34 results on '"Jennifer Silvester"'
Search Results
2. Gene isoforms as expression-based biomarkers predictive of drug response in vitro
- Author
-
Zhaleh Safikhani, Petr Smirnov, Kelsie L. Thu, Jennifer Silvester, Nehme El-Hachem, Rene Quevedo, Mathieu Lupien, Tak W. Mak, David Cescon, and Benjamin Haibe-Kains
- Subjects
Science - Abstract
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.
- Published
- 2017
- Full Text
- View/download PDF
3. CX-5461 is a DNA G-quadruplex stabilizer with selective lethality in BRCA1/2 deficient tumours
- Author
-
Hong Xu, Marco Di Antonio, Steven McKinney, Veena Mathew, Brandon Ho, Nigel J. O’Neil, Nancy Dos Santos, Jennifer Silvester, Vivien Wei, Jessica Garcia, Farhia Kabeer, Daniel Lai, Priscilla Soriano, Judit Banáth, Derek S. Chiu, Damian Yap, Daniel D. Le, Frank B. Ye, Anni Zhang, Kelsie Thu, John Soong, Shu-chuan Lin, Angela Hsin Chin Tsai, Tomo Osako, Teresa Algara, Darren N. Saunders, Jason Wong, Jian Xian, Marcel B. Bally, James D. Brenton, Grant W. Brown, Sohrab P. Shah, David Cescon, Tak W. Mak, Carlos Caldas, Peter C. Stirling, Phil Hieter, Shankar Balasubramanian, and Samuel Aparicio
- Subjects
Science - Abstract
Stabilization of DNA quadruplex structures (G4) is lethal for cells with a compromised DNA repair pathway. Here, the authors show that CX-5461, a small molecule in clinical trials as RNA polymerase inhibitor, has G4-stablization properties and can be repurposed to target DNA repair-defective cancers cells.
- Published
- 2017
- Full Text
- View/download PDF
4. Author Correction: Gene isoforms as expression-based biomarkers predictive of drug response in vitro
- Author
-
Zhaleh Safikhani, Petr Smirnov, Kelsie L. Thu, Jennifer Silvester, Nehme El-Hachem, Rene Quevedo, Mathieu Lupien, Tak W. Mak, David Cescon, and Benjamin Haibe-Kains
- Subjects
Science - Abstract
In the original version of this Article, financial support was not fully acknowledged. This error has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2018
- Full Text
- View/download PDF
5. Inhibition of the MNK1/2–eIF4E Axis Augments Palbociclib-Mediated Antitumor Activity in Melanoma and Breast Cancer
- Author
-
Sathyen A. Prabhu, Omar Moussa, Christophe Gonçalves, Judith H. LaPierre, Hsiang Chou, Fan Huang, Vincent R. Richard, Pault Y. M. Ferruzo, Elizabeth M. Guettler, Isabel Soria-Bretones, Laura Kirby, Natascha Gagnon, Jie Su, Jennifer Silvester, Sai Sakktee Krisna, April A. N. Rose, Karen E. Sheppard, David W. Cescon, Frédérick A. Mallette, Rene P. Zahedi, Christoph H. Borchers, Sonia V. del Rincon, and Wilson H. Miller
- Subjects
Cancer Research ,Oncology - Abstract
Aberrant cell-cycle progression is characteristic of melanoma, and CDK4/6 inhibitors, such as palbociclib, are currently being tested for efficacy in this disease. Despite the promising nature of CDK4/6 inhibitors, their use as single agents in melanoma has shown limited clinical benefit. Herein, we discovered that treatment of tumor cells with palbociclib induces the phosphorylation of the mRNA translation initiation factor eIF4E. When phosphorylated, eIF4E specifically engenders the translation of mRNAs that code for proteins involved in cell survival. We hypothesized that cancer cells treated with palbociclib use upregulated phosphorylated eIF4E (phospho-eIF4E) to escape the antitumor benefits of this drug. Indeed, we found that pharmacologic or genetic disruption of MNK1/2 activity, the only known kinases for eIF4E, enhanced the ability of palbociclib to decrease clonogenic outgrowth. Moreover, a quantitative proteomics analysis of melanoma cells treated with combined MNK1/2 and CDK4/6 inhibitors showed downregulation of proteins with critical roles in cell-cycle progression and mitosis, including AURKB, TPX2, and survivin. We also observed that palbociclib-resistant breast cancer cells have higher basal levels of phospho-eIF4E, and that treatment with MNK1/2 inhibitors sensitized these palbociclib-resistant cells to CDK4/6 inhibition. In vivo we demonstrate that the combination of MNK1/2 and CDK4/6 inhibition significantly increases the overall survival of mice compared with either monotherapy. Overall, our data support MNK1/2 inhibitors as promising drugs to potentiate the antineoplastic effects of palbociclib and overcome therapy-resistant disease.
- Published
- 2022
- Full Text
- View/download PDF
6. Supplementary Data from Inhibition of the MNK1/2–eIF4E Axis Augments Palbociclib-Mediated Antitumor Activity in Melanoma and Breast Cancer
- Author
-
Wilson H. Miller, Sonia V. del Rincon, Christoph H. Borchers, Rene P. Zahedi, Frédérick A. Mallette, David W. Cescon, Karen E. Sheppard, April A. N. Rose, Sai Sakktee Krisna, Jennifer Silvester, Jie Su, Natascha Gagnon, Laura Kirby, Isabel Soria-Bretones, Elizabeth M. Guettler, Pault Y. M. Ferruzo, Vincent R. Richard, Fan Huang, Hsiang Chou, Judith H. LaPierre, Christophe Gonçalves, Omar Moussa, and Sathyen A. Prabhu
- Abstract
List of differentially expressed proteins annotated by cluster with pathway analysis in BLM cells
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 1 from Inhibition of the MNK1/2–eIF4E Axis Augments Palbociclib-Mediated Antitumor Activity in Melanoma and Breast Cancer
- Author
-
Wilson H. Miller, Sonia V. del Rincon, Christoph H. Borchers, Rene P. Zahedi, Frédérick A. Mallette, David W. Cescon, Karen E. Sheppard, April A. N. Rose, Sai Sakktee Krisna, Jennifer Silvester, Jie Su, Natascha Gagnon, Laura Kirby, Isabel Soria-Bretones, Elizabeth M. Guettler, Pault Y. M. Ferruzo, Vincent R. Richard, Fan Huang, Hsiang Chou, Judith H. LaPierre, Christophe Gonçalves, Omar Moussa, and Sathyen A. Prabhu
- Abstract
Supplementary Figure 1
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Tyrosine Threonine Kinase Inhibition Eliminates Lung Cancers by Augmenting Apoptosis and Polyploidy
- Author
-
Ethan Dmitrovsky, Xi Liu, Tak W. Mak, Ignacio I. Wistuba, Jaime Rodriguez-Canales, David W Cescon, Kelsie L. Thu, Jennifer Silvester, Luisa M. Solis, Barbara Mino, Carmen Behrens, Wei Lu, Pamela Villalobos, Jonathan M. Kurie, Lisa Maria Mustachio, Jason Roszik, Yulong Chen, Masanori Kawakami, Zibo Chen, and Lin Zheng
- Abstract
The spindle assembly checkpoint maintains genomic integrity. A key component is tyrosine threonine kinase (TTK, also known as Mps1). TTK antagonism is hypothesized to cause genomic instability and cell death. Interrogating The Cancer Genome Atlas revealed high TTK expression in lung adenocarcinomas and squamous cell cancers versus the normal lung (P < 0.001). This correlated with an unfavorable prognosis in examined lung adenocarcinoma cases (P = 0.007). TTK expression profiles in lung tumors were independently assessed by RNA in situ hybridization. CFI-402257 is a highly selective TTK inhibitor. Its potent antineoplastic effects are reported here against a panel of well-characterized murine and human lung cancer cell lines. Significant antitumorigenic activity followed independent treatments of athymic mice bearing human lung cancer xenografts (6.5 mg/kg, P < 0.05; 8.5 mg/kg, P < 0.01) and immunocompetent mice with syngeneic lung cancers (P < 0.001). CFI-402257 antineoplastic mechanisms were explored. CFI-402257 triggered aneuploidy and apoptotic death of lung cancer cells without changing centrosome number. Reverse phase protein arrays (RPPA) of vehicle versus CFI-402257–treated lung cancers were examined using more than 300 critical growth-regulatory proteins. RPPA bioinformatic analyses discovered CFI-402257 enhanced MAPK signaling, implicating MAPK antagonism in augmenting TTK inhibitory effects. This was independently confirmed using genetic and pharmacologic repression of MAPK that promoted CFI-402257 anticancer actions. TTK antagonism exerted marked antineoplastic effects against lung cancers and MAPK inhibition cooperated. Future work should determine whether CFI-402257 treatment alone or with a MAPK inhibitor is active in the lung cancer clinic.
