158 results on '"Senthil K. Muthuswamy"'
Search Results
2. Parallelized multidimensional analytic framework applied to mammary epithelial cells uncovers regulatory principles in EMT
- Author
-
Indranil Paul, Dante Bolzan, Ahmed Youssef, Keith A. Gagnon, Heather Hook, Gopal Karemore, Michael U. J. Oliphant, Weiwei Lin, Qian Liu, Sadhna Phanse, Carl White, Dzmitry Padhorny, Sergei Kotelnikov, Christopher S. Chen, Pingzhao Hu, Gerald V. Denis, Dima Kozakov, Brian Raught, Trevor Siggers, Stefan Wuchty, Senthil K. Muthuswamy, and Andrew Emili
- Subjects
Science - Abstract
Epithelial-to-mesenchymal transition (EMT) is a complex process regulated at multiple molecular levels. Here, the authors implement an analytic framework - PAMAF - to integrate data from twelve distinct omics modalities, which they use to understand the molecular changes and regulation during EMT in vitro.
- Published
- 2023
- Full Text
- View/download PDF
3. Polarity protein SCRIB interacts with SLC3A2 to regulate proliferation and tamoxifen resistance in ER+ breast cancer
- Author
-
Yasuhiro Saito, Shiori Matsuda, Naomi Ohnishi, Keiko Endo, Sanae Ashitani, Maki Ohishi, Ayano Ueno, Masaru Tomita, Koji Ueda, Tomoyoshi Soga, and Senthil K. Muthuswamy
- Subjects
Biology (General) ,QH301-705.5 - Abstract
A complex involving polarity protein SCRIB and the leucine amino acid transporter SLC7A5 promotes cell proliferation and tamoxifen resistance in estrogen receptor-positive breast cancer cells.
- Published
- 2022
- Full Text
- View/download PDF
4. Elevated levels of mitochondrial CoQ10 induce ROS-mediated apoptosis in pancreatic cancer
- Author
-
Tulin Dadali, Anne R. Diers, Shiva Kazerounian, Senthil K. Muthuswamy, Pallavi Awate, Ryan Ng, Saie Mogre, Carrie Spencer, Katerina Krumova, Hannah E. Rockwell, Justice McDaniel, Emily Y. Chen, Fei Gao, Karl T. Diedrich, Vijetha Vemulapalli, Leonardo O. Rodrigues, Viatcheslav R. Akmaev, Khampaseuth Thapa, Manuel Hidalgo, Arindam Bose, Vivek K. Vishnudas, A. James Moser, Elder Granger, Michael A. Kiebish, Stephane Gesta, Niven R. Narain, and Rangaprasad Sarangarajan
- Subjects
Medicine ,Science - Abstract
Abstract Reactive oxygen species (ROS) are implicated in triggering cell signalling events and pathways to promote and maintain tumorigenicity. Chemotherapy and radiation can induce ROS to elicit cell death allows for targeting ROS pathways for effective anti-cancer therapeutics. Coenzyme Q10 is a critical cofactor in the electron transport chain with complex biological functions that extend beyond mitochondrial respiration. This study demonstrates that delivery of oxidized Coenzyme Q10 (ubidecarenone) to increase mitochondrial Q-pool is associated with an increase in ROS generation, effectuating anti-cancer effects in a pancreatic cancer model. Consequent activation of cell death was observed in vitro in pancreatic cancer cells, and both human patient-derived organoids and tumour xenografts. The study is a first to demonstrate the effectiveness of oxidized ubidecarenone in targeting mitochondrial function resulting in an anti-cancer effect. Furthermore, these findings support the clinical development of proprietary formulation, BPM31510, for treatment of cancers with high ROS burden with potential sensitivity to ubidecarenone.
- Published
- 2021
- Full Text
- View/download PDF
5. PDX-derived organoids model in vivo drug response and secrete biomarkers
- Author
-
Ling Huang, Bruno Bockorny, Indranil Paul, Dipikaa Akshinthala, Pierre-Oliver Frappart, Omar Gandarilla, Arindam Bose, Veronica Sanchez-Gonzalez, Emily E. Rouse, Sylvain D. Lehoux, Nicole Pandell, Christine M. Lim, John G. Clohessy, Joseph Grossman, Raul Gonzalez, Sofia Perea Del Pino, George Daaboul, Mandeep S. Sawhney, Steven D. Freedman, Alexander Kleger, Richard D. Cummings, Andrew Emili, Lakshmi B. Muthuswamy, Manuel Hidalgo, and Senthil K. Muthuswamy
- Subjects
Oncology ,Medicine - Abstract
Patient-derived organoid models are proving to be a powerful platform for both basic and translational studies. Here we conduct a methodical analysis of pancreatic ductal adenocarcinoma (PDAC) tumor organoid drug response in paired patient-derived xenograft (PDX) and PDX-derived organoid (PXO) models grown under WNT-free culture conditions. We report a specific relationship between area under the curve value of organoid drug dose response and in vivo tumor growth, irrespective of the drug treatment. In addition, we analyzed the glycome of PDX and PXO models and demonstrate that PXOs recapitulate the in vivo glycan landscape. In addition, we identify a core set of 57 N-glycans detected in all 10 models that represent 50%–94% of the relative abundance of all N-glycans detected in each of the models. Last, we developed a secreted biomarker discovery pipeline using media supernatant of organoid cultures and identified potentially new extracellular vesicle (EV) protein markers. We validated our findings using plasma samples from patients with PDAC, benign gastrointestinal diseases, and chronic pancreatitis and discovered that 4 EV proteins are potential circulating biomarkers for PDAC. Thus, we demonstrate the utility of organoid cultures to not only model in vivo drug responses but also serve as a powerful platform for discovering clinically actionable serologic biomarkers.
