25 results on '"Joelle Baddour"'
Search Results
2. Lactate-mediated epigenetic reprogramming regulates formation of human pancreatic cancer-associated fibroblasts
- Author
-
Tushar D Bhagat, Dagny Von Ahrens, Meelad Dawlaty, Yiyu Zou, Joelle Baddour, Abhinav Achreja, Hongyun Zhao, Lifeng Yang, Brijesh Patel, Changsoo Kwak, Gaurav S Choudhary, Shanisha Gordon-Mitchell, Srinivas Aluri, Sanchari Bhattacharyya, Srabani Sahu, Prafulla Bhagat, Yiting Yu, Matthias Bartenstein, Orsi Giricz, Masako Suzuki, Davendra Sohal, Sonal Gupta, Paola A Guerrero, Surinder Batra, Michael Goggins, Ulrich Steidl, John Greally, Beamon Agarwal, Kith Pradhan, Debabrata Banerjee, Deepak Nagrath, Anirban Maitra, and Amit Verma
- Subjects
pancreas ,stroma ,methylation ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Even though pancreatic ductal adenocarcinoma (PDAC) is associated with fibrotic stroma, the molecular pathways regulating the formation of cancer associated fibroblasts (CAFs) are not well elucidated. An epigenomic analysis of patient-derived and de-novo generated CAFs demonstrated widespread loss of cytosine methylation that was associated with overexpression of various inflammatory transcripts including CXCR4. Co-culture of neoplastic cells with CAFs led to increased invasiveness that was abrogated by inhibition of CXCR4. Metabolite tracing revealed that lactate produced by neoplastic cells leads to increased production of alpha-ketoglutarate (aKG) within mesenchymal stem cells (MSCs). In turn, aKG mediated activation of the demethylase TET enzyme led to decreased cytosine methylation and increased hydroxymethylation during de novo differentiation of MSCs to CAF. Co-injection of neoplastic cells with TET-deficient MSCs inhibited tumor growth in vivo. Thus, in PDAC, a tumor-mediated lactate flux is associated with widespread epigenomic reprogramming that is seen during CAF formation.
- Published
- 2019
- Full Text
- View/download PDF
3. Amplification of USP13 drives ovarian cancer metabolism
- Author
-
Cecil Han, Lifeng Yang, Hyun Ho Choi, Joelle Baddour, Abhinav Achreja, Yunhua Liu, Yujing Li, Jiada Li, Guohui Wan, Cheng Huang, Guang Ji, Xinna Zhang, Deepak Nagrath, and Xiongbin Lu
- Subjects
Science - Abstract
Cancer cells need to reprogramme their metabolism to allow rapid cell proliferation. Here, the authors show that USP13is amplified in ovarian cancer and its protein product, a deubiquitinase, drives tumour progression by rewiring the metabolism of cancer cells by stabilising two critical metabolic enzymes.
- Published
- 2016
- Full Text
- View/download PDF
4. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism
- Author
-
Hongyun Zhao, Lifeng Yang, Joelle Baddour, Abhinav Achreja, Vincent Bernard, Tyler Moss, Juan C Marini, Thavisha Tudawe, Elena G Seviour, F Anthony San Lucas, Hector Alvarez, Sonal Gupta, Sourindra N Maiti, Laurence Cooper, Donna Peehl, Prahlad T Ram, Anirban Maitra, and Deepak Nagrath
- Subjects
cancer metabolism ,tumor microenvironment ,exosomes ,metabolic flux analysis ,reductive carboxylation ,macropinocytosis ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Cancer-associated fibroblasts (CAFs) are a major cellular component of tumor microenvironment in most solid cancers. Altered cellular metabolism is a hallmark of cancer, and much of the published literature has focused on neoplastic cell-autonomous processes for these adaptations. We demonstrate that exosomes secreted by patient-derived CAFs can strikingly reprogram the metabolic machinery following their uptake by cancer cells. We find that CAF-derived exosomes (CDEs) inhibit mitochondrial oxidative phosphorylation, thereby increasing glycolysis and glutamine-dependent reductive carboxylation in cancer cells. Through 13C-labeled isotope labeling experiments we elucidate that exosomes supply amino acids to nutrient-deprived cancer cells in a mechanism similar to macropinocytosis, albeit without the previously described dependence on oncogenic-Kras signaling. Using intra-exosomal metabolomics, we provide compelling evidence that CDEs contain intact metabolites, including amino acids, lipids, and TCA-cycle intermediates that are avidly utilized by cancer cells for central carbon metabolism and promoting tumor growth under nutrient deprivation or nutrient stressed conditions.
- Published
- 2016
- Full Text
- View/download PDF
5. Abstract 3425: The discovery and characterization of CFT1946: A potent, selective, and orally bioavailable degrader of mutant BRAF for the treatment of BRAF-driven cancers
- Author
-
Yanke Liang, Mathew E. Sowa, Katrina L. Jackson, Jeffrey R. Simard, Bridget Kreger, Ping Li, Laura Poling, Joelle Baddour, Andrew Good, Hongwei Huang, Scott Eron, Christopher G. Nasveschuk, Robert Yu, Mark Fitzgerald, Victoria Garza, Morgan W. O’Shea, Gesine Veits, Jeremy Y. Yap, Moses Moustakim, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe S. Patel, Richard Deibler, Kyle S. Cole, David Cocozziello, Fazlur Rahman, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Roy M. Pollock, and Stewart L. Fisher
- Subjects
Cancer Research ,Oncology - Abstract
The BRAF kinase plays a critical role in the MAPK signaling pathway and is mutated in ~8% of all human cancers including melanoma (~60%), thyroid (~60%), and lung adenocarcinoma (~10%). The most common mutation in BRAF is V600E (Class I), which is found in >70% in these cancers. Despite the clinical success of approved small molecule inhibitors of BRAF V600E (vemurafenib, dabrafenib and encorafenib), this remains an area of unmet medical need because nearly all patients progress, due to either primary or acquired resistance. A bifunctional degradation activating compound (or BiDACTM degrader) may address the liabilities of approved drugs by overcoming, or preventing the emergence of, resistance to BRAF inhibitors. Here we describe CFT1946, an orally bioavailable cereblon-based BiDAC degrader of BRAF V600 mutant proteins, and provide an overview of the medicinal chemistry path leading to its discovery. CFT1946 degrades BRAF V600 mutant proteins, while maintaining exquisite selectivity against the proteome, sparing wild type BRAF (BRAF-WT), ARAF, and CRAF. In A375 cells, CFT1946 potently degraded BRAF V600E and inhibited ERK phosphorylation and cell growth while having no effect in the mutant KRAS, BRAF-WT driven cell line HCT116. In the A375 xenograft model, oral delivery of CFT1946 at 10 mg/kg PO BID resulted in deeper and more durable tumor regression compared to a clinically relevant dose of encorafenib. Further evaluation of CFT1946 in an engineered, clinically relevant BRAFi-resistant A375 cell line (endogenous BRAF V600E + engineered expression of NRAS Q61K) demonstrated that CFT1946 both degraded BRAF V600E and caused a loss of viability in these cells, while treatment with encorafenib had no effect. In xenografts derived from this BRAFi-resistant cell line, oral dosing of CFT1946 as a single agent led to tumor growth inhibition, while treatment with a clinically relevant dose of encorafenib had no effect on tumor growth. Furthermore, dosing CFT1946 in combination with the MEK inhibitor, trametinib, resulted in significant tumor regression, whereas combining encorafenib with the same dose of trametinib had no effect. The medicinal chemistry campaign resulting in CFT1946 focused on the improvement of in vivo pharmacokinetics and rational linker design to achieve high oral bioavailability in a beyond Rule of 5 heterobifunctional degrader. The preclinical data presented herein support the planned Phase 1/2 clinical trial of CFT1946 for the treatment of BRAF-V600 mutant solid tumors. Citation Format: Yanke Liang, Mathew E. Sowa, Katrina L. Jackson, Jeffrey R. Simard, Bridget Kreger, Ping Li, Laura Poling, Joelle Baddour, Andrew Good, Hongwei Huang, Scott Eron, Christopher G. Nasveschuk, Robert Yu, Mark Fitzgerald, Victoria Garza, Morgan W. O’Shea, Gesine Veits, Jeremy Y. Yap, Moses Moustakim, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe S. Patel, Richard Deibler, Kyle S. Cole, David Cocozziello, Fazlur Rahman, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Roy M. Pollock, Stewart L. Fisher. The discovery and characterization of CFT1946: A potent, selective, and orally bioavailable degrader of mutant BRAF for the treatment of BRAF-driven cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3425.