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Inhibition of the MNK1/2–eIF4E Axis Augments Palbociclib-Mediated Antitumor Activity in Melanoma and Breast Cancer
- Author
-
Wilson H. Miller, Sonia V. del Rincon, Christoph H. Borchers, Rene P. Zahedi, Frédérick A. Mallette, David W. Cescon, Karen E. Sheppard, April A. N. Rose, Sai Sakktee Krisna, Jennifer Silvester, Jie Su, Natascha Gagnon, Laura Kirby, Isabel Soria-Bretones, Elizabeth M. Guettler, Pault Y. M. Ferruzo, Vincent R. Richard, Fan Huang, Hsiang Chou, Judith H. LaPierre, Christophe Gonçalves, Omar Moussa, and Sathyen A. Prabhu
- Abstract
Aberrant cell-cycle progression is characteristic of melanoma, and CDK4/6 inhibitors, such as palbociclib, are currently being tested for efficacy in this disease. Despite the promising nature of CDK4/6 inhibitors, their use as single agents in melanoma has shown limited clinical benefit. Herein, we discovered that treatment of tumor cells with palbociclib induces the phosphorylation of the mRNA translation initiation factor eIF4E. When phosphorylated, eIF4E specifically engenders the translation of mRNAs that code for proteins involved in cell survival. We hypothesized that cancer cells treated with palbociclib use upregulated phosphorylated eIF4E (phospho-eIF4E) to escape the antitumor benefits of this drug. Indeed, we found that pharmacologic or genetic disruption of MNK1/2 activity, the only known kinases for eIF4E, enhanced the ability of palbociclib to decrease clonogenic outgrowth. Moreover, a quantitative proteomics analysis of melanoma cells treated with combined MNK1/2 and CDK4/6 inhibitors showed downregulation of proteins with critical roles in cell-cycle progression and mitosis, including AURKB, TPX2, and survivin. We also observed that palbociclib-resistant breast cancer cells have higher basal levels of phospho-eIF4E, and that treatment with MNK1/2 inhibitors sensitized these palbociclib-resistant cells to CDK4/6 inhibition. In vivo we demonstrate that the combination of MNK1/2 and CDK4/6 inhibition significantly increases the overall survival of mice compared with either monotherapy. Overall, our data support MNK1/2 inhibitors as promising drugs to potentiate the antineoplastic effects of palbociclib and overcome therapy-resistant disease.
- Published
- 2023
- Full Text
- View/download PDF
10. Figures S1-S6 from Tyrosine Threonine Kinase Inhibition Eliminates Lung Cancers by Augmenting Apoptosis and Polyploidy
- Author
-
Ethan Dmitrovsky, Xi Liu, Tak W. Mak, Ignacio I. Wistuba, Jaime Rodriguez-Canales, David W Cescon, Kelsie L. Thu, Jennifer Silvester, Luisa M. Solis, Barbara Mino, Carmen Behrens, Wei Lu, Pamela Villalobos, Jonathan M. Kurie, Lisa Maria Mustachio, Jason Roszik, Yulong Chen, Masanori Kawakami, Zibo Chen, and Lin Zheng
- Abstract
Figure S1: The structure of CFI-402257. Figure S2: TTK mRNA expression profiles in lung cancer cases with different smoking patterns, as shown in panel A. In this left panel N means no smoking history while Y indicates the presence of a smoking history. That was displayed in the right panel as never, former or current smokers. Panel B displays the anti-proliferative effects of CFI-402257 relative to vehicle treatments in lung adenocarcinoma cell lines. The area under the curve was calculated based on the proportion of control values and log10 dosage values to generate dose-response curves. Error bars are standard deviations. Figure S3: Immunoblot analysis of ED1 (A) and A549 (B) lung cancer cells after independent transfection with TTK-targeting (siRNA1 and siRNA2) or control siRNAs. Figure S4: Representative photomicrographs of 393P (A) or H1299 (B) lung cancer cell line-derived tumors after 21 days of CFI-402257 or vehicle treatments. (C) Syngeneic 393P tumor-bearing mice were treated with vehicle, 8.5 mg/kg CFI-402257 orally for 5 days, after which tumors were harvested for RPPA analyses (n = 5 mice per group). Comparison of the 8.5mg/kg CFI-402257-treated group to vehicle-treated groups. The proteins displayed above the red line are significantly different (P < 0.05). Figure S5: (A) The association between survival and expression of MAP2K1/MAP2K2 and TTK in combination using TCGA. Green: MAP2K low/TTK low, purple: MAP2K low/TTK high, orange: MAP2K high/TTK low and red: MAP2K high/TTK high. (B) The combination index (CI) for each dosage pair was calculated. The dash line indicates CI=1.0. Figure S6: TTK mutations in lung adenocarcinomas (A) and squamous cell carcinomas (B) in TCGA database.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 2 from Inhibition of the MNK1/2–eIF4E Axis Augments Palbociclib-Mediated Antitumor Activity in Melanoma and Breast Cancer
- Author
-
Wilson H. Miller, Sonia V. del Rincon, Christoph H. Borchers, Rene P. Zahedi, Frédérick A. Mallette, David W. Cescon, Karen E. Sheppard, April A. N. Rose, Sai Sakktee Krisna, Jennifer Silvester, Jie Su, Natascha Gagnon, Laura Kirby, Isabel Soria-Bretones, Elizabeth M. Guettler, Pault Y. M. Ferruzo, Vincent R. Richard, Fan Huang, Hsiang Chou, Judith H. LaPierre, Christophe Gonçalves, Omar Moussa, and Sathyen A. Prabhu
- Abstract
Supplementary Figure 2
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 3 from Inhibition of the MNK1/2–eIF4E Axis Augments Palbociclib-Mediated Antitumor Activity in Melanoma and Breast Cancer
- Author
-
Wilson H. Miller, Sonia V. del Rincon, Christoph H. Borchers, Rene P. Zahedi, Frédérick A. Mallette, David W. Cescon, Karen E. Sheppard, April A. N. Rose, Sai Sakktee Krisna, Jennifer Silvester, Jie Su, Natascha Gagnon, Laura Kirby, Isabel Soria-Bretones, Elizabeth M. Guettler, Pault Y. M. Ferruzo, Vincent R. Richard, Fan Huang, Hsiang Chou, Judith H. LaPierre, Christophe Gonçalves, Omar Moussa, and Sathyen A. Prabhu
- Abstract
Supplementary Figure 3
- Published
- 2023
- Full Text
- View/download PDF
13. The spindle assembly checkpoint is a therapeutic vulnerability of CDK4/6 inhibitor-resistant ER
- Author
-
Isabel, Soria-Bretones, Kelsie L, Thu, Jennifer, Silvester, Jennifer, Cruickshank, Samah, El Ghamrasni, Wail, Ba-Alawi, Graham C, Fletcher, Reza, Kiarash, Mitchell J, Elliott, Jordan J, Chalmers, Andrea C, Elia, Albert, Cheng, April A N, Rose, Mark R, Bray, Benjamin, Haibe-Kains, Tak W, Mak, and David W, Cescon
- Subjects
Drug Resistance, Neoplasm ,Cell Line, Tumor ,Neoplasms ,M Phase Cell Cycle Checkpoints - Abstract
Inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6i) are standard first-line treatments for metastatic ER
- Published
- 2022
14. Tyrosine Threonine Kinase Inhibition Eliminates Lung Cancers by Augmenting Apoptosis and Polyploidy
- Author
-