- Published
- 2020
- Full Text
- View/download PDF
6. Differential Functions of Splicing Factors in Mammary Transformation and Breast Cancer Metastasis
- Author
-
SungHee Park, Mattia Brugiolo, Martin Akerman, Shipra Das, Laura Urbanski, Adam Geier, Anil K. Kesarwani, Martin Fan, Nathan Leclair, Kuan-Ting Lin, Leo Hu, Ian Hua, Joshy George, Senthil K. Muthuswamy, Adrian R. Krainer, and Olga Anczuków
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Misregulation of alternative splicing is a hallmark of human tumors, yet to what extent and how it contributes to malignancy are only beginning to be unraveled. Here, we define which members of the splicing factor SR and SR-like families contribute to breast cancer and uncover differences and redundancies in their targets and biological functions. We identify splicing factors frequently altered in human breast tumors and assay their oncogenic functions using breast organoid models. We demonstrate that not all splicing factors affect mammary tumorigenesis in MCF-10A cells. Specifically, the upregulation of SRSF4, SRSF6, or TRA2β disrupts acinar morphogenesis and promotes cell proliferation and invasion in MCF-10A cells. By characterizing the targets of these oncogenic splicing factors, we identify shared spliced isoforms associated with well-established cancer hallmarks. Finally, we demonstrate that TRA2β is regulated by the MYC oncogene, plays a role in metastasis maintenance in vivo, and its levels correlate with breast cancer patient survival. : Park et al. demonstrate that >50% of human breast tumors exhibit an alteration in one of the splicing factors from the SR protein family. Using in vitro and in vivo breast cancer models, they identify three splicing factors that promote cell proliferation and invasion by regulating isoforms associated with cancer hallmarks. Keywords: alternative RNA splicing, breast cancer, splicing factor, SR protein, metastasis, MYC, TRA2-beta, triple negative breast cancer
- Published
- 2019
- Full Text
- View/download PDF
7. Personalized tumor vaccine for pancreatic cancer
- Author
-
Shira, Orr, Ling, Huang, James, Moser, Dina, Stroopinsky, Omar, Gandarilla, Cori, DeCicco, Jessica, Liegel, Cansu, Tacettin, Adam, Ephraim, Giulia, Cheloni, Daniela, Torres, Donald, Kufe, Jacalyn, Rosenblatt, Manuel, Hidalgo, Senthil K, Muthuswamy, and David, Avigan
- Subjects
Cancer Research ,Oncology ,Immunology ,Immunology and Allergy - Abstract
Pancreatic cancer is a highly lethal malignancy often presenting with advanced disease and characterized by resistance to standard chemotherapy. Immune-based therapies such checkpoint inhibition have been largely ineffective such that pancreatic cancer is categorized as an immunologically "cold tumor". In the present study, we examine the therapeutic efficacy of a personalized cancer vaccine in which tumor cells are fused with dendritic cells (DC) resulting in the broad induction of antitumor immunity.In the KPC spontaneous pancreatic cancer murine model, we demonstrated that vaccination with DC/KPC fusions led to expansion of pancreatic cancer specific lymphocytes with an activated phenotype. Remarkably, vaccination led to a reduction in tumor bulk and near doubling of median survival in this highly aggressive model. In a second murine pancreatic model (Panc02), vaccination with DC/tumor fusions similarly led to expansion of tumor antigen specific lymphocytes and their infiltration to the tumor site. Having shown efficacy in immunocompetent murine models, we subsequently demonstrated that DC/tumor fusions generated from primary human pancreatic cancer and autologous DCs potently stimulate tumor specific cytotoxic lymphocyte responses.DC/tumor fusions induce the activation and expansion of tumor reactive lymphocytes with the capacity to infiltrate into the pancreatic cancer tumor bed.
- Published
- 2022
- Full Text
- View/download PDF
8. Data from Insights into Immune Escape During Tumor Evolution and Response to Immunotherapy Using a Rat Model of Breast Cancer
- Author
-
Kornelia Polyak, Senthil K. Muthuswamy, Deborah A. Dillon, Gordon J. Freeman, Sara M. Tolaney, Eliezer M. Van Allen, Tanya E. Keenan, Katherine C. Murphy, Bethlehem Lulseged, Ethan D. Krop, Shanshan Xie, Michael U.J. Oliphant, Nicholas W. Harper, Ernesto Rojas Jimenez, Maša Alečković, Anne Trinh, and Carlos R. Gil Del Alcazar
- Abstract
Animal models are critical for the preclinical validation of cancer immunotherapies. Unfortunately, mouse breast cancer models do not faithfully reproduce the molecular subtypes and immune environment of the human disease. In particular, there are no good murine models of estrogen receptor–positive (ER+) breast cancer, the predominant subtype in patients. Here, we show that Nitroso-N-methylurea–induced mammary tumors in outbred Sprague-Dawley rats recapitulate the heterogeneity for mutational profiles, ER expression, and immune evasive mechanisms observed in human breast cancer. We demonstrate the utility of this model for preclinical studies by dissecting mechanisms of response to immunotherapy using combination TGFBR inhibition and PD-L1 blockade. Short-term treatment of early-stage tumors induced durable responses. Gene expression profiling and spatial mapping classified tumors as inflammatory and noninflammatory, and identified IFNγ, T-cell receptor (TCR), and B-cell receptor (BCR) signaling, CD74/MHC II, and epithelium-interacting CD8+ T cells as markers of response, whereas the complement system, M2 macrophage phenotype, and translation in mitochondria were associated with resistance. We found that the expression of CD74 correlated with leukocyte fraction and TCR diversity in human breast cancer. We identified a subset of rat ER+ tumors marked by expression of antigen-processing genes that had an active immune environment and responded to treatment. A gene signature characteristic of these tumors predicted disease-free survival in patients with ER+ Luminal A breast cancer and overall survival in patients with metastatic breast cancer receiving anti–PD-L1 therapy. We demonstrate the usefulness of this preclinical model for immunotherapy and suggest examination to expand immunotherapy to a subset of patients with ER+ disease.See related Spotlight by Roussos Torres, p. 672
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure from Insights into Immune Escape During Tumor Evolution and Response to Immunotherapy Using a Rat Model of Breast Cancer
- Author
-
Kornelia Polyak, Senthil K. Muthuswamy, Deborah A. Dillon, Gordon J. Freeman, Sara M. Tolaney, Eliezer M. Van Allen, Tanya E. Keenan, Katherine C. Murphy, Bethlehem Lulseged, Ethan D. Krop, Shanshan Xie, Michael U.J. Oliphant, Nicholas W. Harper, Ernesto Rojas Jimenez, Maša Alečković, Anne Trinh, and Carlos R. Gil Del Alcazar
- Abstract
Supplementary Figure from Insights into Immune Escape During Tumor Evolution and Response to Immunotherapy Using a Rat Model of Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table from Insights into Immune Escape During Tumor Evolution and Response to Immunotherapy Using a Rat Model of Breast Cancer
- Author
-
Kornelia Polyak, Senthil K. Muthuswamy, Deborah A. Dillon, Gordon J. Freeman, Sara M. Tolaney, Eliezer M. Van Allen, Tanya E. Keenan, Katherine C. Murphy, Bethlehem Lulseged, Ethan D. Krop, Shanshan Xie, Michael U.J. Oliphant, Nicholas W. Harper, Ernesto Rojas Jimenez, Maša Alečković, Anne Trinh, and Carlos R. Gil Del Alcazar
- Abstract
Supplementary Table from Insights into Immune Escape During Tumor Evolution and Response to Immunotherapy Using a Rat Model of Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
11. Tables S1 – S6 from Discovery of New Targets to Control Metastasis in Pancreatic Cancer by Single-cell Transcriptomics Analysis of Circulating Tumor Cells
- Author
-
Manuel Hidalgo, Pedro P. Lopez-Casas, Fatima Al-Shahrour, Senthil K. Muthuswamy, Sofia Perea, Ling Huang, Yolanda Duran, Camino Menendez, Victoria Bonilla, Natalia Baños, Manuel Muñoz, Ana Dopazo, Bruno Bockorny, Javier Perales-Patón, and Spas Dimitrov-Markov
- Abstract
Supplementary Tables
- Published
- 2023
- Full Text
- View/download PDF
12. Abnormal exocrine–endocrine cell cross-talk promotes β-cell dysfunction and loss in MODY8
- Author
-
Sevim Kahraman, Ercument Dirice, Giorgio Basile, Danielle Diegisser, Jahedul Alam, Bente B. Johansson, Manoj K. Gupta, Jiang Hu, Ling Huang, Chew-Li Soh, Danwei Huangfu, Senthil K. Muthuswamy, Helge Raeder, Anders Molven, and Rohit N. Kulkarni
- Subjects
Physiology (medical) ,Endocrinology, Diabetes and Metabolism ,Internal Medicine ,Cell Biology - Published
- 2022
- Full Text
- View/download PDF
13. sup 2 from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental Figure 2. A) Relative expression Left panel: Mean of fluorescence and right panel: qPCR validation of miR-21-5p expression level in the set of cell lines, 3D-models and organoid compared to normal controls. B) qPCR analysis of PDCD4 and PTEN expression levels in the PANC1 stable cell lines (Lenti-21) inhibiting miR- 21 activity (pLenti-III-miR-off). C) qRT-PCR of miR-21-5p expression after lipofectamine transfection of PANC1, BxPC3 and PL-45 cells with anti-miR-21 inhibitor (mirVana ®) at the dose of 50nM after 48h. For qPCR each miRNA sample was normalized on the basis of it's 18s content and on the basis of GAPDH for mRNAs.
- Published
- 2023
- Full Text
- View/download PDF
14. sup 3 from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental Figure 3. A) iRGD-TAMRA (red) binding after 15 min at 4 degree in normal cells (Low integrins-NRP1) and in human and mice PDAC cells (High integrins-NRP1). Scale bars 200μm B) qRT-PCR of miR-21-5p expression after treatment of PANC1, BxPC3 and PL-45 cells with TPN-21 at the dose of 100nM after 48h. C) Representation of 3D-model treatment course with TPN-21. For qPCR each miRNA sample was normalized on the basis of its 18s content. Error bars, mean {plus minus} s.d *P = 0.01-0.05; **P = 0.001-0.01; ***P < 0.001; ****P < 0.0001 N.S., not significant, two-tailed t-test; n = 3 biological replicates.
- Published
- 2023
- Full Text
- View/download PDF
15. sup 1 from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental Figure 1. A) Left panel: Visualization of the miRNA profile of normal pancreas samples (n=3 control). Right panel: Visualization of the miRNA profile of PDAC PDX tumor samples (n=27 cases). Relative expression is shown as mean of fluorescence (MFI). miRNAs profiling was done through Firefly Circulating miRNA Assay and normalized using two miRNAs that are not significantly deregulated between all samples miR-22-3p and miR-30b-5p defined by the geNorm-like 37 algorithm. B) Visualization of miRNA profiles of PDAC cell lines (n=4), PDAC 3Dmodel (n=2) and Patient-derived-organoid (PDO 286 and PDO 281) compared to normal pancreatic cell and organoid (n=2 control). miRNAs profiling was done via Firefly Circulating miRNA Assay and normalized using miR-181b-5p, miR-103-3p, miR-30b-5p defined by the geNorm-like algorithm. Normalized miRNA signal intensities are presented as fold-change (log10 of the ratio between a probe value to the average of all the other samples for that probe). Green represents highly expressed miRNAs and red represents lowly expressed miRNAs.
- Published
- 2023
- Full Text
- View/download PDF
16. Data from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Purpose: Since drug responses vary between patients, it is crucial to develop pre-clinical or co-clinical strategies that forecast patient response. In this study, we tested whether RNA-based therapeutics were suitable for personalized medicine by using patient-derived-organoid (PDO) and patient-derived-xenograft (PDX) models.Experimental Design: We performed microRNA (miRNA) profiling of PDX samples to determine the status of miRNA deregulation in individual pancreatic ductal adenocarcinoma (PDAC) patients. To deliver personalized RNA-based-therapy targeting oncogenic miRNAs that form part of this common PDAC miRNA over-expression signature, we packaged antimiR oligonucleotides against one of these miRNAs in tumor-penetrating nanocomplexes (TPN) targeting cell surface proteins on PDAC tumors.Results: As a validation for our pre-clinical strategy, the therapeutic potential of one of our nano-drugs, TPN-21, was first shown to decrease tumor cell growth and survival in PDO avatars for individual patients, then in their PDX avatars.Conclusions: This general approach appears suitable for co-clinical validation of personalized RNA medicine and paves the way to prospectively identify patients with eligible miRNA profiles for personalized RNA-based therapy. Clin Cancer Res; 24(7); 1734–47. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental legend from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental legend
- Published
- 2023
- Full Text
- View/download PDF
18. sup 4 from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental Figure 4. A) qPCR analysis of miR-21-5p expression level after 4 treatments with TPN-21 in PDO 286. B) Relative tumor burden after (4 I.V injection of PBS (n=6) TPN-control n=6 or TPN-21 n=5 (5mg/kg). For qPCR each miRNA sample was normalized on the basis of its 18s content.
- Published
- 2023
- Full Text
- View/download PDF
19. table 1 a from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental table 1A List of miRNAs, miRBase accession number and mature sequence of 46 selected-miRNAs used to build the Firefly 46plex circulating custom panel. There miRNAs have previously been linked to pancreatic cancer or to KRAS.
- Published
- 2023
- Full Text
- View/download PDF
20. table 1 b from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental table 1B List of PDX pancreatic samples. Information regarding KRAS-mutation and gemcitabine resistance profile. S for sensitive, R for Resistant.