- Published
- 2023
- Full Text
- View/download PDF
6. Abstract 2158: Preclinical evaluation of CFT1946 as a selective degrader of mutant BRAF for the treatment of BRAF driven cancers
- Author
-
Mathew E. Sowa, Bridget Kreger, Joelle Baddour, Yanke Liang, Jeffrey R. Simard, Laura Poling, Ping Li, Robert Yu, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe Sahil Patel, Richard Deibler, Kyle S. Cole, Scott Eron, David Cocozziello, Fazlur Rahman, Moses Moustakim, Christopher G. Nasveschuk, Katrina L. Jackson, Mark Fitzgerald, Victoria Garza, Morgan O’Shea, Gesine Veits, Jeremy L. Yap, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Stewart L. Fisher, and Roy M. Pollock
- Subjects
Cancer Research ,Oncology - Abstract
The BRAF kinase is a critical node in the MAPK signaling pathway and is mutated in approximately 8% of human cancers including melanoma (~60%), thyroid (~60%), and lung adenocarcinoma (~10%). The most common mutation in BRAF is V600E (Class I), occurring in half of malignant melanomas. This mutation hyperactivates ERK and signals as a RAF inhibitor-sensitive monomer. BRAF inhibitors including vemurafenib, dabrafenib and encorafenib have produced impressive responses in V600X patients, however resistance usually emerges within a year, including RAS mutation, BRAFV600E amplification, and BRAFV600E intragenic deletion or splice variants. These inhibitors are also ineffective against non-V600 BRAF mutants (Class II & III). To address some of these limitations we have developed CFT1946, a bifunctional degradation activating compound (BiDAC™) degrader comprising a BRAF kinase domain targeting ligand linked to a cereblon ligand. CFT1946 is capable of degrading BRAF V600E (Class I), G469A (Class II), G466V (Class III) mutations, and the p61-BRAFV600E splice variant while maintaining exquisite selectivity against the proteome including WT BRAF and CRAF. In A375 cells, CFT1946 potently degraded BRAFV600E (Emax = 26%; DC50 = 14nM at 24hr) and, inhibited ERK phosphorylation (IC50 = 11nM at 24hr) and cell growth (GI50 = 94nM at 96hr) while having no effect in the mutant KRAS driven cell line HCT116. In A375 xenografts, oral delivery of CFT1946 resulted in deeper tumor regressions when dosed at 10 mg/kg PO BID and compared favorably to a clinically relevant dose of encorafenib. We further evaluated CFT1946 in an engineered A375-BRAFV600E/NRASQ61K double mutant model of BRAF inhibitor resistance. CFT1946 was able to degrade BRAFV600E in these cells and was much more effective than encorafenib at inhibiting viability in vitro. In this model, in vivo dosing of single agent CFT1946 caused robust tumor growth inhibition and combination with the MEK inhibitor, trametinib, resulted in tumor regressions. The combination of encorafenib and trametinib showed no activity in the same model. Next, we demonstrated that CFT1946 was able to degrade additional BRAF mutant proteins including G469A (Class II), G466V (Class III), and the p61-BRAFV600E splice variant using heterologous expression in HEK293T cells. Additionally, we also showed that CFT1946, but not encorafenib, inhibited proliferation of the BRAFG466V heterozygous lung tumor cell line H1666. Based on its activity in preclinical models, including models of BRAF inhibitor resistance, and its drug-like properties we are progressing CFT1946 as a candidate for clinical development in patients with solid tumors bearing BRAF V600X mutations. Further, given CFT1946’s activity on non-V600 BRAF mutations, we are continuing to explore CFT1946 and related BiDAC degraders as therapeutic options for patients bearing Class II or Class III BRAF mutations. Citation Format: Mathew E. Sowa, Bridget Kreger, Joelle Baddour, Yanke Liang, Jeffrey R. Simard, Laura Poling, Ping Li, Robert Yu, Ashley Hart, Roman V. Agafonov, Grace Sarkissian, Joe Sahil Patel, Richard Deibler, Kyle S. Cole, Scott Eron, David Cocozziello, Fazlur Rahman, Moses Moustakim, Christopher G. Nasveschuk, Katrina L. Jackson, Mark Fitzgerald, Victoria Garza, Morgan O’Shea, Gesine Veits, Jeremy L. Yap, Andrew J. Phillips, Elizabeth Norton, Adam S. Crystal, Stewart L. Fisher, Roy M. Pollock. Preclinical evaluation of CFT1946 as a selective degrader of mutant BRAF for the treatment of BRAF driven cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2158.
- Published
- 2022
- Full Text
- View/download PDF
7. Development of an AchillesTAG degradation system and its application to control CAR-T activity
- Author
-
Joe Sahil Patel, Eunice S. Park, Chi-Li Chen, Andrew J. K. Phillips, Linda Lee, Thomas G. Scott, Christina S. Henderson, Ashley A. Hart, David A. Proia, James A. Henderson, W. Austin Elam, Catherine S. Oakes, Trang N. Tieu, Joelle Baddour, Mark C. Norley, Mathew E. Sowa, Jessica Freda, Marius S. Pop, Brendon Ladd, Minsheng He, Christopher G. Nasveschuk, Harit U. Vora, Gunther Kern, Gesine Kerstin Veits, Abigail Vogelaar, Stewart L. Fisher, Richard W. Deibler, Rhamy Zeid, Mark W. Carlson, Roman V. Agafonov, Arushi Jain, Graham P. Marsh, Prasoon Chaturvedi, Hannah J. Maple, Scott J. Eron, and Emily S. Kibbler
- Subjects
Chemistry ,Drug discovery ,Chemical biology ,Medicinal chemistry ,Computational biology ,QD415-436 ,Protein degradation ,Biochemistry ,Target validation ,CAR PROTEIN ,Pharmacology (medical) ,Target protein ,Car t cells ,Targeted protein degradation ,aTAG - Abstract
In addition to the therapeutic applicability of targeted protein degradation (TPD), the modality also harbors unique properties that enable the development of innovative chemical biology tools to interrogate complex biology. TPD offers an all-chemical strategy capable of the potent, durable, selective, reversible, and time-resolved control of the levels of a given target protein in both in vitro and in vivo contexts. These properties are particularly well-suited for enabling the precise perturbation of a given gene to understand its biology, identify dependencies/vulnerabilities in disease contexts, and as a strategy to control gene therapies. To leverage these elegant properties, we developed the AchillesTag (aTAG) degradation system to serve as a tool in target identification and validation efforts. The aTAG degradation system provides a novel degradation tag based on the MTH1 protein paired with three fully validated bifunctional degraders with both in vitro and in vivo applicability. We catalog the development of the aTAG system from selection and validation of the novel MTH1 aTAG, alongside a comprehensive SAR campaign to identify high performing tool degraders. To demonstrate the utility of the aTAG system to dissect a complex biological system, we apply the technology to the control of Chimeric Antigen Receptor (CAR) activity. Using aTAG, we demonstrate the ability to potently and selectively control CAR protein levels, resulting in the exquisite rheostat control of CAR mediated T-cell activity. Furthermore, we showcase the in vivo application of the system via degradation of the aTAG-fused CAR protein in a human xenograft model. The aTAG degradation system provides a complete chemical biology tool to aid foundational target validation efforts that inspire drug discovery campaigns towards therapeutic applicability.