Wei Lu, Ignacio I. Wistuba, David W. Cescon, Lin Zheng, Tak W. Mak, Pamela Villalobos, Jennifer Silvester, Lisa Maria Mustachio, Barbara Mino, Ethan Dmitrovsky, Luisa M. Solis, Zibo Chen, Masanori Kawakami, Kelsie L. Thu, Xi Liu, Carmen Behrens, Jonathan M. Kurie, Yulong Chen, Jaime Rodriguez-Canales, and Jason Roszik
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Genome instability ,Cancer Research ,Programmed cell death ,Lung Neoplasms ,Carcinogenesis ,Apoptosis ,In situ hybridization ,Biology ,Polyploidy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,Cell Proliferation ,Centrosome ,Kinase ,Protein-Tyrosine Kinases ,medicine.disease ,Pyrimidines ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Pyrazoles ,Adenocarcinoma ,Anaphase - Abstract
The spindle assembly checkpoint maintains genomic integrity. A key component is tyrosine threonine kinase (TTK, also known as Mps1). TTK antagonism is hypothesized to cause genomic instability and cell death. Interrogating The Cancer Genome Atlas revealed high TTK expression in lung adenocarcinomas and squamous cell cancers versus the normal lung (P < 0.001). This correlated with an unfavorable prognosis in examined lung adenocarcinoma cases (P = 0.007). TTK expression profiles in lung tumors were independently assessed by RNA in situ hybridization. CFI-402257 is a highly selective TTK inhibitor. Its potent antineoplastic effects are reported here against a panel of well-characterized murine and human lung cancer cell lines. Significant antitumorigenic activity followed independent treatments of athymic mice bearing human lung cancer xenografts (6.5 mg/kg, P < 0.05; 8.5 mg/kg, P < 0.01) and immunocompetent mice with syngeneic lung cancers (P < 0.001). CFI-402257 antineoplastic mechanisms were explored. CFI-402257 triggered aneuploidy and apoptotic death of lung cancer cells without changing centrosome number. Reverse phase protein arrays (RPPA) of vehicle versus CFI-402257–treated lung cancers were examined using more than 300 critical growth-regulatory proteins. RPPA bioinformatic analyses discovered CFI-402257 enhanced MAPK signaling, implicating MAPK antagonism in augmenting TTK inhibitory effects. This was independently confirmed using genetic and pharmacologic repression of MAPK that promoted CFI-402257 anticancer actions. TTK antagonism exerted marked antineoplastic effects against lung cancers and MAPK inhibition cooperated. Future work should determine whether CFI-402257 treatment alone or with a MAPK inhibitor is active in the lung cancer clinic.
- Published
- 2019
- Full Text
- View/download PDF
15. Computational pharmacogenomic screen identifies drugs that phenocopy dipyridamole and potentiate anti-breast cancer activity of statins
- Author
-
Joseph Longo, David W. Cescon, Jenna E. van Leeuwen, Deena M.A. Gendoo, Jennifer Silvester, Linda Z. Penn, Benjamin Haibe-Kains, Emily Branchard, and Wail Ba alawi
- Subjects
Phenocopy ,Dipyridamole ,Oncology ,medicine.medical_specialty ,Anti breast cancer ,business.industry ,Internal medicine ,Pharmacogenomics ,medicine ,business ,medicine.drug - Abstract
Statins are a family of FDA-approved cholesterol-lowering drugs that inhibit the rate-limiting enzyme of the metabolic mevalonate pathway, which have been shown to have anti-cancer activity. As therapeutic efficacy increases when drugs are used in combination, we sought to identify agents, like dipyridamole, that potentiate statin-induced tumor cell death. As an antiplatelet agent dipyridamole will not be suitable for all cancer patients. Thus, we developed an integrative pharmacogenomics pipeline to identify agents that were similar to dipyridamole at the level of drug structure, in vitro sensitivity and molecular perturbation. To enrich for compounds expected to target the mevalonate pathway, we took a pathway-centric approach towards computational selection, which we called mevalonate drug network fusion (MVA-DNF). We validated two of the top ranked compounds, nelfinavir and honokiol and demonstrated that, like dipyridamole, they synergize with fluvastatin to potentiate tumour cell death by blocking the restorative feedback loop. This is achieved by inhibiting activation of the key transcription factor that induces mevalonate pathway gene transcription, sterol regulatory element-binding protein 2 (SREBP2). Mechanistically, the synergistic response of fluvastatin-nelfinavir and fluvastatin-honokiol was associated with similar transcriptomic and proteomic pathways, indicating a similar mechanism of action between nelfinavir and honokiol when combined with fluvastatin. Further analysis identified the canonical epithelial-mesenchymal transition (EMT) gene, E-cadherin as a biomarker of these synergistic responses across a large panel of breast cancer cell lines. Thus, our computational pharmacogenomic approach can identify novel compounds that phenocopy a compound of interest in a pathway-specific manner.
- Published
- 2021
- Full Text
- View/download PDF
16. Computational pharmacogenomic screen identifies drugs that potentiate the anti-breast cancer activity of statins
- Author
-
Jenna E. van Leeuwen, Wail Ba-Alawi, Emily Branchard, Jennifer Cruickshank, Wiebke Schormann, Joseph Longo, Jennifer Silvester, Peter L. Gross, David W. Andrews, David W. Cescon, Benjamin Haibe-Kains, Linda Z. Penn, and Deena M. A. Gendoo
- Subjects
Multidisciplinary ,Nelfinavir ,General Physics and Astronomy ,Mevalonic Acid ,Breast Neoplasms ,General Chemistry ,Dipyridamole ,Cadherins ,General Biochemistry, Genetics and Molecular Biology ,Cholesterol ,Vemurafenib ,Pharmacogenetics ,Humans ,Female ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Clotrimazole - Abstract
Statins, a family of FDA-approved cholesterol-lowering drugs that inhibit the rate-limiting enzyme of the mevalonate metabolic pathway, have demonstrated anticancer activity. Evidence shows that dipyridamole potentiates statin-induced cancer cell death by blocking a restorative feedback loop triggered by statin treatment. Leveraging this knowledge, we develop an integrative pharmacogenomics pipeline to identify compounds similar to dipyridamole at the level of drug structure, cell sensitivity and molecular perturbation. To overcome the complex polypharmacology of dipyridamole, we focus our pharmacogenomics pipeline on mevalonate pathway genes, which we name mevalonate drug-network fusion (MVA-DNF). We validate top-ranked compounds, nelfinavir and honokiol, and identify that low expression of the canonical epithelial cell marker, E-cadherin, is associated with statin-compound synergy. Analysis of remaining prioritized hits led to the validation of additional compounds, clotrimazole and vemurafenib. Thus, our computational pharmacogenomic approach identifies actionable compounds with pathway-specific activities.