- Published
- 2023
- Full Text
- View/download PDF
21. sup 5 from Personalized RNA Medicine for Pancreatic Cancer
- Author
-
Frank J. Slack, Sangeeta N. Bhatia, Manuel Hidalgo, Senthil K. Muthuswamy, Jong Cheol Jeong, Emilia Pulver, Pedro P. Lopez-Casas, Rajesha Rupaimoole, Ling Huang, Liangliang Hao, and Maud-Emmanuelle Gilles
- Abstract
Supplemental Figure 5. A) Representatives PDO 286 images and metabolic activity measured from MTT assay after repeated TPN-21 or gemcitabine treatments (100nM) or both treatments. Scale bars 1000μm. B) Quantification of organoid size after treatments. Organoids size was measure through ImageJ software on a total of ⩾ 35 spheroids and are presented in arbitrary units (UA). Error bars, mean {plus minus} s.d *P = 0.01- 0.05; **P = 0.001-0.01; ***P < 0.001; ****P < 0.0001 N.S., not significant, two-tailed ttest; n = 3 biological replicates.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Data from Organoid Sensitivity Correlates with Therapeutic Response in Patients with Pancreatic Cancer
- Author
-
Manuel Hidalgo, Senthil K. Muthuswamy, Mark Callery, Tara S. Kent, Martin Smith, Tyler M. Berzin, Douglas Pleskow, Mandeep S. Sawhney, Robert Besaw, Roger B. Davis, Christine Lim, Supraja Narasimhan, Catherine Conahan, Mary Linton B. Peters, Benjamin Schlechter, Andrea J. Bullock, Bruno Bockorny, Jonah Cohen, Leo L. Tsai, Raul S. Gonzalez, Sofia Perea, Dipikaa Akshinthala, Ling Huang, Lakshmi Muthuswamy, and Joseph E. Grossman
- Abstract
Supplementary Data from Organoid Sensitivity Correlates with Therapeutic Response in Patients with Pancreatic Cancer
- Published
- 2023
- Full Text
- View/download PDF
23. Data from Organoid Sensitivity Correlates with Therapeutic Response in Patients with Pancreatic Cancer
- Author
-
Manuel Hidalgo, Senthil K. Muthuswamy, Mark Callery, Tara S. Kent, Martin Smith, Tyler M. Berzin, Douglas Pleskow, Mandeep S. Sawhney, Robert Besaw, Roger B. Davis, Christine Lim, Supraja Narasimhan, Catherine Conahan, Mary Linton B. Peters, Benjamin Schlechter, Andrea J. Bullock, Bruno Bockorny, Jonah Cohen, Leo L. Tsai, Raul S. Gonzalez, Sofia Perea, Dipikaa Akshinthala, Ling Huang, Lakshmi Muthuswamy, and Joseph E. Grossman
- Abstract
Purpose:Pancreatic ductal adenocarcinoma (PDAC) remains a significant health issue. For most patients, there are no options for targeted therapy, and existing treatments are limited by toxicity. The HOPE trial (Harnessing Organoids for PErsonalized Therapy) was a pilot feasibility trial aiming to prospectively generate patient-derived organoids (PDO) from patients with PDAC and test their drug sensitivity and correlation with clinical outcomes.Experimental Design:PDOs were established from a heterogeneous population of patients with PDAC including both basal and classical PDAC subtypes.Results:A method for classifying PDOs as sensitive or resistant to chemotherapy regimens was developed to predict the clinical outcome of patients. Drug sensitivity testing on PDOs correlated with clinical responses to treatment in individual patients.Conclusions:These data support the investigation of PDOs to guide treatment in prospective interventional trials in PDAC.
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Figure 3 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Figure 3 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure 4 from Mislocalization of the Cell Polarity Protein Scribble Promotes Mammary Tumorigenesis and Is Associated with Basal Breast Cancer
- Author
-
Senthil K. Muthuswamy, Robert D. Cardiff, Jennifer A. Pietenpol, Hal K. Berman, Brian D. Lehmann, Bernard Martin, Lakshmi B. Muthuswamy, Avi Z. Rosenberg, Kiyomi Araki, S. Dipikaa Akshinthala, and Michael E. Feigin
- Abstract
PDF file - 103K, Mislocalization of SCRIB enhances EGF-induced AKT activation.
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figure 2 from Mislocalization of the Cell Polarity Protein Scribble Promotes Mammary Tumorigenesis and Is Associated with Basal Breast Cancer
- Author
-
Senthil K. Muthuswamy, Robert D. Cardiff, Jennifer A. Pietenpol, Hal K. Berman, Brian D. Lehmann, Bernard Martin, Lakshmi B. Muthuswamy, Avi Z. Rosenberg, Kiyomi Araki, S. Dipikaa Akshinthala, and Michael E. Feigin
- Abstract
PDF file - 187K, H&E staining of hyperplasia and tumors from hSCRIBP305L mice.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figure 1 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Figure 1 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Figure 6 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Figure 6 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
29. Supplementary Figure Legends from Mislocalization of the Cell Polarity Protein Scribble Promotes Mammary Tumorigenesis and Is Associated with Basal Breast Cancer
- Author
-
Senthil K. Muthuswamy, Robert D. Cardiff, Jennifer A. Pietenpol, Hal K. Berman, Brian D. Lehmann, Bernard Martin, Lakshmi B. Muthuswamy, Avi Z. Rosenberg, Kiyomi Araki, S. Dipikaa Akshinthala, and Michael E. Feigin
- Abstract
PDF file - 68K
- Published
- 2023
- Full Text
- View/download PDF
30. Data from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
c-Myc is an oncogene transcription factor that causes cancer in many settings, including solid tumors that arise in the context of organized tissue structures. Given that disruption of tissue architecture frequently occurs in cancer, there is considerable interest in how cell organization impacts oncogene function. A previous report found that organization of mammary epithelial cells into defined 3-dimensional structures renders them insensitive to the effects of retrovirus-mediated overexpression of Myc, leading to the notion that organization tempers the sensitivity of individual cells to Myc activity. In this article, we report that epithelial cell organization does not profoundly alter Myc activity but, instead, suppresses Myc by modulating its expression. We show that the morphogenesis of mammary epithelial cells into organized acinar structures in vitro is accompanied by widespread changes in gene expression patterns, including a substantial decrease in the expression of Myc. Concomitant with the decrease in endogenous Myc expression, we observe a decrease in transcription from retroviral vectors during morphogenesis and find that Myc transgene expression in acini is much lower than in unorganized cells. This decrease in Myc transgene activity is responsible for the apparent recalcitrance of organized cells to ectopic Myc, as adenovirus-mediated expression of Myc in organized structures potently induces apoptosis. These observations reveal that organization does not alter the inherent response of epithelial cells to Myc and suggest that other tumor suppression mechanisms, apart from structure, antagonize Myc in the development of solid tumors. Cancer Res; 71(11); 3822–30. ©2011 AACR.
- Published
- 2023
- Full Text
- View/download PDF
31. Supplementary Figure 4 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Figure 4 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Figure 5 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Figure 5 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Tables 1-5 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Tables 1-5 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
34. Data from The Polarity Protein Par6 Induces Cell Proliferation and Is Overexpressed in Breast Cancer
- Author
-
Senthil K. Muthuswamy, D. Craig Allred, Srinjan Basu, Balasubramanian Lakshmi, Sangjun Lee, Victoria Aranda, and Marissa E. Nolan
- Abstract
The polarity protein complex Par6/atypical protein kinase (aPKC)/Cdc42 regulates polarization processes during epithelial morphogenesis, astrocyte migration, and axon specification. We, as well as others, have shown that this complex is also required for disruption of apical-basal polarity during the oncogene ErbB2-induced transformation and transforming growth factor β–induced epithelial-mesenchymal transition of mammary epithelial cells. Here, we report that expression of Par6 by itself in mammary epithelial cells induces epidermal growth factor–independent cell proliferation and development of hyperplastic three-dimensional acini without affecting apical-basal polarity. This is dependent on the ability of Par6 to interact with aPKC and Cdc42, but not Lgl and Par3, and its ability to promote sustained activation of MEK/ERK signaling. Down-regulation of Cdc42 or aPKC expression suppresses the ability of Par6 to induce proliferation, demonstrating that Par6 promotes cell proliferation by interacting with aPKC and Cdc42. We also show that Par6 is overexpressed in breast cancer–derived cell lines and in both precancerous breast lesions and advanced primary human breast cancers, suggesting that Par6 overexpression regulates tumor initiation and progression. Thus, in addition to regulating cell polarization processes, Par6 is an inducer of cell proliferation in breast epithelial cells. [Cancer Res 2008;68(20):8201–9]
- Published
- 2023
- Full Text
- View/download PDF
35. Supplementary Methods, Figure and Table Legends from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Methods, Figure and Table Legends from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
36. Supplementary Figure 2 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Author
-
William P. Tansey, Senthil K. Muthuswamy, Zhongming Zhao, Siyuan Zheng, Min Yu, and David R. Simpson
- Abstract
Supplementary Figure 2 from Epithelial Cell Organization Suppresses Myc Function by Attenuating Myc Expression
- Published
- 2023
- Full Text
- View/download PDF
37. Supplementary Figures 1-6 from The Polarity Protein Par6 Induces Cell Proliferation and Is Overexpressed in Breast Cancer
- Author
-
Senthil K. Muthuswamy, D. Craig Allred, Srinjan Basu, Balasubramanian Lakshmi, Sangjun Lee, Victoria Aranda, and Marissa E. Nolan
- Abstract
Supplementary Figures 1-6 from The Polarity Protein Par6 Induces Cell Proliferation and Is Overexpressed in Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
38. Supplementary Figure 1 from Mislocalization of the Cell Polarity Protein Scribble Promotes Mammary Tumorigenesis and Is Associated with Basal Breast Cancer
- Author
-
Senthil K. Muthuswamy, Robert D. Cardiff, Jennifer A. Pietenpol, Hal K. Berman, Brian D. Lehmann, Bernard Martin, Lakshmi B. Muthuswamy, Avi Z. Rosenberg, Kiyomi Araki, S. Dipikaa Akshinthala, and Michael E. Feigin
- Abstract
PDF file - 99K, Expression of SCRIB mRNA in human breast cancer.
- Published
- 2023
- Full Text
- View/download PDF
39. Supplementary Figure 3 from Mislocalization of the Cell Polarity Protein Scribble Promotes Mammary Tumorigenesis and Is Associated with Basal Breast Cancer
- Author
-
Senthil K. Muthuswamy, Robert D. Cardiff, Jennifer A. Pietenpol, Hal K. Berman, Brian D. Lehmann, Bernard Martin, Lakshmi B. Muthuswamy, Avi Z. Rosenberg, Kiyomi Araki, S. Dipikaa Akshinthala, and Michael E. Feigin
- Abstract
PDF file - 204K, Characterization of hSCRIBP305L tumors.
- Published
- 2023
- Full Text
- View/download PDF
40. A Large-Scale Proteomics Resource of Circulating Extracellular Vesicles for Biomarker Discovery in Pancreatic Cancer
- Author
-
Bruno Bockorny, Lakshmi Muthuswamy, Ling Huang, Marco Hadisurya, Christine Maria Lim, Leo L. Tsai, Ritu R. Gill, Jesse L. Wei, Andrea J. Bullock, Joseph E. Grossman, Robert J. Besaw, Supraja Narasimhan, W. Andy Tao, Sofia Perea, Mandeep S. Sawhney, Steven D. Freedman, Manuel Hidalgo, Anton Iliuk, and Senthil K. Muthuswamy
- Subjects
Article - Abstract
Pancreatic cancer has the worst prognosis of all common tumors. Earlier cancer diagnosis could increase survival rates and better assessment of metastatic disease could improve patient care. As such, there is an urgent need to develop biomarkers to diagnose this deadly malignancy earlier. Analyzing circulating extracellular vesicles (cEVs) using ‘liquid biopsies’ offers an attractive approach to diagnose and monitor disease status. However, it is important to differentiate EV-associated proteins enriched in patients with pancreatic ductal adenocarcinoma (PDAC) from those with benign pancreatic diseases such as chronic pancreatitis and intraductal papillary mucinous neoplasm (IPMN). To meet this need, we combined the novel EVtrap method for highly efficient isolation of EVs from plasma and conducted proteomics analysis of samples from 124 individuals, including patients with PDAC, benign pancreatic diseases and controls. On average, 912 EV proteins were identified per 100µL of plasma. EVs containing high levels of PDCD6IP, SERPINA12 and RUVBL2 were associated with PDAC compared to the benign diseases in both discovery and validation cohorts. EVs with PSMB4, RUVBL2 and ANKAR were associated with metastasis, and those with CRP, RALB and CD55 correlated with poor clinical prognosis. Finally, we validated a 7-EV protein PDAC signature against a background of benign pancreatic diseases that yielded an 89% prediction accuracy for the diagnosis of PDAC. To our knowledge, our study represents the largest proteomics profiling of circulating EVs ever conducted in pancreatic cancer and provides a valuable open-source atlas to the scientific community with a comprehensive catalogue of novel cEVs that may assist in the development of biomarkers and improve the outcomes of patients with PDAC.
- Published
- 2023
- Full Text
- View/download PDF
41. Organoid Sensitivity Correlates with Therapeutic Response in Patients with Pancreatic Cancer
- Author
-
Lakshmi Muthuswamy, Ling Huang, Raul S. Gonzalez, Jonah Cohen, Senthil K. Muthuswamy, Catherine Conahan, Supraja Narasimhan, Tyler M. Berzin, Mandeep S. Sawhney, Andrea J. Bullock, Mary Linton B. Peters, Sofia Perea, Roger B. Davis, Benjamin Schlechter, Bruno Bockorny, Dipikaa Akshinthala, Robert J. Besaw, Mark P. Callery, Douglas K. Pleskow, Leo L. Tsai, Martin P. Smith, Christine Maria Lim, Manuel Hidalgo, Tara S. Kent, and Joseph E. Grossman
- Subjects
Drug ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pancreatic ductal adenocarcinoma ,endocrine system diseases ,media_common.quotation_subject ,medicine.medical_treatment ,Article ,Targeted therapy ,03 medical and health sciences ,Basal (phylogenetics) ,0302 clinical medicine ,Hope trial ,Internal medicine ,Pancreatic cancer ,medicine ,Humans ,In patient ,Prospective Studies ,030304 developmental biology ,media_common ,0303 health sciences ,Chemotherapy ,business.industry ,medicine.disease ,digestive system diseases ,3. Good health ,Organoids ,Pancreatic Neoplasms ,030220 oncology & carcinogenesis ,business ,Carcinoma, Pancreatic Ductal - Abstract
Purpose: Pancreatic ductal adenocarcinoma (PDAC) remains a significant health issue. For most patients, there are no options for targeted therapy, and existing treatments are limited by toxicity. The HOPE trial (Harnessing Organoids for PErsonalized Therapy) was a pilot feasibility trial aiming to prospectively generate patient-derived organoids (PDO) from patients with PDAC and test their drug sensitivity and correlation with clinical outcomes. Experimental Design: PDOs were established from a heterogeneous population of patients with PDAC including both basal and classical PDAC subtypes. Results: A method for classifying PDOs as sensitive or resistant to chemotherapy regimens was developed to predict the clinical outcome of patients. Drug sensitivity testing on PDOs correlated with clinical responses to treatment in individual patients. Conclusions: These data support the investigation of PDOs to guide treatment in prospective interventional trials in PDAC.