- Published
- 2021
8. Design of BET Inhibitor Prodrugs with Superior Efficacy and Devoid of Systemic Toxicities
- Author
-
Jeremiah A. Johnson, DeborahJ. C. Ehrlich, MatthewR. Golder, Allison M. Neenan, Peter Blume-Jensen, James C. Ackley, William K. Dahlberg, Kyriakos D. Economides, Jennifer K. Saucier-Sawyer, Lawrence A. Reiter, Paul W. Kopesky, Elisa de Stanchina, Olga Burenkova, David J. Turnquist, Jannik N. Andersen, Sattanathan Paramasivan, Jenny Liu, Bhavatarini Vangamudi, Sung Jin Huh, Nolan M. Gallagher, Samantha W. Brady, Michael J. Jessel, Donald E. Chickering, Peyton Shieh, Farrukh Vohidov, Joelle Baddour, Julie Kim, Michail Shipitsin, Hung V.-T. Nguyen, and Gaurab Kc
- Subjects
BET inhibitor ,Therapeutic index ,Drug development ,Tolerability ,In vivo ,business.industry ,medicine.medical_treatment ,medicine ,Prodrug ,Pharmacology ,business ,Targeted therapy ,Bromodomain - Abstract
Prodrugs engineered for preferential activation in diseased versus normal tissues offer immense potential to improve the therapeutic index of preclinical and clinical-stage active pharmaceutical ingredients that either cannot be developed otherwise or whose efficacy or tolerability it is highly desirable to improve. Such approaches, however, often suffer from trial-and-error design, precluding predictive design and optimization. Here, using BET bromodomain inhibitors (BETi)—a class of epigenetic regulators with proven anti-cancer activity but clinical development hindered by systemic adverse effects–– we introduce a platform that overcomes these challenges. Through tuning of traceless linkers appended to a “brush prodrug” scaffold, we demonstrate that it is possible to correlate in vitro prodrug activation kinetics with in vivo tumor pharmacokinetics, leading to novel BETi prodrugs with enhanced anti-tumor efficacy and devoid of dose-limiting toxicities. This work has immediate clinical implications, introducing principles for the predictive design of prodrugs and potentially overcoming hurdles in drug development.
- Published
- 2020
- Full Text
- View/download PDF
9. Lactate-mediated epigenetic reprogramming regulates formation of human pancreatic cancer-associated fibroblasts
- Author
-
Orsolya Giricz, Deepak Nagrath, Yiyu Zou, Beamon Agarwal, John M. Greally, Matthias Bartenstein, Paola A. Guerrero, Srabani Sahu, Anirban Maitra, Srinivas Aluri, Changsoo Kwak, Tushar D. Bhagat, Shanisha Gordon-Mitchell, Brijesh Patel, Hongyun Zhao, Michael Goggins, Debabrata Banerjee, Davendra Sohal, Masako Suzuki, Lifeng Yang, Dagny Von Ahrens, Joelle Baddour, Sonal Gupta, Sanchari Bhattacharyya, Surinder K. Batra, Amit Verma, Gaurav Choudhary, Meelad M. Dawlaty, Ulrich Steidl, Prafulla Bhagat, Abhinav Achreja, Kith Pradhan, and Yiting Yu
- Subjects
0301 basic medicine ,Mouse ,Epigenesis, Genetic ,Mice ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,pancreas ,Biology (General) ,Epigenomics ,Cancer Biology ,biology ,Chemistry ,General Neuroscience ,General Medicine ,Cellular Reprogramming ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,DNA methylation ,5-Methylcytosine ,Medicine ,Ketoglutaric Acids ,Reprogramming ,Carcinoma, Pancreatic Ductal ,Research Article ,Receptors, CXCR4 ,QH301-705.5 ,Science ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,stroma ,Animals ,Humans ,Neoplasm Invasiveness ,Epigenetics ,Lactic Acid ,Cell Proliferation ,General Immunology and Microbiology ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,DNA Methylation ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,biology.protein ,Cancer research ,Demethylase ,methylation ,Stromal Cells ,Transcriptome - Abstract
Even though pancreatic ductal adenocarcinoma (PDAC) is associated with fibrotic stroma, the molecular pathways regulating the formation of cancer associated fibroblasts (CAFs) are not well elucidated. An epigenomic analysis of patient-derived and de-novo generated CAFs demonstrated widespread loss of cytosine methylation that was associated with overexpression of various inflammatory transcripts including CXCR4. Co-culture of neoplastic cells with CAFs led to increased invasiveness that was abrogated by inhibition of CXCR4. Metabolite tracing revealed that lactate produced by neoplastic cells leads to increased production of alpha-ketoglutarate (aKG) within mesenchymal stem cells (MSCs). In turn, aKG mediated activation of the demethylase TET enzyme led to decreased cytosine methylation and increased hydroxymethylation during de novo differentiation of MSCs to CAF. Co-injection of neoplastic cells with TET-deficient MSCs inhibited tumor growth in vivo. Thus, in PDAC, a tumor-mediated lactate flux is associated with widespread epigenomic reprogramming that is seen during CAF formation.
- Published
- 2019
10. Reduction of liver fibrosis by rationally designed macromolecular telmisartan prodrugs
- Author
-
Sattanathan Paramasivan, Kyriakos D. Economides, Deborah C. Ehrlich, Eric J. Held, Joelle Baddour, Peter Blume-Jensen, Matthew R. Golder, Allison M. Neenan, Jeremiah A. Johnson, Samantha W. Brady, James C. Ackley, Jennifer K. Saucier-Sawyer, Bhavatarini Vangamudi, Farrukh Vohidov, Paul W. Kopesky, Sung Jin Huh, Hung V.-T. Nguyen, Donald E. Chickering, Michail Shipitsin, Jannik N. Andersen, Jenny Liu, and Lawrence A. Reiter
- Subjects
0301 basic medicine ,Liver Cirrhosis ,Male ,medicine.medical_specialty ,medicine.drug_class ,Polymers ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Pharmacology ,010402 general chemistry ,01 natural sciences ,Article ,Rats, Sprague-Dawley ,03 medical and health sciences ,Mice ,Medicine ,Animals ,Prodrugs ,Telmisartan ,Antihypertensive drug ,Carbon Tetrachloride ,chemistry.chemical_classification ,Mice, Inbred BALB C ,business.industry ,Prodrug ,Molecular medicine ,3. Good health ,0104 chemical sciences ,Computer Science Applications ,Rats ,Disease Models, Animal ,030104 developmental biology ,Enzyme ,chemistry ,Gene Expression Regulation ,Liver ,Drug Design ,Drug delivery ,Histopathology ,Female ,Hepatic fibrosis ,business ,Angiotensin II Type 1 Receptor Blockers ,Biotechnology ,medicine.drug ,Half-Life - Abstract
At present there are no drugs for the treatment of chronic liver fibrosis that have been approved by the Food and Drug Administration of the United States. Telmisartan, a small-molecule antihypertensive drug, displays antifibrotic activity, but its clinical use is limited because it causes systemic hypotension. Here, we report the scalable and convergent synthesis of macromolecular telmisartan prodrugs optimized for preferential release in diseased liver tissue. We have optimized the release of active telmisartan in fibrotic liver to be depot-like (that is, a constant therapeutic concentration) through the molecular design of telmisartan brush-arm star polymers, and show that these lead to improved efficacy and to the avoidance of dose-limiting hypotension in both metabolically and chemically induced mouse models of hepatic fibrosis, as determined by histopathology, enzyme levels in the liver, intact-tissue protein markers, hepatocyte necrosis protection and gene-expression analyses. In rats and dogs, the prodrugs are retained long term in liver tissue, and have a well-tolerated safety profile. Our findings support the further development of telmisartan prodrugs that enable infrequent dosing in the treatment of liver fibrosis.