- Published
- 2021
17. Computational pharmacogenomics screen identifies synergistic statin-compound combinations as anti-breast cancer therapies
- Author
-
Jenna E. van Leeuwen, David W. Cescon, Wail Ba-alawi, Jennifer Silvester, Deena M.A. Gendoo, Linda Z. Penn, Benjamin Haibe-Kains, Emily Branchard, and Joseph Longo
- Subjects
Honokiol ,Statin ,medicine.drug_class ,business.industry ,Pharmacology ,Dipyridamole ,chemistry.chemical_compound ,Nelfinavir ,chemistry ,Mechanism of action ,Pharmacogenomics ,medicine ,Mevalonate pathway ,medicine.symptom ,business ,medicine.drug ,Fluvastatin - Abstract
Statins are a family of FDA-approved cholesterol-lowering drugs that inhibit the rate-limiting enzyme of the metabolic mevalonate pathway, which have been shown to have anti-cancer activity. As therapeutic efficacy is increased when drugs are used in combination, we sought to identify agents, like dipyridamole, that potentiate statin-induced tumor cell death. As an antiplatelet agent dipyridamole will not be suitable for all cancer patients. Thus, we developed an integrative pharmacogenomics pipeline to identify agents that were similar to dipyridamole at the level of drug structure, in vitro sensitivity and molecular perturbation. To enrich for compounds expected to target the mevalonate pathway, we took a pathway-centric approach towards computational selection, which we called mevalonate drug network fusion (MVA-DNF). We validated two of the top ranked compounds, nelfinavir and honokiol and demonstrated that, like dipyridamole, they synergize with fluvastatin to potentiate tumor cell death by blocking the restorative feedback loop. This is achieved by inhibiting activation of the key transcription factor that induces mevalonate pathway gene transcription, sterol regulatory element-binding protein 2 (SREBP2). Mechanistically, the synergistic response of fluvastatin+nelfinavir and fluvastatin+honokiol was associated with similar transcriptomic and proteomic pathways, indicating a similar mechanism of action between nelfinavir and honokiol when combined with fluvastatin. Further analysis identified the canonical epithelial-mesenchymal transition (EMT) gene, E-cadherin as a biomarker of these synergistic responses across a large panel of breast cancer cell lines. Thus, our computational pharmacogenomic approach can identify novel compounds that phenocopy a compound of interest in a pathway-specific manner.Significance StatementWe provide a rapid and cost-effective strategy to expand a class of drugs with a similar phenotype. Our parent compound, dipyridamole, potentiated statin-induced tumor cell death by blocking the statin-triggered restorative feedback response that dampens statins pro-apoptotic activity. To identify compounds with this activity we performed a pharmacogenomic analysis to distinguish agents similar to dipyridamole in terms of structure, cell sensitivity and molecular perturbations. As dipyridamole has many reported activities, we focused our molecular perturbation analysis on the pathway inhibited by statins, the metabolic mevalonate pathway. Our strategy was successful as we validated nelfinavir and honokiol as dipyridamole-like drugs at both the phenotypic and molecular levels. Our pathway-specific pharmacogenomics approach will have broad applicability.
- Published
- 2020
- Full Text
- View/download PDF
18. Noncoding somatic and inherited single-nucleotide variants converge to promote ESR1 expression in breast cancer
- Author
-
Rossanna C. Pezo, Trevor J. Pugh, Mark Dowar, David W. Cescon, Ken Kron, Jennifer Silvester, Tak W. Mak, Mathieu Lupien, Benjamin Haibe-Kains, Aislinn E. Treloar, S. Y. Cindy Yang, Philippe L. Bedard, Richard C Sallari, Parisa Mazrooei, Kinjal Desai, Kelsie L. Thu, Xue Wu, Nicholas A Sinnott-Armstrong, and Swneke D. Bailey
- Subjects
0301 basic medicine ,Estrogen receptor ,Breast Neoplasms ,Regulatory Sequences, Nucleic Acid ,Biology ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Enhancer ,Regulation of gene expression ,Mutation ,Estrogen Receptor alpha ,Cancer ,medicine.disease ,Gene Expression Regulation, Neoplastic ,body regions ,030104 developmental biology ,Regulatory sequence ,MCF-7 Cells ,Female ,CRISPR-Cas Systems ,Estrogen receptor alpha ,Transcription Factors - Abstract
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.
- Published
- 2016
- Full Text
- View/download PDF
19. Reactive oxygen species modulate macrophage immunosuppressive phenotype through the up-regulation of PD-L1
- Author
-
Mandy F. Chu, Kelsey Hodgson, Luis Palomero, Jennifer Silvester, Tak W. Mak, Miguel Angel Pujana, Gordon Duncan, Paola Cappello, Tracy L. McGaha, Thorsten Berger, Andrew Wakeham, Mar Garcia-Valero, Cecilia Roux, Chiara Gorrini, Rahul Shinde, David W. Cescon, and Soode Moghadas Jafari
- Subjects
Paclitaxel ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,B7-H1 Antigen ,chemistry.chemical_compound ,Mice ,Drug Therapy ,Chemotherapy ,Immune suppression ,Macrophages ,Programmed death ligand-1 ,Reactive oxygen species ,PD-L1 ,Cell Line, Tumor ,Tumor Microenvironment ,Medicine ,Animals ,Humans ,Buthionine sulfoximine ,RNA, Messenger ,Buthionine Sulfoximine ,Triple-negative breast cancer ,Tumor microenvironment ,Multidisciplinary ,biology ,business.industry ,Glutathione ,Immune checkpoint ,Blockade ,Up-Regulation ,Phenotype ,chemistry ,PNAS Plus ,Tumor progression ,Cancer research ,biology.protein ,Female ,Chemokines ,business ,Reactive Oxygen Species ,Immunosuppressive Agents - Abstract
The combination of immune checkpoint blockade with chemotherapy is currently under investigation as a promising strategy for the treatment of triple negative breast cancer (TNBC). Tumor-associated macrophages (TAMs) are the most prominent component of the breast cancer microenvironment because they influence tumor progression and the response to therapies. Here we show that macrophages acquire an immunosuppressive phenotype and increase the expression of programmed death ligand-1 (PD-L1) when treated with reactive oxygen species (ROS) inducers such as the glutathione synthesis inhibitor, buthionine sulphoximine (BSO), and paclitaxel. Mechanistically, these agents cause accumulation of ROS that in turn activate NF-κB signaling to promote PD-L1 transcription and the release of immunosuppressive chemokines. Systemic in vivo administration of paclitaxel promotes PD-L1 accumulation on the surface of TAMS in a mouse model of TNBC, consistent with in vitro results. Combinatorial treatment with paclitaxel and an anti-mouse PD-L1 blocking antibody significantly improved the therapeutic efficacy of paclitaxel by reducing tumor burden and increasing the number of tumor-associated cytotoxic T cells. Our results provide a strong rationale for the use of anti–PD-L1 blockade in the treatment of TNBC patients. Furthermore, interrogation of chemotherapy-induced PD-L1 expression in TAMs is warranted to define appropriate patient selection in the use of PD-L1 blockade.