- Published
- 2021
- Full Text
- View/download PDF
42. Insights into immune escape during tumor evolution and response to immunotherapy using a rat model of breast cancer
- Author
-
Carlos R. Gil Del Alcazar, Anne Trinh, Maša Alečković, Ernesto Rojas Jimenez, Nicholas W. Harper, Michael U.J. Oliphant, Shanshan Xie, Ethan D. Krop, Bethlehem Lulseged, Katherine C. Murphy, Tanya E. Keenan, Eliezer M. Van Allen, Sara M. Tolaney, Gordon J. Freeman, Deborah A. Dillon, Senthil K. Muthuswamy, and Kornelia Polyak
- Subjects
Cancer Research ,Immunology ,Receptors, Antigen, T-Cell ,Mammary Neoplasms, Experimental ,Breast Neoplasms ,Hormones ,Article ,Rats ,Rats, Sprague-Dawley ,Mice ,Immunologic Factors ,Animals ,Humans ,Female ,Immunotherapy ,Progesterone - Abstract
Animal models are critical for the preclinical validation of cancer immunotherapies. Unfortunately, mouse breast cancer models do not faithfully reproduce the molecular subtypes and immune environment of the human disease. In particular, there are no good murine models of estrogen receptor–positive (ER+) breast cancer, the predominant subtype in patients. Here, we show that Nitroso-N-methylurea–induced mammary tumors in outbred Sprague-Dawley rats recapitulate the heterogeneity for mutational profiles, ER expression, and immune evasive mechanisms observed in human breast cancer. We demonstrate the utility of this model for preclinical studies by dissecting mechanisms of response to immunotherapy using combination TGFBR inhibition and PD-L1 blockade. Short-term treatment of early-stage tumors induced durable responses. Gene expression profiling and spatial mapping classified tumors as inflammatory and noninflammatory, and identified IFNγ, T-cell receptor (TCR), and B-cell receptor (BCR) signaling, CD74/MHC II, and epithelium-interacting CD8+ T cells as markers of response, whereas the complement system, M2 macrophage phenotype, and translation in mitochondria were associated with resistance. We found that the expression of CD74 correlated with leukocyte fraction and TCR diversity in human breast cancer. We identified a subset of rat ER+ tumors marked by expression of antigen-processing genes that had an active immune environment and responded to treatment. A gene signature characteristic of these tumors predicted disease-free survival in patients with ER+ Luminal A breast cancer and overall survival in patients with metastatic breast cancer receiving anti–PD-L1 therapy. We demonstrate the usefulness of this preclinical model for immunotherapy and suggest examination to expand immunotherapy to a subset of patients with ER+ disease. See related Spotlight by Roussos Torres, p. 672
- Published
- 2022
43. Empirical identification and validation of tumor-targeting T cell receptors from circulation using autologous pancreatic tumor organoids
- Author
-
Qingda Meng, Shanshan Xie, G Kenneth Gray, Mohammad H Dezfulian, Omar Gandarilla, Weilin Li, Ling Huang, Dipikaa Akshinthala, Elizabeth Ferrer, Catherine Conahan, Sofia Perea Del Pino, Joseph Grossman, Stephen J Elledge, Manuel Hidalgo, and Senthil K Muthuswamy
- Subjects
Cancer Research ,T cell ,medicine.medical_treatment ,Immunology ,Receptors, Antigen, T-Cell ,immunologic techniques ,antigen-mediated ,clonal selection ,gastrointestinal neoplasms ,chemistry.chemical_compound ,Mice ,Immune system ,Antigen ,Cancer immunotherapy ,TIGIT ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Humans ,RC254-282 ,Pharmacology ,Clinical/Translational Cancer Immunotherapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Carboxyfluorescein succinimidyl ester ,Organoids ,Pancreatic Neoplasms ,medicine.anatomical_structure ,Oncology ,chemistry ,Cancer research ,Molecular Medicine ,CD8 - Abstract
BackgroundTumor-specific cytotoxic T cells and T cell receptors are effective tools for cancer immunotherapy. Most efforts to identify them rely on known antigens or lymphocytes that have infiltrated into the tumor bed. Approaches to empirically identify tumor-targeting T cells and T cell receptors by exploiting all antigens expressed on tumor cell surfaces are not well developed for most carcinomas, including pancreatic cancer.MethodsAutologous tumor organoids were stimulated with T cells from the patients’ peripheral blood for 2 weeks to generate the organoid-primed T (opT) cells. opT cell phenotype was analyzed by monitoring changes in the expression levels of 28 cell surface and checkpoint proteins. Expression of ligands of the immune checkpoints was investigated by immunohistochemistry staining. T cells were labeled with carboxyfluorescein succinimidyl ester (CFSE) and assayed by flow cytometry to monitor tumor-induced T cell proliferation changes. opT cell-mediated killing of three-dimensional organoids was measured using an M30 ELISA kit. T cell receptors (TCRs) were identified by deep sequencing of gDNA isolated from T cells, and the TCR specificity was confirmed by transferring TCRs to the T cell line SKW-3 or donor T cells.ResultsThe co-culture was effective in the generation of CD8 + or CD4+opT cells. The opT cells killed autologous tumors in a granzyme B or Fas-Fas ligand-dependent manner and expressed markers of tissue-resident memory phenotype. Each patient-derived opT cell culture displayed a unique complement of checkpoint proteins. Interestingly, only NKG2A blockade showed a potent increase in the interferon-γ production compared with blocking programmed cell death protein 1 (PD-1) or programmed cell death ligand 1 (PD-L1) or TIM3 or TIGIT or LAG3. Importantly, TCR sequencing demonstrated a dramatic clonal expansion of T cells with a restricted subset of TCRs. Cloning and transferring the TCRs to heterologous T cells was sufficient to confer tumor cell recognition and cytotoxic properties in a patient-specific manner.ConclusionWe report a platform for expanding tumor-targeting T cells from the peripheral blood of patients with pancreatic cancer. We identify the NKG2A-HLA-E axis as a potentially important checkpoint for CD8 +T cells for pancreatic cancer. Lastly, we demonstrate empirical identification of tumor-targeting TCRs that can be used for TCR-therapeutics.
- Published
- 2021
44. Genome-wide synthetic lethal screen unveils novel CAIX-NFS1/xCT axis as a targetable vulnerability in hypoxic solid tumors
- Author
-
Shawn C. Chafe, Franco J. Vizeacoumar, Ling Huang, Wells S. Brown, Paul C. McDonald, Oksana Nemirovsky, Fabrizio Carta, Shannon Awrey, Frederick S. Vizeacoumar, David F. Schaeffer, Andrew Metcalfe, Claudiu T. Supuran, Senthil K. Muthuswamy, Daniel J. Renouf, Geetha Venkateswaran, Shoukat Dedhar, and Joanna M. Karasinska
- Subjects
Iron ,Intracellular pH ,Regulator ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Carbonic anhydrase ,medicine ,Humans ,Carbonic Anhydrase IX ,Hypoxia ,Research Articles ,Cancer ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Reactive oxygen species ,Multidisciplinary ,biology ,Chemistry ,SciAdv r-articles ,Bicarbonate transport ,Cell Biology ,Adenosine ,Cell Hypoxia ,3. Good health ,Bicarbonates ,Carbon-Sulfur Lyases ,Enzyme ,Tumor progression ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Research Article ,medicine.drug - Abstract
Targeting carbonic anhydrase IX acidifies intracellular pH, disrupts redox homeostasis, and creates vulnerability to ferroptosis., The metabolic mechanisms involved in the survival of tumor cells within the hypoxic niche remain unclear. We carried out a synthetic lethal CRISPR screen to identify survival mechanisms governed by the tumor hypoxia–induced pH regulator carbonic anhydrase IX (CAIX). We identified a redox homeostasis network containing the iron-sulfur cluster enzyme, NFS1. Depletion of NFS1 or blocking cyst(e)ine availability by inhibiting xCT, while targeting CAIX, enhanced ferroptosis and significantly inhibited tumor growth. Suppression of CAIX activity acidified intracellular pH, increased cellular reactive oxygen species accumulation, and induced susceptibility to alterations in iron homeostasis. Mechanistically, inhibiting bicarbonate production by CAIX or sodium-driven bicarbonate transport, while targeting xCT, decreased adenosine 5′-monophosphate–activated protein kinase activation and increased acetyl–coenzyme A carboxylase 1 activation. Thus, an alkaline intracellular pH plays a critical role in suppressing ferroptosis, a finding that may lead to the development of innovative therapeutic strategies for solid tumors to overcome hypoxia- and acidosis-mediated tumor progression and therapeutic resistance.
- Published
- 2021
- Full Text
- View/download PDF
45. Polarity protein SCRIB interacts with SLC3A2 to regulate proliferation and tamoxifen resistance in ER+ breast cancer
- Author
-
Yasuhiro Saito, Shiori Matsuda, Naomi Ohnishi, Keiko Endo, Sanae Ashitani, Maki Ohishi, Ayano Ueno, Masaru Tomita, Koji Ueda, Tomoyoshi Soga, and Senthil K. Muthuswamy
- Subjects
Fusion Regulatory Protein 1, Heavy Chain ,Tumor Suppressor Proteins ,Medicine (miscellaneous) ,Membrane Proteins ,Breast Neoplasms ,Estrogens ,General Biochemistry, Genetics and Molecular Biology ,Large Neutral Amino Acid-Transporter 1 ,Cytoskeletal Proteins ,Tamoxifen ,Receptors, Estrogen ,Drug Resistance, Neoplasm ,Humans ,Female ,General Agricultural and Biological Sciences ,Cell Proliferation - Abstract
Estrogen receptor (ER) positive breast cancer represents 75% of all breast cancers in women. Although patients with ER+ cancers receive endocrine therapies, more than 30% develop resistance and succumb to the disease, highlighting the need to understand endocrine resistance. Here we show an unexpected role for the cell polarity protein SCRIB as a tumor-promoter and a regulator of endocrine resistance in ER-positive breast cancer cells. SCRIB expression is induced by estrogen signaling in a MYC-dependent manner. SCRIB interacts with SLC3A2, a heteromeric component of leucine amino acid transporter SLC7A5. SLC3A2 binds to the N-terminus of SCRIB to facilitate the formation of SCRIB/SLC3A2/LLGL2/SLC7A5 quaternary complex required for membrane localization of the amino acid transporter complex. Both SCRIB and SLC3A2 are required for cell proliferation and tamoxifen resistance in ER+ cells identifying a new role for the SCRIB/SLC3A2 complex in ER+ breast cancer.
- Published
- 2021
46. Differential Functions of Splicing Factors in Mammary Transformation and Breast Cancer Metastasis
- Author
-
Adrian R. Krainer, Shipra Das, Martin Fan, Nathan K. Leclair, Leo Hu, Olga Anczuków, Laura Urbanski, Adam Geier, SungHee Park, Martin Akerman, Ian Hua, Senthil K. Muthuswamy, Joshy George, Kuan-Ting Lin, Mattia Brugiolo, and Anil K. Kesarwani
- Subjects
0301 basic medicine ,RNA Splicing ,Breast Neoplasms ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Metastasis ,03 medical and health sciences ,Splicing factor ,0302 clinical medicine ,Breast cancer ,SR protein ,medicine ,Humans ,Neoplasm Metastasis ,skin and connective tissue diseases ,lcsh:QH301-705.5 ,Triple-negative breast cancer ,Alternative splicing ,Cancer ,medicine.disease ,030104 developmental biology ,lcsh:Biology (General) ,RNA splicing ,Cancer research ,RNA Splicing Factors ,030217 neurology & neurosurgery - Abstract
Summary: Misregulation of alternative splicing is a hallmark of human tumors, yet to what extent and how it contributes to malignancy are only beginning to be unraveled. Here, we define which members of the splicing factor SR and SR-like families contribute to breast cancer and uncover differences and redundancies in their targets and biological functions. We identify splicing factors frequently altered in human breast tumors and assay their oncogenic functions using breast organoid models. We demonstrate that not all splicing factors affect mammary tumorigenesis in MCF-10A cells. Specifically, the upregulation of SRSF4, SRSF6, or TRA2β disrupts acinar morphogenesis and promotes cell proliferation and invasion in MCF-10A cells. By characterizing the targets of these oncogenic splicing factors, we identify shared spliced isoforms associated with well-established cancer hallmarks. Finally, we demonstrate that TRA2β is regulated by the MYC oncogene, plays a role in metastasis maintenance in vivo, and its levels correlate with breast cancer patient survival. : Park et al. demonstrate that >50% of human breast tumors exhibit an alteration in one of the splicing factors from the SR protein family. Using in vitro and in vivo breast cancer models, they identify three splicing factors that promote cell proliferation and invasion by regulating isoforms associated with cancer hallmarks. Keywords: alternative RNA splicing, breast cancer, splicing factor, SR protein, metastasis, MYC, TRA2-beta, triple negative breast cancer
- Published
- 2019
47. LLGL2 rescues nutrient stress by promoting leucine uptake in ER+ breast cancer
- Author
-
Lewyn Li, John M. Asara, Augustin Luna, Yasuhiro Saito, Senthil K. Muthuswamy, Chris Sander, Myles Brown, Brian Raught, and Etienne Coyaud
- Subjects
0301 basic medicine ,Multidisciplinary ,Cell growth ,Cell ,Cancer ,Biology ,medicine.disease ,law.invention ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Breast cancer ,Cell culture ,law ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Suppressor ,Endocrine system ,Receptor - Abstract
Drosophila Lgl and its mammalian homologues, LLGL1 and LLGL2, are scaffolding proteins that regulate the establishment of apical-basal polarity in epithelial cells1,2. Whereas Lgl functions as a tumour suppressor in Drosophila1, the roles of mammalian LLGL1 and LLGL2 in cancer are unclear. The majority (about 75%) of breast cancers express oestrogen receptors (ERs)3, and patients with these tumours receive endocrine treatment4. However, the development of resistance to endocrine therapy and metastatic progression are leading causes of death for patients with ER+ disease4. Here we report that, unlike LLGL1, LLGL2 is overexpressed in ER+ breast cancer and promotes cell proliferation under nutrient stress. LLGL2 regulates cell surface levels of a leucine transporter, SLC7A5, by forming a trimeric complex with SLC7A5 and a regulator of membrane fusion, YKT6, to promote leucine uptake and cell proliferation. The oestrogen receptor targets LLGL2 expression. Resistance to endocrine treatment in breast cancer cells was associated with SLC7A5- and LLGL2-dependent adaption to nutrient stress. SLC7A5 was necessary and sufficient to confer resistance to tamoxifen treatment, identifying SLC7A5 as a potential therapeutic target for overcoming resistance to endocrine treatments in breast cancer. Thus, LLGL2 functions as a promoter of tumour growth and not as a tumour suppressor in ER+ breast cancer. Beyond breast cancer, adaptation to nutrient stress is critically important5, and our findings identify an unexpected role for LLGL2 in this process.
- Published
- 2019
- Full Text
- View/download PDF
48. SY11-2 Tumor organoid models for translational research and co-clinical applications
- Author
-
Senthil K. Muthuswamy
- Subjects
Oncology ,Hematology - Published
- 2022
- Full Text
- View/download PDF
49. A multi-tiered map of EMT defines major transition points and identifies vulnerabilities
- Author
-
Indranil Paul, Dante Bolzan, Ahmed Youssef, Keith A. Gagnon, Heather Hook, Gopal Karemore, Michael UJ Oliphant, Weiwei Lin, Qian Liu, Sadhna Phanse, Carl White, Dzmitry Padhorny, Sergei Kotelnikov, Guillaume P. Andrieu, Christopher S. Chen, Pingzhao Hu, Gerald V. Denis, Dima Kozakov, Brian Raught, Trevor Siggers, Stefan Wuchty, Senthil K. Muthuswamy, and Andrew Emili
- Subjects
Transcriptome ,Metabolomics ,medicine.anatomical_structure ,biology ,Proteome ,biology.protein ,medicine ,Cytochrome P450 ,Epithelial–mesenchymal transition ,Protein phosphatase 2 ,Nucleus ,Proto-oncogene tyrosine-protein kinase Src ,Cell biology - Abstract
SummaryEpithelial to mesenchymal transition (EMT) is a complex cellular program proceeding through a hybrid E/M state linked to cancer-associated stemness, migration and chemoresistance. Deeper molecular understanding of this dynamic physiological landscape is needed to define events which regulate the transition and entry into and exit from the E/M state. Here, we quantified >60,000 molecules across ten time points and twelve omic layers in human mammary epithelial cells undergoing TGFβ-induced EMT. Deep proteomic profiles of whole cells, nuclei, extracellular vesicles, secretome, membrane and phosphoproteome defined state-specific signatures and major transition points. Parallel metabolomics showed metabolic reprogramming preceded changes in other layers, while single-cell RNA sequencing identified transcription factors controlling entry into E/M. Covariance analysis exposed unexpected discordance between the molecular layers. Integrative causal modeling revealed co-dependencies governing entry into E/M that were verified experimentally using combinatorial inhibition. Overall, this dataset provides an unprecedented resource on TGFβ signaling, EMT and cancer.
- Published
- 2021
- Full Text
- View/download PDF
50. Elevated levels of mitochondrial CoQ10 induce ROS-mediated apoptosis in pancreatic cancer
- Author
-
Anne R. Diers, Vivek K. Vishnudas, Arindam Bose, Karl T. Diedrich, Shiva Kazerounian, Michael A. Kiebish, Manuel Hidalgo, Stephane Gesta, Carrie Spencer, Emily Y. Chen, Rangaprasad Sarangarajan, Fei Gao, Hannah E. Rockwell, Justice McDaniel, Vijetha Vemulapalli, Niven R. Narain, Pallavi Awate, Ryan Ng, Khampaseuth Thapa, A. James Moser, Saie Mogre, Senthil K. Muthuswamy, Viatcheslav R. Akmaev, Elder Granger, Katerina Krumova, Tulin Dadali, and Leonardo O. Rodrigues
- Subjects
Cell signaling ,Programmed cell death ,Cell Survival ,Ubiquinone ,Science ,Cell Respiration ,Mice, Nude ,Apoptosis ,Article ,Substrate Specificity ,chemistry.chemical_compound ,Oxygen Consumption ,Cell Line, Tumor ,Pancreatic cancer ,Organoid ,medicine ,Animals ,Humans ,Cancer ,Cell Proliferation ,Membrane Potential, Mitochondrial ,Coenzyme Q10 ,chemistry.chemical_classification ,Glycerol-3-Phosphate Dehydrogenase (NAD+) ,Reactive oxygen species ,Multidisciplinary ,Electron Transport Complex II ,medicine.disease ,Cancer metabolism ,In vitro ,Mitochondria ,Organoids ,Pancreatic Neoplasms ,Oxidative Stress ,chemistry ,Cancer research ,Medicine ,Reactive Oxygen Species - Abstract
Reactive oxygen species (ROS) are implicated in triggering cell signalling events and pathways to promote and maintain tumorigenicity. Chemotherapy and radiation can induce ROS to elicit cell death allows for targeting ROS pathways for effective anti-cancer therapeutics. Coenzyme Q10 is a critical cofactor in the electron transport chain with complex biological functions that extend beyond mitochondrial respiration. This study demonstrates that delivery of oxidized Coenzyme Q10 (ubidecarenone) to increase mitochondrial Q-pool is associated with an increase in ROS generation, effectuating anti-cancer effects in a pancreatic cancer model. Consequent activation of cell death was observed in vitro in pancreatic cancer cells, and both human patient-derived organoids and tumour xenografts. The study is a first to demonstrate the effectiveness of oxidized ubidecarenone in targeting mitochondrial function resulting in an anti-cancer effect. Furthermore, these findings support the clinical development of proprietary formulation, BPM31510, for treatment of cancers with high ROS burden with potential sensitivity to ubidecarenone.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.