- Published
- 2019
11. Eye tissue regeneration and engineering
- Author
-
Joelle Baddour, Konstantinos Sousounis, and Panagiotis A. Tsonis
- Subjects
Standard of care ,genetic structures ,business.industry ,Corneal Diseases ,Cell degeneration ,eye diseases ,Transplantation ,medicine.anatomical_structure ,Intraocular lenses ,Everyday tasks ,medicine ,Human eye ,sense organs ,Stem cell ,business ,Neuroscience - Abstract
Vision is a major driver of our everyday tasks. The human eye is composed of different specialized tissues that facilitate sight. Dysregulation of these homeostatic mechanisms can lead to eye diseases that are usually detrimental to the patient’s quality of life. Regenerative engineering is utilizing cutting edge technology to restore the lost function of the faulty components of the human eye. In this chapter, the current methods used to treat eye diseases will be briefly discussed and the shortcomings of the current standard of care requiring novel intervention will also be addressed. The topics covered in this chapter include in vitro expansion and transplantation of corneal endothelial cells and limbal stem cells for the treatment of corneal diseases, intraocular lenses for cataract treatment, and subretinal microchips for the treatment of photoreceptor or ganglion cell degeneration. We also touch upon the subject of 3D printing and how the intrinsic regenerative capabilities of certain organisms such as fishes and amphibians can shape the future of regenerative engineering.
- Published
- 2017
- Full Text
- View/download PDF
12. Energy stress-induced lncRNA FILNC1 represses c-Myc-mediated energy metabolism and inhibits renal tumor development
- Author
-
Xifeng Wu, Hyemin Lee, Li Zhuang, Zhen Dong Xiao, Han Liang, Christopher G. Wood, Deepak Nagrath, Joelle Baddour, Yilei Zhang, Jian Gu, Boyi Gan, and Leng Han
- Subjects
0301 basic medicine ,Genes, myc ,General Physics and Astronomy ,urologic and male genital diseases ,Kidney ,0302 clinical medicine ,Heterogeneous-Nuclear Ribonucleoprotein D ,lcsh:Science ,Multidisciplinary ,female genital diseases and pregnancy complications ,Kidney Neoplasms ,Up-Regulation ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,RNA, Long Noncoding ,hormones, hormone substitutes, and hormone antagonists ,medicine.medical_specialty ,Science ,Down-Regulation ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Downregulation and upregulation ,Stress, Physiological ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Heterogeneous Nuclear Ribonucleoprotein D0 ,Lactic Acid ,Transcription factor ,Carcinoma, Renal Cell ,Messenger RNA ,fungi ,Kidney metabolism ,RNA ,Cancer ,General Chemistry ,medicine.disease ,030104 developmental biology ,Endocrinology ,Glucose ,HEK293 Cells ,Cancer cell ,Cancer research ,lcsh:Q ,Energy Metabolism - Abstract
The roles of long non-coding RNAs in cancer metabolism remain largely unexplored. Here we identify FILNC1 (FoxO-induced long non-coding RNA 1) as an energy stress-induced long non-coding RNA by FoxO transcription factors. FILNC1 deficiency in renal cancer cells alleviates energy stress-induced apoptosis and markedly promotes renal tumor development. We show that FILNC1 deficiency leads to enhanced glucose uptake and lactate production through upregulation of c-Myc. Upon energy stress, FILNC1 interacts with AUF1, a c-Myc mRNA-binding protein, and sequesters AUF1 from binding c-Myc mRNA, leading to downregulation of c-Myc protein. FILNC1 is specifically expressed in kidney, and is downregulated in renal cell carcinoma; also, its low expression correlates with poor clinical outcomes in renal cell carcinoma. Together, our study not only identifies FILNC1 as a negative regulator of renal cancer with potential clinical value, but also reveals a regulatory mechanism by long non-coding RNAs to control energy metabolism and tumor development., FoxO are commonly down-regulated transcription factors and tumor suppressors in renal cell cancer (RCC). Here, the authors show that upon energy stress FoxOs induce the expression of the long non-coding RNA FILNC1, which inhibits survival of RCC by downregulating c-Myc and c-Myc-dependent metabolic rewiring.
- Published
- 2016
13. Genomic deletion of malic enzyme 2 confers collateral lethality in pancreatic cancer
- Author
-
Huamin Wang, Jason B. Fleming, Y. Alan Wang, Tony Gutschner, Andrea Viale, Alina Chen, Anirban Maitra, Wen Ting Liao, Lifeng Yang, Di Zhao, Nikunj Satani, Florian L. Muller, Ya'an Kang, Giannicola Genovese, Giulio Draetta, Edward F. Chang, Zangdao Lan, Deepak Nagrath, Haoqiang Ying, Joelle Baddour, Ronald A. DePinho, Prasenjit Dey, Chia Chin Wu, Jiyoon Lee, and Abhinav Achreja
- Subjects
0301 basic medicine ,Male ,AMP-Activated Protein Kinases ,Pregnancy Proteins ,Mice ,AMP-activated protein kinase ,Malate Dehydrogenase ,2.1 Biological and endogenous factors ,NADPH regeneration ,Amino Acids ,Aetiology ,Cancer ,chemistry.chemical_classification ,Multidisciplinary ,Ovarian Cancer ,Mitochondria ,Biochemistry ,Pancreatic Ductal ,5.1 Pharmaceuticals ,Ketoglutaric Acids ,Development of treatments and therapeutic interventions ,Sterol Regulatory Element Binding Protein 1 ,Biotechnology ,Carcinoma, Pancreatic Ductal ,Gene isoform ,General Science & Technology ,Malic enzyme ,Oxidative phosphorylation ,Biology ,Article ,Minor Histocompatibility Antigens ,03 medical and health sciences ,Pancreatic Cancer ,Rare Diseases ,Animals ,Humans ,Gene ,Transaminases ,Carcinoma ,Branched-Chain ,Sterol regulatory element-binding protein ,Pancreatic Neoplasms ,030104 developmental biology ,Enzyme ,chemistry ,biology.protein ,Biocatalysis ,Digestive Diseases ,Reactive Oxygen Species ,Amino Acids, Branched-Chain ,Gene Deletion ,NADP - Abstract
The genome of pancreatic ductal adenocarcinoma (PDAC) frequently contains deletions of tumour suppressor gene loci, most notably SMAD4, which is homozygously deleted in nearly one-third of cases. As loss of neighbouring housekeeping genes can confer collateral lethality, we sought to determine whether loss of the metabolic gene malic enzyme 2 (ME2) in the SMAD4 locus would create cancer-specific metabolic vulnerability upon targeting of its paralogous isoform ME3. The mitochondrial malic enzymes (ME2 and ME3) are oxidative decarboxylases that catalyse the conversion of malate to pyruvate and are essential for NADPH regeneration and reactive oxygen species homeostasis. Here we show that ME3 depletion selectively kills ME2-null PDAC cells in a manner consistent with an essential function for ME3 in ME2-null cancer cells. Mechanistically, integrated metabolomic and molecular investigation of cells deficient in mitochondrial malic enzymes revealed diminished NADPH production and consequent high levels of reactive oxygen species. These changes activate AMP activated protein kinase (AMPK), which in turn directly suppresses sterol regulatory element-binding protein 1 (SREBP1)-directed transcription of its direct targets including the BCAT2 branched-chain amino acid transaminase 2) gene. BCAT2 catalyses the transfer of the amino group from branched-chain amino acids to α-ketoglutarate (α-KG) thereby regenerating glutamate, which functions in part to support de novo nucleotide synthesis. Thus, mitochondrial malic enzyme deficiency, which results in impaired NADPH production, provides a prime 'collateral lethality' therapeutic strategy for the treatment of a substantial fraction of patients diagnosed with this intractable disease.
- Published
- 2016
14. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism
- Author
-
Tyler J. Moss, Lifeng Yang, Sonal Gupta, Deepak Nagrath, Joelle Baddour, F. Anthony San Lucas, Vincent Bernard, Hongyun Zhao, Prahlad T. Ram, Anirban Maitra, Sourindra Maiti, Juan C. Marini, Héctor M. Alvarez, Donna M. Peehl, Laurence J.N. Cooper, Abhinav Achreja, Elena G. Seviour, and Thavisha Tudawe
- Subjects
0301 basic medicine ,macropinocytosis ,QH301-705.5 ,Science ,cancer metabolism ,Oxidative phosphorylation ,exosomes ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,metabolic flux analysis ,Neoplasms ,medicine ,Humans ,tumor microenvironment ,Glycolysis ,Biology (General) ,Human Biology and Medicine ,reductive carboxylation ,Tumor microenvironment ,General Immunology and Microbiology ,General Neuroscience ,Pinocytosis ,Cancer ,General Medicine ,Metabolism ,Cell Biology ,medicine.disease ,Microvesicles ,Cell biology ,030104 developmental biology ,Biochemistry ,Cancer cell ,Medicine ,Research Article ,Human - Abstract
Cancer-associated fibroblasts (CAFs) are a major cellular component of tumor microenvironment in most solid cancers. Altered cellular metabolism is a hallmark of cancer, and much of the published literature has focused on neoplastic cell-autonomous processes for these adaptations. We demonstrate that exosomes secreted by patient-derived CAFs can strikingly reprogram the metabolic machinery following their uptake by cancer cells. We find that CAF-derived exosomes (CDEs) inhibit mitochondrial oxidative phosphorylation, thereby increasing glycolysis and glutamine-dependent reductive carboxylation in cancer cells. Through 13C-labeled isotope labeling experiments we elucidate that exosomes supply amino acids to nutrient-deprived cancer cells in a mechanism similar to macropinocytosis, albeit without the previously described dependence on oncogenic-Kras signaling. Using intra-exosomal metabolomics, we provide compelling evidence that CDEs contain intact metabolites, including amino acids, lipids, and TCA-cycle intermediates that are avidly utilized by cancer cells for central carbon metabolism and promoting tumor growth under nutrient deprivation or nutrient stressed conditions. DOI: http://dx.doi.org/10.7554/eLife.10250.001, eLife digest Cancer cells behave differently from healthy cells in many ways. Healthy cells rely on structures called mitochondria to provide them with energy via a process that requires oxygen. However cancer cells don’t rely on this process, and instead release energy by breaking down sugars outside of the mitochondria. This may explain why cancer cells are able to thrive even when little oxygen is available. Cancer cells also interact with neighboring cells called fibroblasts, which are a major part of a tumor’s microenvironment, and recruit them into the tumors. The fibroblasts communicate with cancer cells, in part, by releasing chemical messengers packaged into tiny bubble-like structures called exosomes. Recent studies have suggested that these exosomes may help cancer cells to thrive, but there are many questions remaining about how they might do this. Now, Zhao et al. show that the fibroblasts smuggle essential nutrients to cancer cells via the exosomes and disable oxygen-based energy production in cancer cells. First, exosomes released by cancer-associated fibroblasts from people with prostate cancer were collected and marked with a green dye. Next, the green-labeled exosomes were mixed with prostate cancer cells, and shown to be absorbed by the cells. Oxygen-based energy release was dramatically reduced in the exosome-absorbing cells, and sugar-based energy release increased. Next, Zhao et al examined the contents of the exosomes, and found that they contain the building blocks of proteins, fats, and other important molecules. Next, the experiments revealed that both prostate cancer and pancreatic cancer cells deprived of nutrients can use these smuggled resources to continue to grow. Importantly, this process did not involve the protein Kras, which previous studies had show helps cancer cells absorb nutrients. These findings suggest that preventing exosomes from smuggling resources to starving cancer cells might be an effective strategy to treat cancers. DOI: http://dx.doi.org/10.7554/eLife.10250.002
- Published
- 2016
15. Organ repair and regeneration: An overview
- Author
-
Panagiotis A. Tsonis, Joelle Baddour, and Konstantinos Sousounis
- Subjects
Embryology ,Pathology ,medicine.medical_specialty ,Decellularization ,Tissue Engineering ,Stem Cells ,Regeneration (biology) ,General Medicine ,Biology ,Regenerative Medicine ,Embryonic stem cell ,Regenerative medicine ,Organ Specificity ,Models, Animal ,medicine ,Animals ,Humans ,Regeneration ,Stem cell ,Progenitor cell ,Induced pluripotent stem cell ,Stem Cell Transplantation ,Developmental Biology - Abstract
A number of organs have the intrinsic ability to regenerate, a distinctive feature that varies among organisms. Organ regeneration is a process not fully yet understood. However, when its underlying mechanisms are unraveled, it holds tremendous therapeutic potential for humans. In this review, we chose to summarize the repair and regenerative potential of the following organs and organ systems: thymus, adrenal gland, thyroid gland, intestine, lungs, heart, liver, blood vessels, germ cells, nervous system, eye tissues, hair cells, kidney and bladder, skin, hair follicles, pancreas, bone, and cartilage. For each organ, a review of the following is presented: (a) factors, pathways, and cells that are involved in the organ's intrinsic regenerative ability, (b) contribution of exogenous cells - such as progenitor cells, embryonic stem cells, induced pluripotent stem cells, and bone marrow-, adipose- and umbilical cord blood-derived stem cells - in repairing and regenerating organs in the absence of an innate intrinsic regenerative capability, (c) and the progress made in engineering bio-artificial scaffolds, tissues, and organs. Organ regeneration is a promising therapy that can alleviate humans from diseases that have not been yet cured. It is also superior to already existing treatments that utilize exogenous sources to substitute for the organ's lost structure and/or function(s).
- Published
- 2012
- Full Text
- View/download PDF
16. Publisher Correction: Reduction of liver fibrosis by rationally designed macromolecular telmisartan prodrugs
- Author
-
Sattanathan Paramasivan, Eric J. Held, Hung V.-T. Nguyen, Jennifer K. Saucier-Sawyer, James C. Ackley, Donald E. Chickering, Jeremiah A. Johnson, Matthew R. Golder, Deborah C. Ehrlich, Paul W. Kopesky, Peter Blume-Jensen, Joelle Baddour, Jannik N. Andersen, Sung Jin Huh, Michail Shipitsin, Allison M. Neenan, Jenny Liu, Lawrence A. Reiter, Samantha W. Brady, Farrukh Vohidov, Bhavatarini Vangamudi, and Kyriakos D. Economides
- Subjects
Statement (logic) ,Published Erratum ,Liver fibrosis ,Philosophy ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,02 engineering and technology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Linguistics ,0104 chemical sciences ,Computer Science Applications ,0210 nano-technology ,Biotechnology - Abstract
In the version of this Article originally published, the author Peter Blume-Jensen was not denoted as a corresponding author; this has now been amended and the author’s email address has been added. The ‘Correspondence and requests for materials’ statement was similarly affected and has now been updated with the author’s initials ‘P.B-J.’
- Published
- 2018
- Full Text
- View/download PDF
17. Abstract 777: Malic enzyme 3 as a collateral lethality target in pancreatic cancer
- Author
-
Youngsoo Kim, Ronald A. DePinho, Deepak Nagrath, Joelle Baddour, Florian L. Muller, Chia Chin Wu, Andrea Viale, Anirban Maitra, Huamin Wang, Robert Macloed, Prasenjit Dey, Alan Wang, Giulio Draetta, and Haoqiang Ying
- Subjects
Cancer Research ,biology ,medicine.disease ,Sterol regulatory element-binding protein ,Small hairpin RNA ,Oncology ,AMP-activated protein kinase ,Apoptosis ,Pancreatic cancer ,Cancer cell ,Cancer research ,biology.protein ,medicine ,Null cell ,Gene - Abstract
Cancer genomes possess many deletion events targeting tumor suppressor genes (TSG) and neighboring genes in these loci. These deletion patterns prompted us to consider a systematic approach, termed “collateral lethality”, designed to identify cancer-specific vulnerabilities resulting from the deletion of neighboring genes. These bystander genes do not appear to be involved in cancer pathogenesis, yet encode cell-essential functions and are members of multi-gene families that are functionally redundant and co-expressed. Homozygous deletion of SMAD4 is a frequent event in pancreas cancer and other cancer types, totaling >30,000 cases in US annually. SMAD4 deletion often results in co-deletion of the neighboring mitochondrial malic enzyme 2 (ME2) gene. In mammalian cells, two genes (ME2 and ME3) encode redundant cell-essential mitochondrial ME activity. Together, ME2 and ME3 function to generate pyruvate to fuel the TCA cycle, and NADPH to maintain ROS homeostasis. These observations prompted us to hypothesize that the genetic or pharmacological extinction of ME3 activity in a ME2 null cell would specifically compromise cancer cells yet be tolerated in normal host cells possessing ME2 activity. Inducible shRNA strategies were employed to genetically deplete ME3 in ME2-null versus ME2-intact cells followed by apoptosis measurements, integrated metabolomics, and molecular investigations. In collaboration with IONIS Pharmaceuticals, we tested and validated anti-sense oligonucleotide (ASO) to target ME3. Genetic depletion of ME3 in ME2 null, but not ME2 intact, cells resulted in apoptosis and blocked tumorigenic potential. Mechanistically, integrated metabolomic and molecular investigation of mitochondrial ME-deficient cells revealed diminished NADPH production and consequent high ROS, which activates AMP activated protein kinase (AMPK) and which in turn directly suppresses sterol regulatory element-binding protein 1 (SREBP1)-directed transcription of its direct targets including the BCAT2 (Branched chain amino acid transaminase 2) gene. We also determined that mitochondrial MEs regulate the utilization of branched chain amino acid (BCAA) via BCAT2, a transaminase required for BCAA catabolism. Notably, enforced expression of BCAT2 can restore tumorigenic potential of ME2/3 deficiency, and free nucleotides can restore proliferation in cell culture. Specific targeting of ME3 using ASOs could dramatically reduce tumor burden in a subQ tumor model of pancreatic cancer. Thus, a key mechanism driving cancer cell lethality involves BCAAs as crucial metabolites under the critical regulation of the mitochondrial MEs. These studies reveal a collateral lethal vulnerability in pancreas and other cancers that can be targeted pharmacologically in genotype-defined patient populations. We propose that highly specific ME3 inhibitors could provide an effective therapy across a substantial number of cancer patients. Citation Format: Prasenjit Dey, Joelle Baddour, Youngsoo Kim, Robert Macloed, Florian Muller, Chia Chin Wu, Huamin Wang, Andrea Viale, Haoqiang Ying, Giulio Draetta, Anirban Maitra, Alan Wang, Deepak Nagrath, Ronald DePinho. Malic enzyme 3 as a collateral lethality target in pancreatic cancer [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 777.
- Published
- 2018
- Full Text
- View/download PDF
18. Author response: Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism
- Author
-
Laurence J.N. Cooper, Deepak Nagrath, Joelle Baddour, Donna M. Peehl, Sourindra Maiti, F. Anthony San Lucas, Héctor M. Alvarez, Hongyun Zhao, Prahlad T. Ram, Abhinav Achreja, Elena G. Seviour, Thavisha Tudawe, Sonal Gupta, Anirban Maitra, Tyler J. Moss, Vincent Bernard, Lifeng Yang, and Juan C. Marini
- Subjects
Tumor microenvironment ,Cancer cell ,Cancer research ,Metabolism ,Biology ,Microvesicles - Published
- 2015
- Full Text
- View/download PDF
19. HSulf-1 deficiency dictates a metabolic reprograming of glycolysis and TCA cycle in ovarian cancer
- Author
-
Debarshi Roy, Katherine Stilles, Val J. Lowe, Thomas Dierks, Ashwani Khurana, Samuel Leung, Susmita Mondal, Viji Shridhar, Edward Hammond, Steve E. Kalloger, Juliana Camacho-Pereira, Keith Dredge, Seth Padmabandu, Deepak Nagrath, Joelle Baddour, Blake Gilks, Lifeng Yang, and Eduardo N. Chini
- Subjects
PG545 ,Glucose uptake ,Citric Acid Cycle ,Biology ,Mice ,Cell Line, Tumor ,Animals ,Humans ,Glycolysis ,Cell Proliferation ,Mice, Knockout ,Ovarian Neoplasms ,HSulf-1 ,Pyruvate dehydrogenase complex ,Microarray Analysis ,Warburg effect ,3. Good health ,Citric acid cycle ,HB-EGF ,ovarian cancer ,c-Myc ,Oncology ,Biochemistry ,Anaerobic glycolysis ,biology.protein ,Phosphorylation ,GLUT1 ,Female ,Sulfotransferases ,Signal Transduction ,Research Paper - Abstract
Warburg effect has emerged as a potential hallmark of many cancers. However, the molecular mechanisms that led to this metabolic state of aerobic glycolysis, particularly in ovarian cancer (OVCA) have not been completely elucidated. HSulf-1 predominantly functions by limiting the bioavailability of heparan binding growth factors and hence their downstream signaling. Here we report that HSulf-1, a known putative tumor suppressor, is a negative regulator of glycolysis. Silencing of HSulf-1 expression in OV202 cell line increased glucose uptake and lactate production by upregulating glycolytic genes such as Glut1, HKII, LDHA, as well as metabolites. Conversely, HSulf-1 overexpression in TOV21G cells resulted in the down regulation of glycolytic enzymes and reduced glycolytic phenotype, supporting the role of HSulf-1 loss in enhanced aerobic glycolysis. HSulf-1 deficiency mediated glycolytic enhancement also resulted in increased inhibitory phosphorylation of pyruvate dehydrogenase (PDH) thus blocking the entry of glucose flux into TCA cycle. Consistent with this, metabolomic and isotope tracer analysis showed reduced glucose flux into TCA cycle. Moreover, HSulf-1 loss is associated with lower oxygen consumption rate (OCR) and impaired mitochondrial function. Mechanistically, lack of HSulf-1 promotes c-Myc induction through HB-EGF-mediated p-ERK activation. Pharmacological inhibition of c-Myc reduced HB-EGF induced glycolytic enzymes implicating a major role of c-Myc in loss of HSulf-1 mediated altered glycolytic pathway in OVCA. Similarly, PG545 treatment, an agent that binds to heparan binding growth factors and sequesters growth factors away from their ligand also blocked HB-EGF signaling and reduced glucose uptake in vivo in HSulf-1 deficient cells.
- Published
- 2015
20. Aging and Regeneration in Vertebrates
- Author
-
Joelle Baddour, Konstantinos Sousounis, and Panagiotis A. Tsonis
- Subjects
DNA damage ,Regeneration (biology) ,Stem cell theory of aging ,medicine ,Anatomy ,Progenitor cell ,Stem cell ,Biology ,medicine.disease_cause ,Tissue homeostasis ,Oxidative stress ,Cell biology ,Telomere - Abstract
Aging is marked by changes that affect organs and resident stem cell function. Shorting of telomeres, DNA damage, oxidative stress, deregulation of genes and proteins, impaired cell-cell communication, and an altered systemic environment cause the eventual demise of cells. At the same time, reparative activities also decline. It is intriguing to correlate aging with the decline of regenerative abilities. Animal models with strong regenerative capabilities imply that aging processes might not be affecting regeneration. In this review, we selectively present age-dependent changes in stem/progenitor cells that are vital for tissue homeostasis and repair. In addition, the aging effect on regeneration following injury in organs such as lung, skeletal muscle, heart, nervous system, cochlear hair, lens, and liver are discussed. These tissues are also known for diseases such as heart attack, stroke, cognitive impairment, cataract, and hearing loss that occur mostly during aging in humans. Conclusively, vertebrate regeneration declines with age with the loss of stem/progenitor cell function. Future studies on improving the function of stem cells, along with studies in fish and amphibians where regeneration does not decline with age, will undoubtedly provide insights into both processes.
- Published
- 2014
- Full Text
- View/download PDF
21. Role of Increased n-acetylaspartate Levels in Cancer
- Author
-
Rajesha Rupaimoole, Guillermo N. Armaiz-Pena, Michael A. Davies, Menashe Bar Eli, Lifeng Yang, Wei Hu, Alpa M. Nick, Behrouz Zand, Christopher McCullough, Gabriel Lopez-Berestein, Lingegowda S. Mangala, Ying Wang, Archana S. Nagaraja, Michele Guindani, Einav Shoshan, Susan K. Lutgendorf, Keith A. Baggerly, Kshipra M. Gharpure, Takashi Mitamura, Deepak Nagrath, Joelle Baddour, Anil K. Sood, Jinsong Liu, Abhinav Achreja, Chad V. Pecot, Heather J. Dalton, Pratip K. Bhattacharya, Rebecca A. Previs, Niki M. Zacharias, Sherry Y. Wu, Cristian Rodriguez-Aguayo, Sunila Pradeep, and Cristina Ivan
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Cell Survival ,Apoptosis ,Kaplan-Meier Estimate ,Biology ,Gene Expression Regulation, Enzymologic ,Article ,Mice ,03 medical and health sciences ,Acetyltransferases ,Tandem Mass Spectrometry ,Uterine cancer ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Chromatography, High Pressure Liquid ,Cell Proliferation ,Ovarian Neoplasms ,Aspartic Acid ,Cell growth ,Melanoma ,Ovary ,medicine.disease ,Cystadenocarcinoma, Serous ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Cancer cell ,Cancer research ,FOXM1 ,Female ,Neoplasm Grading ,Ovarian cancer - Abstract
The clinical and biological effects of metabolic alterations in cancer are not fully understood.In high-grade serous ovarian cancer (HGSOC) samples (n = 101), over 170 metabolites were profiled and compared with normal ovarian tissues (n = 15). To determine NAT8L gene expression across different cancer types, we analyzed the RNA expression of cancer types using RNASeqV2 data available from the open access The Cancer Genome Atlas (TCGA) website (http://www.cbioportal.org/public-portal/). Using NAT8L siRNA, molecular techniques and histological analysis, we determined cancer cell viability, proliferation, apoptosis, and tumor growth in in vitro and in vivo (n = 6-10 mice/group) settings. Data were analyzed with the Student's t test and Kaplan-Meier analysis. Statistical tests were two-sided.Patients with high levels of tumoral NAA and its biosynthetic enzyme, aspartate N-acetyltransferase (NAT8L), had worse overall survival than patients with low levels of NAA and NAT8L. The overall survival duration of patients with higher-than-median NAA levels (3.6 years) was lower than that of patients with lower-than-median NAA levels (5.1 years, P = .03). High NAT8L gene expression in other cancers (melanoma, renal cell, breast, colon, and uterine cancers) was associated with worse overall survival. NAT8L silencing reduced cancer cell viability (HEYA8: control siRNA 90.61% ± 2.53, NAT8L siRNA 39.43% ± 3.00, P.001; A2780: control siRNA 90.59% ± 2.53, NAT8L siRNA 7.44% ± 1.71, P.001) and proliferation (HEYA8: control siRNA 74.83% ± 0.92, NAT8L siRNA 55.70% ± 1.54, P.001; A2780: control siRNA 50.17% ± 4.13, NAT8L siRNA 26.52% ± 3.70, P.001), which was rescued by addition of NAA. In orthotopic mouse models (ovarian cancer and melanoma), NAT8L silencing reduced tumor growth statistically significantly (A2780: control siRNA 0.52 g ± 0.15, NAT8L siRNA 0.08 g ± 0.17, P.001; HEYA8: control siRNA 0.79 g ± 0.42, NAT8L siRNA 0.24 g ± 0.18, P = .008, A375-SM: control siRNA 0.55 g ± 0.22, NAT8L siRNA 0.21 g ± 0.17 g, P = .001). NAT8L silencing downregulated the anti-apoptotic pathway, which was mediated through FOXM1.These findings indicate that the NAA pathway has a prominent role in promoting tumor growth and represents a valuable target for anticancer therapy.Altered energy metabolism is a hallmark of cancer (1). Proliferating cancer cells have much greater metabolic requirements than nonproliferating differentiated cells (2,3). Moreover, altered cancer metabolism elevates unique metabolic intermediates, which can promote cancer survival and progression (4,5). Furthermore, emerging evidence suggests that proliferating cancer cells exploit alternative metabolic pathways to meet their high demand for energy and to accumulate biomass (6-8).
