX2-amplified SCC cell survival. Together, these findings demonstrate that the action of SOX2 in SCC differs substantially from its role in pluripotency. The identification of the SCC-associated interaction between SOX2 and p63 will enable deeper characterization the downstream targets of this interaction in SCC and normal squamous epithelial physiology.

Collaboration


Dive into the Joshua M. Francis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge