Network


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

Hotspot


Dive into the research topics where Jennifer Silvester is active.

Publication


Featured researches published by Jennifer Silvester.


Genes & Development | 2011

Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress

Kathrin Zaugg; Yi Yao; Patrick T. Reilly; Karuppiah Kannan; Reza Kiarash; Jacqueline M. Mason; Ping Huang; Suzanne K. Sawyer; Benjamin J. Fuerth; Brandon Faubert; Tuula Kalliomaki; Andrew Elia; Xunyi Luo; Vincent Nadeem; David Bungard; Sireesha Yalavarthi; Joseph D. Growney; Andrew Wakeham; Yasmin Moolani; Jennifer Silvester; Annick You Ten; Walbert J. Bakker; Katsuya Tsuchihara; Shelley L. Berger; Richard P. Hill; Russell G. Jones; Ming Tsao; Murray Robinson; Craig B. Thompson; Guohua Pan

Tumor cells gain a survival/growth advantage by adapting their metabolism to respond to environmental stress, a process known as metabolic transformation. The best-known aspect of metabolic transformation is the Warburg effect, whereby cancer cells up-regulate glycolysis under aerobic conditions. However, other mechanisms mediating metabolic transformation remain undefined. Here we report that carnitine palmitoyltransferase 1C (CPT1C), a brain-specific metabolic enzyme, may participate in metabolic transformation. CPT1C expression correlates inversely with mammalian target of rapamycin (mTOR) pathway activation, contributes to rapamycin resistance in murine primary tumors, and is frequently up-regulated in human lung tumors. Tumor cells constitutively expressing CPT1C show increased fatty acid (FA) oxidation, ATP production, and resistance to glucose deprivation or hypoxia. Conversely, cancer cells lacking CPT1C produce less ATP and are more sensitive to metabolic stress. CPT1C depletion via siRNA suppresses xenograft tumor growth and metformin responsiveness in vivo. CPT1C can be induced by hypoxia or glucose deprivation and is regulated by AMPKα. Cpt1c-deficient murine embryonic stem (ES) cells show sensitivity to hypoxia and glucose deprivation and altered FA homeostasis. Our results indicate that cells can use a novel mechanism involving CPT1C and FA metabolism to protect against metabolic stress. CPT1C may thus be a new therapeutic target for the treatment of hypoxic tumors.


Oncogene | 2012

The interaction between caveolin-1 and Rho-GTPases promotes metastasis by controlling the expression of alpha5-integrin and the activation of Src, Ras and Erk.

Enrico Arpaia; H Blaser; M Quintela-Fandino; Gordon S. Duncan; H S Leong; A Ablack; S C Nambiar; Evan F. Lind; Jennifer Silvester; C K Fleming; Alessandro Rufini; M W Tusche; Anne Brüstle; Pamela S. Ohashi; J D Lewis; Tak W. Mak

Proteins containing a caveolin-binding domain (CBD), such as the Rho-GTPases, can interact with caveolin-1 (Cav1) through its caveolin scaffold domain. Rho-GTPases are important regulators of p130Cas, which is crucial for both normal cell migration and Src kinase-mediated metastasis of cancer cells. However, although Rho-GTPases (particularly RhoC) and Cav1 have been linked to cancer progression and metastasis, the underlying molecular mechanisms are largely unknown. To investigate the function of Cav1–Rho-GTPase interaction in metastasis, we disrupted Cav1–Rho-GTPase binding in melanoma and mammary epithelial tumor cells by overexpressing CBD, and examined the loss-of-function of RhoC in metastatic cancer cells. Cancer cells overexpressing CBD or lacking RhoC had reduced p130Cas phosphorylation and Rac1 activation, resulting in an inhibition of migration and invasion in vitro. The activity of Src and the activation of its downstream targets FAK, Pyk2, Ras and extracellular signal-regulated kinase (Erk)1/2 were also impaired. A reduction in α5-integrin expression, which is required for binding to fibronectin and thus cell migration and survival, was observed in CBD-expressing cells and cells lacking RhoC. As a result of these defects, CBD-expressing melanoma cells had a reduced ability to metastasize in recipient mice, and impaired extravasation and survival in secondary sites in chicken embryos. Our data indicate that interaction between Cav1 and Rho-GTPases (most likely RhoC but not RhoA) promotes metastasis by stimulating α5-integrin expression and regulating the Src-dependent activation of p130Cas/Rac1, FAK/Pyk2 and Ras/Erk1/2 signaling cascades.


Genes & Development | 2013

Mule/Huwe1/Arf-BP1 suppresses Ras-driven tumorigenesis by preventing c-Myc/Miz1-mediated down-regulation of p21 and p15

Satoshi Inoue; Zhenyue Hao; Andrew J. Elia; David W. Cescon; Lily Zhou; Jennifer Silvester; Bryan E. Snow; Isaac S. Harris; Masato Sasaki; Wanda Y. Li; Momoe Itsumi; Kazuo Yamamoto; Takeshi Ueda; Carmen Dominguez-Brauer; Chiara Gorrini; Iok In Christine Chio; Jillian Haight; Annick You-Ten; Susan McCracken; Andrew Wakeham; Danny Ghazarian; Linda Penn; Gerry Melino; Tak W. Mak

Tumorigenesis results from dysregulation of oncogenes and tumor suppressors that influence cellular proliferation, differentiation, apoptosis, and/or senescence. Many gene products involved in these processes are substrates of the E3 ubiquitin ligase Mule/Huwe1/Arf-BP1 (Mule), but whether Mule acts as an oncogene or tumor suppressor in vivo remains controversial. We generated K14Cre;Mule(flox/flox(y)) (Mule kKO) mice and subjected them to DMBA/PMA-induced skin carcinogenesis, which depends on oncogenic Ras signaling. Mule deficiency resulted in increased penetrance, number, and severity of skin tumors, which could be reversed by concomitant genetic knockout of c-Myc but not by knockout of p53 or p19Arf. Notably, in the absence of Mule, c-Myc/Miz1 transcriptional complexes accumulated, and levels of p21CDKN1A (p21) and p15INK4B (p15) were down-regulated. In vitro, Mule-deficient primary keratinocytes exhibited increased proliferation that could be reversed by Miz1 knockdown. Transfer of Mule-deficient transformed cells to nude mice resulted in enhanced tumor growth that again could be abrogated by Miz1 knockdown. Our data demonstrate in vivo that Mule suppresses Ras-mediated tumorigenesis by preventing an accumulation of c-Myc/Miz1 complexes that mediates p21 and p15 down-regulation.


Proceedings of the National Academy of Sciences of the United States of America | 2014

TAp73 is required for spermatogenesis and the maintenance of male fertility

Satoshi Inoue; Richard Tomasini; Alessandro Rufini; Andrew J. Elia; Massimiliano Agostini; Ivano Amelio; Dave Cescon; David Dinsdale; Lily Zhou; Isaac S. Harris; Sophie Lac; Jennifer Silvester; Wanda Y. Li; Masato Sasaki; Jillian Haight; Anne Brüstle; Andrew Wakeham; Colin McKerlie; Andrea Jurisicova; Gerry Melino; Tak W. Mak

Significance Defects in spermatogenesis, many of which are unexplained, underlie the infertility problems of ∼20% of couples. Although specific roles for the p53 family members in female fertility have been described, their involvement in spermatogenesis is largely unexpected. Using gene-targeted mice, we have demonstrated that deficiency of TAp73, but not p53 or ∆Np73, leads to male infertility caused by severely impaired germ cell differentiation and maturation to viable sperms in the testes. Importantly, our work has established that TAp73, but not p53, regulates many genes involved in spermatogenesis. Thus, our results provide previously unidentified in vivo evidence that TAp73 is a “guardian” of male germ cells and may point toward a novel avenue for the diagnosis and management of male infertility. The generation of viable sperm proceeds through a series of coordinated steps, including germ cell self-renewal, meiotic recombination, and terminal differentiation into functional spermatozoa. The p53 family of transcription factors, including p53, p63, and p73, are critical for many physiological processes, including female fertility, but little is known about their functions in spermatogenesis. Here, we report that deficiency of the TAp73 isoform, but not p53 or ΔNp73, results in male infertility because of severe impairment of spermatogenesis. Mice lacking TAp73 exhibited increased DNA damage and cell death in spermatogonia, disorganized apical ectoplasmic specialization, malformed spermatids, and marked hyperspermia. We demonstrated that TAp73 regulates the mRNA levels of crucial genes involved in germ stem/progenitor cells (CDKN2B), spermatid maturation/spermiogenesis (metalloproteinase and serine proteinase inhibitors), and steroidogenesis (CYP21A2 and progesterone receptor). These alterations of testicular histology and gene expression patterns were specific to TAp73 null mice and not features of mice lacking p53. Our work provides previously unidentified in vivo evidence that TAp73 has a unique role in spermatogenesis that ensures the maintenance of mitotic cells and normal spermiogenesis. These results may have implications for the diagnosis and management of human male infertility.


Journal of Experimental Medicine | 2012

The E3 ubiquitin ligase Mule acts through the ATM–p53 axis to maintain B lymphocyte homeostasis

Zhenyue Hao; Gordon S. Duncan; Yu-Wen Su; Wanda Y. Li; Jennifer Silvester; Claire Hong; Han You; Dirk Brenner; Chiara Gorrini; Jillian Haight; Andrew Wakeham; Annick You-Ten; Susan McCracken; Andrew Elia; Qinxi Li; Jacqui Detmar; Andrea Jurisicova; Elias Hobeika; Michael Reth; Yi Sheng; Philipp A. Lang; Pamela S. Ohashi; Qing Zhong; Xiaodong Wang; Tak W. Mak

Genetic manipulation reveals that Mule is vital for B cell development, proliferation, and homeostasis as a result of its ability to regulate p53 and ATM.


Nature Immunology | 2013

ARIH2 is essential for embryogenesis, and its hematopoietic deficiency causes lethal activation of the immune system

Amy E Lin; Gregor Ebert; Yongkai Ow; Simon Preston; Jesse G. Toe; James Cooney; Hamish W. Scott; Masato Sasaki; Samuel D. Saibil; Dilan Dissanayake; Raymond H. Kim; Andrew Wakeham; Annick You-Ten; Arda Shahinian; Gordon S. Duncan; Jennifer Silvester; Pamela S. Ohashi; Tak W. Mak; Marc Pellegrini

The E3 ligase ARIH2 has an unusual structure and mechanism of elongating ubiquitin chains. To understand its physiological role, we generated gene-targeted mice deficient in ARIH2. ARIH2 deficiency resulted in the embryonic death of C57BL/6 mice. On a mixed genetic background, the lethality was attenuated, with some mice surviving beyond weaning and then succumbing to an aggressive multiorgan inflammatory response. We found that in dendritic cells (DCs), ARIH2 caused degradation of the inhibitor IκBβ in the nucleus, which abrogated its ability to sequester, protect and transcriptionally coactivate the transcription factor subunit p65 in the nucleus. Loss of ARIH2 caused dysregulated activation of the transcription factor NF-κB in DCs, which led to lethal activation of the immune system in ARIH2-sufficent mice reconstituted with ARIH2-deficient hematopoietic stem cells. Our data have therapeutic implications for targeting ARIH2 function.


Nature Genetics | 2016

Noncoding somatic and inherited single-nucleotide variants converge to promote ESR1 expression in breast cancer.

Swneke D. Bailey; Kinjal Desai; Ken Kron; Parisa Mazrooei; Nicholas A Sinnott-Armstrong; Aislinn E. Treloar; Mark Dowar; Kelsie L. Thu; David W. Cescon; Jennifer Silvester; S.Y. Cindy Yang; Xue Wu; Rossanna C. Pezo; Benjamin Haibe-Kains; Tak W. Mak; Philippe L. Bedard; Trevor J. Pugh; Richard C. Sallari; Mathieu Lupien

Sustained expression of the estrogen receptor-α (ESR1) drives two-thirds of breast cancer and defines the ESR1-positive subtype. ESR1 engages enhancers upon estrogen stimulation to establish an oncogenic expression program. Somatic copy number alterations involving the ESR1 gene occur in approximately 1% of ESR1-positive breast cancers, suggesting that other mechanisms underlie the persistent expression of ESR1. We report significant enrichment of somatic mutations within the set of regulatory elements (SRE) regulating ESR1 in 7% of ESR1-positive breast cancers. These mutations regulate ESR1 expression by modulating transcription factor binding to the DNA. The SRE includes a recurrently mutated enhancer whose activity is also affected by rs9383590, a functional inherited single-nucleotide variant (SNV) that accounts for several breast cancer risk–associated loci. Our work highlights the importance of considering the combinatorial activity of regulatory elements as a single unit to delineate the impact of noncoding genetic alterations on single genes in cancer.


Nature Communications | 2017

Gene isoforms as expression-based biomarkers predictive of drug response in vitro

Zhaleh Safikhani; Petr Smirnov; Kelsie L. Thu; Jennifer Silvester; Nehme El-Hachem; Rene Quevedo; Mathieu Lupien; Tak W. Mak; David W. Cescon; Benjamin Haibe-Kains

Next-generation sequencing technologies have recently been used in pharmacogenomic studies to characterize large panels of cancer cell lines at the genomic and transcriptomic levels. Among these technologies, RNA-sequencing enable profiling of alternatively spliced transcripts. Given the high frequency of mRNA splicing in cancers, linking this feature to drug response will open new avenues of research in biomarker discovery. To identify robust transcriptomic biomarkers for drug response across studies, we develop a meta-analytical framework combining the pharmacological data from two large-scale drug screening datasets. We use an independent pan-cancer pharmacogenomic dataset to test the robustness of our candidate biomarkers across multiple cancer types. We further analyze two independent breast cancer datasets and find that specific isoforms of IGF2BP2, NECTIN4, ITGB6, and KLHDC9 are significantly associated with AZD6244, lapatinib, erlotinib, and paclitaxel, respectively. Our results support isoform expressions as a rich resource for biomarkers predictive of drug response.Altered mRNA splicing features in many cancers, but it has not been linked to drug response. Here, with their meta-analytic framework, the authors analyse pharmacogenomic data to identify isoform-based biomarkers predictive of in vitro drug response, and show them to frequently be strong predictors.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Disruption of the anaphase-promoting complex confers resistance to TTK inhibitors in triple-negative breast cancer

Kelsie L. Thu; Jennifer Silvester; Meghan J. Elliott; W. Ba-alawi; M. H. Duncan; A. C. Elia; A. S. Mer; Petr Smirnov; Zhaleh Safikhani; Benjamin Haibe-Kains; Tak W. Mak; David W. Cescon

Significance Using functional genomic screens, we have identified resistance mechanisms to the clinical TTK protein kinase inhibitor (TTKi) CFI-402257 in breast cancer. As this and other TTKi are currently in clinical trials, understanding determinants of tumor drug response could permit rational selection of patients for treatment. We found that TTKi resistance is conferred by impairing anaphase-promoting complex/cyclosome (APC/C) function to minimize the lethal effects of mitotic segregation errors. Discovery of this mechanism in aneuploid cancer cells builds on previous reports indicating that weakening the APC/C promotes tolerance of chromosomal instability in diploid cells. Our work suggests that APC/C functional capacity may serve as a clinically useful biomarker of tumor response to TTKi that warrants investigation in ongoing clinical trials. TTK protein kinase (TTK), also known as Monopolar spindle 1 (MPS1), is a key regulator of the spindle assembly checkpoint (SAC), which functions to maintain genomic integrity. TTK has emerged as a promising therapeutic target in human cancers, including triple-negative breast cancer (TNBC). Several TTK inhibitors (TTKis) are being evaluated in clinical trials, and an understanding of the mechanisms mediating TTKi sensitivity and resistance could inform the successful development of this class of agents. We evaluated the cellular effects of the potent clinical TTKi CFI-402257 in TNBC models. CFI-402257 induced apoptosis and potentiated aneuploidy in TNBC lines by accelerating progression through mitosis and inducing mitotic segregation errors. We used genome-wide CRISPR/Cas9 screens in multiple TNBC cell lines to identify mechanisms of resistance to CFI-402257. Our functional genomic screens identified members of the anaphase-promoting complex/cyclosome (APC/C) complex, which promotes mitotic progression following inactivation of the SAC. Several screen candidates were validated to confer resistance to CFI-402257 and other TTKis using CRISPR/Cas9 and siRNA methods. These findings extend the observation that impairment of the APC/C enables cells to tolerate genomic instability caused by SAC inactivation, and support the notion that a measure of APC/C function could predict the response to TTK inhibition. Indeed, an APC/C gene expression signature is significantly associated with CFI-402257 response in breast and lung adenocarcinoma cell line panels. This expression signature, along with somatic alterations in genes involved in mitotic progression, represent potential biomarkers that could be evaluated in ongoing clinical trials of CFI-402257 or other TTKis.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Polo-like kinase 4 inhibition produces polyploidy and apoptotic death of lung cancers

Masanori Kawakami; Lisa Maria Mustachio; Lin Zheng; Yulong Chen; Jaime Rodriguez-Canales; Barbara Mino; Jonathan M. Kurie; Jason Roszik; Pamela Villalobos; Kelsie L. Thu; Jennifer Silvester; David W. Cescon; Ignacio I. Wistuba; Tak W. Mak; Xi Liu; Ethan Dmitrovsky

Significance Despite current treatments, lung cancers remain a major public health problem. Innovative ways are needed to treat or prevent these cancers. Centrosomes are critical for fidelity of mitosis. Abnormal centrosome numbers can cause aberrant mitosis and cell death. Polo-like kinase 4 (PLK4) regulates centriole duplication, and its deregulation alters centrosome number and mitosis. The potent PLK4 inhibitor CFI-400945 is reported here to exert marked antineoplastic effects against lung cancers. CDK2 inhibition also deregulates mitosis and was found to cooperate with PLK4 antagonism. CFI-400945 is now undergoing phase I clinical trial testing (NCT01954316). Taken together, targeting PLK4 for inhibition holds promise in lung cancer therapy either as a single agent or when combined with an agent that deregulates mitosis. Polo-like kinase 4 (PLK4) is a serine/threonine kinase regulating centriole duplication. CFI-400945 is a highly selective PLK4 inhibitor that deregulates centriole duplication, causing mitotic defects and death of aneuploid cancers. Prior work was substantially extended by showing CFI-400945 causes polyploidy, growth inhibition, and apoptotic death of murine and human lung cancer cells, despite expression of mutated KRAS or p53. Analysis of DNA content by propidium iodide (PI) staining revealed cells with >4N DNA content (polyploidy) markedly increased after CFI-400945 treatment. Centrosome numbers and mitotic spindles were scored. CFI-400945 treatment produced supernumerary centrosomes and mitotic defects in lung cancer cells. In vivo antineoplastic activity of CFI-400945 was established in mice with syngeneic lung cancer xenografts. Lung tumor growth was significantly inhibited at well-tolerated dosages. Phosphohistone H3 staining of resected lung cancers following CFI-400945 treatment confirmed the presence of aberrant mitosis. PLK4 expression profiles in human lung cancers were explored using The Cancer Genome Atlas (TCGA) and RNA in situ hybridization (RNA ISH) of microarrays containing normal and malignant lung tissues. PLK4 expression was significantly higher in the malignant versus normal lung and conferred an unfavorable survival (P < 0.05). Intriguingly, cyclin dependent kinase 2 (CDK2) antagonism cooperated with PLK4 inhibition. Taken together, PLK4 inhibition alone or as part of a combination regimen is a promising way to combat lung cancer.

Collaboration


Dive into the Jennifer Silvester's collaboration.

Top Co-Authors

Avatar

Tak W. Mak

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Andrew Wakeham

University Health Network

View shared research outputs
Top Co-Authors

Avatar

David W. Cescon

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Kelsie L. Thu

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Elia

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Jillian Haight

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Satoshi Inoue

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masato Sasaki

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Annick You-Ten

University Health Network

View shared research outputs
Researchain Logo
Decentralizing Knowledge