- Published
- 2019
20. Abstract PD2-03: CDK4/6 inhibitor-resistant ER+ breast cancer cell lines are hypersensitive to TTK inhibition
- Author
-
Graham C. Fletcher, Jennifer Silvester, Isabel Soria Bretones, Kelsie L. Thu, Jennifer Cruickshank, Mark R. Bray, Tak W. Mak, David W. Cescon, and Reza Kiarash
- Subjects
Cancer Research ,Cell growth ,Kinase ,business.industry ,Cancer ,Cell cycle ,Palbociclib ,medicine.disease ,Metastatic breast cancer ,Breast cancer ,Oncology ,Apoptosis ,medicine ,Cancer research ,business - Abstract
Inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6i), in combination with hormonal therapies, have become standard of care for the treatment of estrogen receptor-positive (ER+)/HER2-negative metastatic breast cancer. Despite demonstrating significant improvements in progression-free survival, acquired resistance to these inhibitors invariably develops. Recent analyses of clinical samples have identified emergent genomic alterations conferring acquired resistance to CDK4/6i, and begin to define the biology of this new clinical entity. Discovery of vulnerabilities of CDK4/6i-resistant tumours is imperative to improve the survival of this group of patients. We modeled CDK4/6i resistance in ER+ breast cancer cell lines using two complementary approaches: (1) spontaneous development of resistance upon continuous exposure to the palbociclib for 6-9 months, and (2) genetic engineering of RB1 loss of function. In both cases, palbociclib resistance was confirmed by colony formation and cell proliferation assays. To identify potential therapeutic strategies for CDK4/6i-resistant cells, we tested the in vitro activity of novel cell cycle inhibitors using sulforhodamine B (SRB) cytotoxicity assays. CFI-402257, a selective TTK inhibitor now in Phase I testing, induced significantly increased cytotoxicity in different CDK4/6i-resistant models compared to parental cell lines, including but not exclusively those with RB1 loss. CFI-402257 treatment caused defects in cell cycle progression and increased DNA damage and genomic instability in CDK4/6i-resistant cells, while these effects were mild in parental, CDK4/6i-sensitive cell lines. In some cases, these phenotypes were accompanied by an increase in apoptotic signaling. Analysis of the molecular determinants of these effects are being evaluated (additional results will be presented). In xenografts derived from MCF7 cells, CFI-402257 treatment completely abrogated the growth of RB1-KO tumours and had a much less pronounced effect on wild-type tumours. In summary, our results nominate the TTK inhibitor CFI-402257 as a promising therapeutic strategy for breast cancer patients who progress after CDK4/6 inhibition. A clinical trial testing this strategy is being launched. Citation Format: Isabel Soria Bretones, Kelsie L Thu, Jennifer Silvester, Reza Kiarash, Graham C Fletcher, Jennifer Cruickshank, Mark R Bray, Tak W Mak, David W Cescon. CDK4/6 inhibitor-resistant ER+ breast cancer cell lines are hypersensitive to TTK inhibition [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr PD2-03.
- Published
- 2020
- Full Text
- View/download PDF
21. The Antiarrhythmic Drug, Dronedarone, Demonstrates Cytotoxic Effects in Breast Cancer Independent of Thyroid Hormone Receptor Alpha 1 (THRα1) Antagonism
- Author
-
Jessica G. Cockburn, Kelsie L. Thu, Katarzyna J. Jerzak, David W. Cescon, William D. Gwynne, Benjamin Haibe-Kains, Zhaleh Safikhani, Tak W. Mak, Mitchell J. Elliott, Jennifer Silvester, John A. Hassell, and Anita Bane
- Subjects
0301 basic medicine ,Hormone Responsive ,Cell Survival ,lcsh:Medicine ,Antineoplastic Agents ,Breast Neoplasms ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Cytotoxic T cell ,Viability assay ,RNA, Small Interfering ,lcsh:Science ,Receptor ,Dronedarone ,Cell Proliferation ,Gene knockdown ,Multidisciplinary ,business.industry ,lcsh:R ,Drug Repositioning ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,lcsh:Q ,business ,Thyroid Hormone Receptors alpha ,medicine.drug - Abstract
Previous research has suggested that thyroid hormone receptor alpha 1 (THRα1), a hormone responsive splice variant, may play a role in breast cancer progression. Whether THRα1 can be exploited for anti-cancer therapy is unknown. The antiproliferative and antitumor effects of dronedarone, an FDA-approved anti-arrhythmic drug which has been shown to antagonize THRα1, was evaluated in breast cancer cell lines in vitro and in vivo. The THRα1 splice variant and the entire receptor, THRα, were also independently targeted using siRNA to determine the effect of target knockdown in vitro. In our study, dronedarone demonstrates cytotoxic effects in vitro and in vivo in breast cancer cell lines at doses and concentrations that may be clinically relevant. However, knockdown of either THRα1 or THRα did not cause substantial anti-proliferative or cytotoxic effects in vitro, nor did it alter the sensitivity to dronedarone. Thus, we conclude that dronedarone’s cytotoxic effect in breast cancer cell lines are independent of THRα or THRα1 antagonism. Further, the depletion of THRα or THRα1 does not affect cell viability or proliferation. Characterizing the mechanism of dronedarone’s anti-tumor action may facilitate drug repurposing or the development of new anti-cancer agents.
- Published
- 2018
- Full Text
- View/download PDF
22. Reply to Oegema et al.: CFI-400945 and Polo-like kinase 4 inhibition
- Author
-
Tak W. Mak, Xi Liu, Jason Roszik, David W. Cescon, Kelsie L. Thu, Jonathan M. Kurie, Jaime Rodriguez-Canales, Ethan Dmitrovsky, Yulong Chen, Barbara Mino, Jennifer Silvester, Masanori Kawakami, Pamela Villalobos, Ignacio I. Wistuba, Lisa Maria Mustachio, and Lin Zheng
- Subjects
0301 basic medicine ,PLK4 ,Indazoles ,Indoles ,Lung Neoplasms ,Multidisciplinary ,Chemistry ,Kinase ,Aurora B kinase ,Polo-like kinase ,Centrinone ,Polyploidy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer research ,Humans ,Letters - Abstract
In “CFI-400945 is not a selective cellular PLK4 inhibitor,” Oegema et al. (1) raise thoughtful comments about our article (2). We appreciate their interest and critique. They propose that CFI-400945 activity was not from Polo-like kinase (PLK4) inhibition and argue that antineoplastic activity was through Aurora B kinase (1), rather than PLK4 inhibition (2). Kinases often share related catalytic pockets, and targeting one kinase without affecting another is difficult to achieve. This is true for CFI-400945 (2) and centrinone, the PLK4 inhibitor (3) used by Oegema et al. (1). CFI-400945 is (by IC50) 38-fold more potent in causing PLK4 inhibition than in antagonizing Aurora B kinase (4, 5). Despite their claim … [↵][1]1Email: tak.mak{at}uhnresearch.ca. [1]: #xref-corresp-1-1
- Published
- 2018
- Full Text
- View/download PDF
23. Polo-like kinase 4 inhibition produces polyploidy and apoptotic death of lung cancers
- Author
-
Ethan Dmitrovsky, Ignacio I. Wistuba, Lin Zheng, Tak W. Mak, Barbara Mino, Kelsie L. Thu, Masanori Kawakami, Jennifer Silvester, Jason Roszik, David W. Cescon, Yulong Chen, Xi Liu, Jonathan M. Kurie, Lisa Maria Mustachio, Pamela Villalobos, and Jaime Rodriguez-Canales
- Subjects
0301 basic medicine ,PLK4 ,Multidisciplinary ,biology ,Cyclin-dependent kinase 2 ,In situ hybridization ,Polo-like kinase ,Biological Sciences ,medicine.disease ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,Propidium iodide ,KRAS ,Lung cancer ,Mitosis - Abstract
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.
- Published
- 2018
24. Disruption of the anaphase-promoting complex confers resistance to TTK inhibitors in triple-negative breast cancer
- Author
-
David W. Cescon, Wail Ba-alawi, Tak W. Mak, Jennifer Silvester, M. J. Elliott, Zhaleh Safikhani, M. H. Duncan, Benjamin Haibe-Kains, Kelsie L. Thu, Antonia Concetta Elia, Arvind Singh Mer, and Petr Smirnov
- Subjects
0301 basic medicine ,Genome instability ,Somatic cell ,Regulator ,Mitosis ,Cell Cycle Proteins ,Triple Negative Breast Neoplasms ,Protein Serine-Threonine Kinases ,Biology ,Anaphase-Promoting Complex-Cyclosome ,Genomic Instability ,03 medical and health sciences ,Cell Line, Tumor ,Humans ,Protein Kinase Inhibitors ,Triple-negative breast cancer ,Multidisciplinary ,Protein-Tyrosine Kinases ,Spindle checkpoint ,Pyrimidines ,030104 developmental biology ,PNAS Plus ,Drug Resistance, Neoplasm ,Cancer research ,Pyrazoles ,Female ,Anaphase-promoting complex - Abstract
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.
- Published
- 2018
- Full Text
- View/download PDF
25. Gene isoforms as expression-based biomarkers predictive of drug response in vitro
- Author
-
Petr Smirnov, David Cescon, Kelsie L. Thu, Mathieu Lupien, Zhaleh Safikhani, Tak W. Mak, Jennifer Silvester, and Benjamin Haibe-Kains
- Subjects
Drug ,0303 health sciences ,medicine.drug_class ,business.industry ,media_common.quotation_subject ,Computational biology ,Precision medicine ,medicine.disease ,Bioinformatics ,Tyrosine-kinase inhibitor ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Pharmacogenomics ,medicine ,Erlotinib ,Biomarker discovery ,business ,030304 developmental biology ,media_common ,medicine.drug ,EGFR inhibitors - Abstract
BackgroundOne of the main challenges in precision medicine is the identification of molecular features associated to drug response to provide clinicians with tools to select the best therapy for each individual cancer patient. The recent adoption of next-generation sequencing technologies enables accurate profiling of not only gene expression but also alternatively-spliced transcripts in large-scale pharmacogenomic studies. Given that altered mRNA splicing has been shown to be prominent in cancers, linking this feature to drug response will open new avenues of research in biomarker discovery.MethodsTo address the lack of reproducibility of drug sensitivity measurements across studies, we developed a meta-analytical framework combining the pharmacological data generated within the Cancer Cell Line Encyclopedia (CCLE) and the Genomics of Drug Sensitivity in Cancer (GDSC). Predictive models are fitted with CCLE RNA-seq data as predictor variables, controlled for tissue type, and combined GDSC and CCLE drug sensitivity values as dependent variables.ResultsWe first validated the biomarkers identified from GDSC and CCLE using an existing pharmacogenomic dataset of 70 breast cancer cell lines. We further selected four drugs with the most promising biomarkers to test whether their predictive value is robust to change in pharmacological assay. We successfully validated 10 isoform-based biomarkers predictive of drug response in breast cancer, including TGFA-001 for the MEK tyrosine kinase inhibitor (TKI) AZD6244, DUOX-001 for the EGFR inhibitor erlotinib, and CPEB4-001 transcript expression associated with lack of sensitivity to paclitaxel.ConclusionThe results of our meta-analysis of pharmacogenomic data suggest that isoforms represent a rich resource for biomarkers predictive of response to chemo- and targeted therapies. Our study also showed that the validation rate for this type of biomarkers is low (
- Published
- 2017
- Full Text
- View/download PDF
26. TAp73 is required for spermatogenesis and the maintenance of male fertility
- Author
-
Dave Cescon, Alessandro Rufini, Masato Sasaki, Tak W. Mak, Andrea Jurisicova, Colin McKerlie, Isaac S. Harris, Satoshi Inoue, Andrew J. Elia, Richard Tomasini, Jennifer Silvester, Lily Zhou, Ivano Amelio, Anne Brüstle, Massimiliano Agostini, Wanda Y. Li, Jillian Haight, Gerry Melino, Andrew Wakeham, David Dinsdale, and Sophie Lac
- Subjects
Male ,Aging ,Spermiogenesis ,Messenger ,Settore BIO/05 - Zoologia ,Apoptosis ,Cell Count ,Male infertility ,Mice ,Testis ,Progesterone ,Serpin ,Mice, Knockout ,ADAM17 ,Multidisciplinary ,Settore BIO/11 ,MMP13 ,Nuclear Proteins ,Timp ,Biological Sciences ,Apical ectoplasmic specialization ,Spermatozoa ,Cell biology ,DNA-Binding Proteins ,medicine.anatomical_structure ,Female ,Germ cell ,endocrine system ,Knockout ,ADAM17 Protein ,Biology ,ddc:570 ,Matrix Metalloproteinase 13 ,medicine ,Animals ,Humans ,RNA, Messenger ,Progenitor cell ,Spermatogenesis ,Infertility, Male ,Cell Proliferation ,Spermatid ,Tumor Suppressor Proteins ,Tumor Protein p73 ,medicine.disease ,Sperm ,ADAM Proteins ,DNA Damage ,Gene Expression Regulation ,Oxidative Stress ,Fertility ,Infertility ,Immunology ,RNA - Abstract
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. published
- Published
- 2014
- Full Text
- View/download PDF
27. BRCA1 interacts with Nrf2 to regulate antioxidant signaling and cell survival
- Author
-
Peter Bouwman, Satoshi Inoue, Jennifer Cruickshank, Bernard Martin, Bryan E. Snow, Alan Tseng, Dirk Brenner, Pegah S. Baniasadi, David W. Cescon, Andrew J. Elia, Melinda A. Musgrave, Jos Jonkers, Andrew Wakeham, Sam D. Molyneux, Chiara Gorrini, Isaac S. Harris, Rama Khokha, Hal K. Berman, Jennifer Silvester, Mona L. Gauthier, Tak W. Mak, Zoe Winterton-Perks, and Purna A. Joshi
- Subjects
Antioxidant ,endocrine system diseases ,medicine.medical_treatment ,Immunology ,Biology ,medicine.disease_cause ,environment and public health ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,medicine ,Immunology and Allergy ,skin and connective tissue diseases ,030304 developmental biology ,Regulation of gene expression ,chemistry.chemical_classification ,0303 health sciences ,Reactive oxygen species ,respiratory system ,3. Good health ,Cell biology ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,Carcinogenesis ,Oxidative stress - Abstract
Oxidative stress plays an important role in cancer development and treatment. Recent data implicate the tumor suppressor BRCA1 in regulating oxidative stress, but the molecular mechanism and the impact in BRCA1-associated tumorigenesis remain unclear. Here, we show that BRCA1 regulates Nrf2-dependent antioxidant signaling by physically interacting with Nrf2 and promoting its stability and activation. BRCA1-deficient mouse primary mammary epithelial cells show low expression of Nrf2-regulated antioxidant enzymes and accumulate reactive oxygen species (ROS) that impair survival in vivo. Increased Nrf2 activation rescues survival and ROS levels in BRCA1-null cells. Interestingly, 53BP1 inactivation, which has been shown to alleviate several defects associated with BRCA1 loss, rescues survival of BRCA1-null cells without restoring ROS levels. We demonstrate that estrogen treatment partially restores Nrf2 levels in the absence of BRCA1. Our data suggest that Nrf2-regulated antioxidant response plays a crucial role in controlling survival downstream of BRCA1 loss. The ability of estrogen to induce Nrf2 posits an involvement of an estrogen-Nrf2 connection in BRCA1 tumor suppression. Lastly, BRCA1-mutated tumors retain a defective antioxidant response that increases the sensitivity to oxidative stress. In conclusion, the role of BRCA1 in regulating Nrf2 activity suggests important implications for both the etiology and treatment of BRCA1-related cancers.
- Published
- 2013
- Full Text
- View/download PDF
28. CX-5461 is a DNA G-quadruplex stabilizer with selective lethality in BRCA1/2 deficient tumours
- Author
-
Marco Di Antonio, Peter C. Stirling, Jason Wong, Tak W. Mak, Carlos Caldas, Teresa Ruiz de Algara, Daniel D. Le, Frank B Ye, John Soong, Grant W. Brown, Samuel Aparicio, Tomo Osako, Steven McKinney, Veena Mathew, Phil Hieter, Sohrab P. Shah, Shankar Balasubramanian, Nancy Dos Santos, Priscilla Soriano, Judit P. Banáth, Daniel Lai, Shu chuan Lin, Anni Zhang, Nigel J. O'Neil, Jennifer Silvester, James D. Brenton, Brandon Ho, Jessica Garcia, Kelsie L. Thu, Angela Hsin Chin Tsai, Farhia Kabeer, Vivien Wei, Marcel B. Bally, David W. Cescon, Derek S. Chiu, Hong Xu, Damian Yap, Darren N. Saunders, Jian Xian, Di Antonio, Marco [0000-0002-7321-1867], Brenton, James [0000-0002-5738-6683], Caldas, Carlos [0000-0003-3547-1489], Balasubramanian, Shankar [0000-0002-0281-5815], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,COMET ASSAY ,DNA Repair ,Transcription, Genetic ,General Physics and Astronomy ,RECOMBINATION ,Quinolones ,PATHWAY ,chemistry.chemical_compound ,Mice ,Transcription (biology) ,Neoplasms ,Genotype ,Homologous Recombination ,DAMAGE ,Multidisciplinary ,BRCA1 Protein ,3. Good health ,Multidisciplinary Sciences ,TARGET ,Science & Technology - Other Topics ,GROWTH ,Female ,DNA Replication ,DNA damage ,Science ,Poly ADP ribose polymerase ,INHIBITION ,Saccharomyces cerevisiae ,Biology ,G-quadruplex ,DNA, Ribosomal ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,Chromosomal Instability ,Animals ,Humans ,Benzothiazoles ,Naphthyridines ,Caenorhabditis elegans ,REPAIR ,BRCA2 Protein ,Science & Technology ,Base Sequence ,Genome, Human ,GENOME INSTABILITY ,General Chemistry ,Xenograft Model Antitumor Assays ,Benzoxazines ,G-Quadruplexes ,030104 developmental biology ,chemistry ,Cell culture ,Cancer cell ,Cancer research ,DNA ,RNA-POLYMERASE I ,DNA Damage - Abstract
G-quadruplex DNAs form four-stranded helical structures and are proposed to play key roles in different cellular processes. Targeting G-quadruplex DNAs for cancer treatment is a very promising prospect. Here, we show that CX-5461 is a G-quadruplex stabilizer, with specific toxicity against BRCA deficiencies in cancer cells and polyclonal patient-derived xenograft models, including tumours resistant to PARP inhibition. Exposure to CX-5461, and its related drug CX-3543, blocks replication forks and induces ssDNA gaps or breaks. The BRCA and NHEJ pathways are required for the repair of CX-5461 and CX-3543-induced DNA damage and failure to do so leads to lethality. These data strengthen the concept of G4 targeting as a therapeutic approach, specifically for targeting HR and NHEJ deficient cancers and other tumours deficient for DNA damage repair. CX-5461 is now in advanced phase I clinical trial for patients with BRCA1/2 deficient tumours (Canadian trial, NCT02719977, opened May 2016)., Stabilization of DNA quadruplex structures (G4) is lethal for cells with a compromised DNA repair pathway. Here, the authors show that CX-5461, a small molecule in clinical trials as RNA polymerase inhibitor, has G4-stablization properties and can be repurposed to target DNA repair-defective cancers cells.
- Published
- 2016
29. ARIH2 is essential for embryogenesis, and its hematopoietic deficiency causes lethal activation of the immune system
- Author
-
Simon Preston, Yongkai Ow, Jesse G. Toe, Amy E. Lin, Hamish W Scott, Andrew Wakeham, Tak W. Mak, James P Cooney, Gregor Ebert, Jennifer Silvester, Raymond H. Kim, Samuel D. Saibil, Pamela S. Ohashi, Annick You-Ten, Arda Shahinian, Marc Pellegrini, Masato Sasaki, Dilan Dissanayake, and Gordon S. Duncan
- Subjects
Transcriptional Activation ,Multiple Organ Failure ,Ubiquitin-Protein Ligases ,Immunology ,Embryonic Development ,Mice ,Immune system ,Ubiquitin ,Transcription (biology) ,Animals ,Humans ,Immunology and Allergy ,Molecular Targeted Therapy ,Transcription factor ,Cells, Cultured ,Mice, Knockout ,biology ,NF-kappa B ,Ubiquitination ,Dendritic Cells ,NFKB1 ,Hematopoiesis ,Cell biology ,Ubiquitin ligase ,Mice, Inbred C57BL ,Disease Models, Animal ,Haematopoiesis ,Immune System ,biology.protein ,Stem cell - Abstract
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.
- Published
- 2012
- Full Text
- View/download PDF
30. Abstract 1948: Polo-like kinase 4 inhibition produces polyploidy and apoptotic death of lung cancers
- Author
-
Lin Zheng, Tak W. Mak, Pamela Villalobos, Xi Liu, Jason Roszik, Barbara Mino, Ignacio I. Wistuba, Jonathan M. Kurie, Lisa Maria Mustachio, Ethan Dmitrovsky, David W. Cescon, Kelsie L. Thu, Masanori Kawakami, Jaime Rodriguez-Canales, Jennifer Silvester, and Yulong Chen
- Subjects
0301 basic medicine ,PLK4 ,Cancer Research ,Programmed cell death ,Polo-like kinase ,Biology ,medicine.disease ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Oncology ,chemistry ,Centrosome ,medicine ,Cancer research ,KRAS ,Propidium iodide ,Lung cancer ,Mitosis - Abstract
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) is a serine/threonine kinase regulating centriole duplication and its deregulation alters centrosome number and mitosis. CFI-400945 is a highly selective PLK4 inhibitor that deregulates centriole duplication, causing mitotic defects and death of aneuploid cancers. Here, we explored CFI-400945 activity against lung cancer using in vitro and in vivo models. CFI-400945 caused 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 by individually staining treated and control cells with γ-tubulin, α-tubulin, and DAPI. CFI-400945 treatment produced supernumerary centrosomes and mitotic defects in the examined lung cancer cell lines. In vivo antineoplastic activity of CFI-400945 was established in mice with syngeneic lung cancer xenografts. Lung tumor growth was statistically significantly reduced at dosages that were well tolerated. Phospho-histone 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) indicating the clinical relevance of PLK4 expression in lung cancer. Intriguingly, cyclin dependent kinase 2 (CDK2) antagonism (that confers multipolar anaphase catastrophe by inhibiting centrosome clustering) showed synergistic effects (CI < 1) on lung cancer cell growth when combined with CDI-400945. This is an appealing cooperating regimen since each agent increases multipolar cell division and cancer cell death. CFI-400945 is undergoing phase I clinical trial testing (NCT01954316). Taken together, targeting PLK4 for inhibition holds promise for lung cancer therapy either as a single agent or when combined with another agent that deregulates mitosis. Citation Format: Masanori Kawakami, Lisa Maria Mustachio, Lin Zheng, Yulong Chen, Jaime Rodriguez-Canales, Barbara Mino, Jonathan M. Kurie, Jason Roszik, Pamela Andrea Villalobos, Kelsie L. Thu, David W. Cescon, Jennifer Silvester, Ignacio Wistuba, Tak W. Mak, Xi Liu, Ethan Dmitrovsky. Polo-like kinase 4 inhibition produces polyploidy and apoptotic death of lung cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1948.
- Published
- 2018
- Full Text
- View/download PDF
31. Author Correction: Gene isoforms as expression-based biomarkers predictive of drug response in vitro
- Author
-
David W. Cescon, Petr Smirnov, Nehme El-Hachem, Kelsie L. Thu, Jennifer Silvester, Zhaleh Safikhani, Rene Quevedo, Tak W. Mak, Benjamin Haibe-Kains, and Mathieu Lupien
- Subjects
Integrin beta Chains ,ComputerSystemsOrganization_COMPUTERSYSTEMIMPLEMENTATION ,Chemistry, Pharmaceutical ,General Physics and Astronomy ,02 engineering and technology ,01 natural sciences ,Neoplasms ,Drug response ,Protein Isoforms ,lcsh:Science ,Multidisciplinary ,RNA-Binding Proteins ,021001 nanoscience & nanotechnology ,Expression (architecture) ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,0210 nano-technology ,Gene isoform ,Paclitaxel ,Science ,MEDLINE ,Antineoplastic Agents ,Breast Neoplasms ,Computational biology ,Biology ,010402 general chemistry ,Article ,General Biochemistry, Genetics and Molecular Biology ,Erlotinib Hydrochloride ,Text mining ,Humans ,RNA, Messenger ,Author Correction ,Gene ,Genome, Human ,Sequence Analysis, RNA ,business.industry ,Lapatinib ,General Chemistry ,In vitro ,0104 chemical sciences ,Alternative Splicing ,Pharmacogenetics ,Quinazolines ,lcsh:Q ,Benzimidazoles ,Drug Screening Assays, Antitumor ,Carrier Proteins ,Transcriptome ,business ,Cell Adhesion Molecules ,Biomarkers - Abstract
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.
- Published
- 2018
32. Mule Regulates the Intestinal Stem Cell Niche via the Wnt Pathway and Targets EphB3 for Proteasomal and Lysosomal Degradation
- Author
-
Robert Nechanitzky, Johan H. van Es, Aaron Pollett, Ana Marques, Andrew J. Elia, Colin Reardon, David M. Sabatini, Zhenyue Hao, Andrew Wakeham, Ömer H. Yilmaz, Jillian Haight, Maureen A. Cox, Toshiro Sato, Miyeko D. Mana, Satoshi Inoue, Patrick M. Brauer, Jerome Fortin, Jennifer Silvester, Robert Cairns, Iok In Christine Chio, Tak W. Mak, Yi Sheng, Hans Clevers, Leanne Tworzyanski, Carmen Dominguez-Brauer, Massachusetts Institute of Technology. Department of Biology, Koch Institute for Integrative Cancer Research at MIT, Mana, Miyeko, Yilmaz, Omer, Sabatini, David, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
0301 basic medicine ,Carcinogenesis ,medicine.disease_cause ,Regenerative Medicine ,Medical and Health Sciences ,Oral and gastrointestinal ,Mice ,Ubiquitin ,Models ,2.1 Biological and endogenous factors ,Stem Cell Niche ,Aetiology ,Wnt Signaling Pathway ,Cancer ,Mice, Knockout ,biology ,Wnt signaling pathway ,Biological Sciences ,Endocytosis ,Ubiquitin ligase ,Cell biology ,Colo-Rectal Cancer ,Intestines ,Adenomatous Polyposis Coli ,Colonic Neoplasms ,Molecular Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,Tyrosine kinase ,Biotechnology ,Paneth Cells ,Proteasome Endopeptidase Complex ,Knockout ,Ubiquitin-Protein Ligases ,Ephrin-B3 ,Models, Biological ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,medicine ,Genetics ,Journal Article ,Animals ,Humans ,Progenitor cell ,Alleles ,Cell Proliferation ,Cell growth ,Tumor Suppressor Proteins ,HEK 293 cells ,Cell Biology ,Biological ,Stem Cell Research ,030104 developmental biology ,HEK293 Cells ,Mutation ,Proteolysis ,biology.protein ,Lysosomes ,Digestive Diseases ,Developmental Biology - Abstract
The E3 ubiquitin ligase Mule is often overexpressed in human colorectal cancers, but its role in gut tumorigenesis is unknown. Here, we show invivo that Mule controls murine intestinal stem and progenitor cell proliferation by modulating Wnt signaling via c-Myc. Mule also regulates protein levels of thereceptor tyrosine kinase EphB3 by targeting it for proteasomal and lysosomal degradation. In the intestine, EphB/ephrinB interactions position cells along the crypt-villus axis and compartmentalize incipient colorectal tumors. Our study thus unveils an important new avenue by which Mule acts as an intestinal tumor suppressor by regulation of the intestinal stem cell niche.
- Published
- 2016
- Full Text
- View/download PDF
33. 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
- Author
-
Tak W. Mak, M. Quintela-Fandino, Amber Ablack, Michael W. Tusche, Alessandro Rufini, E Arpaia, Pamela S. Ohashi, Anne Brüstle, Heiko Blaser, John D. Lewis, Gordon S. Duncan, C K Fleming, Hon S. Leong, Jennifer Silvester, Shruti Nambiar, and Evan F. Lind
- Subjects
MAPK/ERK pathway ,rho GTP-Binding Proteins ,Cancer Research ,RHOA ,Caveolin 1 ,RhoC ,Chick Embryo ,Integrin alpha5 ,Mice, SCID ,Metastasis ,Mice ,0302 clinical medicine ,Cell Movement ,Neoplasm Metastasis ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,0303 health sciences ,biology ,Cell migration ,3. Good health ,Cell biology ,src-Family Kinases ,rhoC GTP-Binding Protein ,030220 oncology & carcinogenesis ,RNA Interference ,Original Article ,Proto-oncogene tyrosine-protein kinase Src ,Protein Binding ,Src ,caveolin-1 ,Immunoblotting ,Molecular Sequence Data ,RAC1 ,03 medical and health sciences ,Cell Line, Tumor ,intravital microscopy ,Genetics ,medicine ,Animals ,cancer ,Amino Acid Sequence ,Rho-GTPase ,Molecular Biology ,030304 developmental biology ,Sequence Homology, Amino Acid ,Neoplasms, Experimental ,medicine.disease ,Enzyme Activation ,Mice, Inbred C57BL ,Crk-Associated Substrate Protein ,alpha5-integrin ,Cancer cell ,biology.protein ,Cancer research ,ras Proteins - Abstract
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 p130(Cas), 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 p130(Cas) 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 p130(Cas)/Rac1, FAK/Pyk2 and Ras/Erk1/2 signaling cascades.
- Published
- 2011
34. Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress
- Author
-
Suzanne K. Sawyer, Tak W. Mak, Tuula Kalliomaki, Brandon Faubert, Andrew E. H. Elia, Jennifer Silvester, Benjamin J. Fuerth, Ming Tsao, Karuppiah Kannan, Yao Yi, Annick You Ten, Jacqueline M. Mason, Vincent Nadeem, Murray O. Robinson, Ping Huang, Walbert Bakker, Russell G. Jones, Sireesha Yalavarthi, David Bungard, Craig B. Thompson, Kathrin Zaugg, Shelley L. Berger, Joseph D. Growney, Andrew Wakeham, Guohua Pan, Yasmin Moolani, Patrick T. Reilly, Katsuya Tsuchihara, Richard P. Hill, Reza Kiarash, Xunyi Luo, and Other departments
- Subjects
Lung Neoplasms ,Cell Survival ,Transplantation, Heterologous ,Apoptosis ,AMP-Activated Protein Kinases ,Mice ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,AMP-activated protein kinase ,Stress, Physiological ,Cell Line, Tumor ,Genetics ,Animals ,Humans ,Glycolysis ,RNA, Messenger ,Carnitine O-palmitoyltransferase ,Hypoxia ,Cells, Cultured ,Embryonic Stem Cells ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Carnitine O-Palmitoyltransferase ,biology ,TOR Serine-Threonine Kinases ,Reproducibility of Results ,Metabolism ,HCT116 Cells ,Warburg effect ,Up-Regulation ,Cell biology ,Gene Expression Regulation, Neoplastic ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Homeostasis ,Research Paper ,Developmental Biology - Abstract
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.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.