- Published
- 2016
- Full Text
- View/download PDF
22. Abstract 1208: Glutamine modulates cellular NAD+/NADH homeostasis thereby regulating cancer metastasis, drug sensitivity in cancer cells
- Author
-
Tyler J. Moss, Lifeng Yang, Katherine Stilles, Prahlad T. Ram, Deepak Nagrath, Anil K. Sood, Joelle Baddour, Sun Hye Kim, Lisa Chiba, Josh Morse, Juan C. Marini, and Abhinav Achreja
- Subjects
Cancer Research ,Glutaminolysis ,Cancer ,Biology ,medicine.disease ,Glutamine ,Citric acid cycle ,chemistry.chemical_compound ,Oncology ,Biochemistry ,Biosynthesis ,chemistry ,Cancer cell ,Cancer research ,medicine ,NAD+ kinase ,Ovarian cancer - Abstract
Glutamine can play a critical role in cellular growth in multiple cancers. Glutamine-addicted cancer cells are dependent on glutamine for viability, and their metabolism is reprogrammed for glutamine utilization through the tricarboxylic acid (TCA) cycle. Recently, we uncovered a missing link between cancer invasiveness and glutamine dependence. Using isotope tracer and bioenergetic analysis, we found that low-invasive ovarian cancer (OVCA) cells are glutamine independent, whereas high-invasive OVCA cells are markedly glutamine dependent. Consistent with our findings, OVCA patients’ microarray data suggest that glutaminolysis correlates with poor survival. Notably, the ratio of gene expression associated with glutamine anabolism versus catabolism has emerged as a novel biomarker for patient prognosis. Significantly, we found that glutamine regulates the cellular NAD+/NADH homeostasis, which mediates cancer metastasis and progression. On the other hand, the overexpression of NAD+ biosynthesis pathway enhances glutamine's entry into TCA cycle for cancer metastasis, as well as chemo-drug resistance. Our findings suggest that a combined approach of targeting high-invasive OVCA cells by blocking glutamine's entry into the TCA cycle, along with targeting NAD+ biosynthesis pathway may lead to potential therapeutic approaches for treating OVCAs. Our insights will present a unique opportunity for overcoming the drug resistance limitation in clinical trials in ovarian cancers. Citation Format: Lifeng Yang, Abhinav Achreja, Tyler Moss, Joelle Baddour, Katherine Stilles, Lisa Chiba, Sun Hye Kim, Josh Morse, Juan Marini, Anil K. Sood, Prahlad T. Ram, Deepak Nagrath. Glutamine modulates cellular NAD+/NADH homeostasis thereby regulating cancer metastasis, drug sensitivity in cancer cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1208. doi:10.1158/1538-7445.AM2015-1208
- Published
- 2015
- Full Text
- View/download PDF
23. Abstract 2913: Transcriptional reprogramming of pancreatic stroma induces metabolic changes in pancreatic tumor cells
- Author
-
Deepak Nagrath, Joelle Baddour, Janusz Franco-Barraza, Anirban Maitra, Juan C. Marini, Radina Khalid, Chaoxin Hu, Lifeng Yang, Rebecca N. Curtis, Abhinav Achreja, Pari Shah, Thomas Plackemeier, Seth Padmabandu, and Edna Cukierman
- Subjects
Cancer Research ,Tumor microenvironment ,Stromal cell ,Cancer ,Biology ,medicine.disease ,Oncology ,Stroma ,Pancreatic tumor ,Pancreatic cancer ,Cancer cell ,medicine ,Cancer research ,Reprogramming - Abstract
Pancreatic cancer remains one of the most lethal of all solid tumors largely due to an aggressive stroma that constitutes the bulk of the tumor. The notion of a reprogrammed stroma suggests the re-establishment of a physiologically normal tumor microenvironment with quiescent stellate cells and fibroblasts. The induction of a quiescent phenotype hinders the aberrant tumor-stroma crosstalk and enables the increased intratumoral delivery of chemo drugs. A combinatorial transcriptional therapy has proven successful at normalizing the tumor microenvironment by reverting the activated state of stromal cells back to quiescence, both at the phenotypic and genetic levels. Indeed, we have observed a reduction in the activation markers of primary human stellate cells and fibroblasts, accompanied by an increase in the quiescence markers. Observations from methylation/hydroxymethylation profiling and metabolic rewiring through isotopomer flux analysis revealed potential targets for therapeutic intervention. In addition, the differential metabolic rewiring induced by a quiescent stroma in cancer cells suggests a tumor suppressive environment aimed at weakening an otherwise aggressive cancer. Citation Format: Joelle Baddour, Lifeng Yang, Abhinav Achreja, Seth Padmabandu, Pari Shah, Rebecca N. Curtis, Thomas Plackemeier, Radina Khalid, Juan C. Marini, Janusz Franco-Barraza, Edna Cukierman, Chaoxin Hu, Anirban Maitra, Deepak Nagrath. Transcriptional reprogramming of pancreatic stroma induces metabolic changes in pancreatic tumor cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2913. doi:10.1158/1538-7445.AM2015-2913
- Published
- 2015
- Full Text
- View/download PDF
24. Abstract 3379: Metabolic influences of pancreatic tumor microenvironment on pancreatic cancer cell's metabolism
- Author
-
Juan C. Marini, Anirban Maitra, Janusz Franco-Barraza, Deepak Nagrath, Joelle Baddour, Lifeng Yang, Edna Cukierman, and Chaoxin Hu
- Subjects
Cancer Research ,Tumor microenvironment ,Pathology ,medicine.medical_specialty ,Stromal cell ,Cancer ,Biology ,medicine.disease ,Metastasis ,Oncology ,Stroma ,Pancreatic tumor ,Pancreatic cancer ,medicine ,CA19-9 - Abstract
Pancreatic cancer, the most lethal of solid tumors, is associated with a five-year survival rate and high mortality. The lethality of this tumor stems from lack of early symptoms, inability for detection of cancerous pancreatic lesions, and a diagnosis window that is accompanied by tumor resistance and metastasis. The bulk of the tumor mass, the fibrotic stroma, has been deemed an active player in the initiation and progression of pancreatic ductal adinocarcinoma (PDAC). A number of studies have elucidated the interactions between stromal cells and pancreatic cancer cells (PCCs), and the extracellular matrix (ECM) and PCCs. However, the role of the tumor microenvironment on the metabolic machinery of PCCs remains an active field of investigation. In this study, we investigate the effect of pancreatic stellate cells (PSCs), cancer-associated fibroblasts (CAFs), with or without ECM components, in a two-dimensional or three-dimensional setting on the glycolytic and mitochondrial pathways of patient-derived PCCs. The modulation of the expression levels of metabolic enzymes by the tumor microenvironment was also investigated. The metabolic reprogramming induced in PCCs by normal and reactive pancreatic stroma was investigated by isotopomer flux analysis. Our results reveal that pancreatic reactive stroma differentially upregulates glutamine and arginine metabolism in PCCs. The insights obtained from our work will lead to the development of targeted therapies for stroma and pancreatic cancer cells. Citation Format: Joelle Baddour, Lifeng Yang, Juan C. Marini, Janusz Franco-Barraza, Edna Cukierman, Chaoxin Hu, Anirban Maitra, Deepak Nagrath. Metabolic influences of pancreatic tumor microenvironment on pancreatic cancer cell's metabolism. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3379. doi:10.1158/1538-7445.AM2014-3379
- Published
- 2014
- Full Text
- View/download PDF
25. Nitric oxide is a positive regulator of the Warburg effect in ovarian cancer cells
- Author
-
Deepak Nagrath, Joelle Baddour, Julia Win, Lifeng Yang, Christine Caneba, Sean M. Hartig, Rebecca N. Curtis, and Juan C. Marini
- Subjects
Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,Immunology ,Citric Acid Cycle ,Biology ,medicine.disease_cause ,Nitric Oxide ,Cellular and Molecular Neuroscience ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Glycolysis ,Ovarian Neoplasms ,Tumor microenvironment ,Glutaminolysis ,Cell Biology ,medicine.disease ,Warburg effect ,Glutathione ,3. Good health ,Mitochondria ,Endocrinology ,Tumor progression ,Cancer cell ,Cancer research ,Original Article ,Female ,Ovarian cancer ,Carcinogenesis ,Reactive Oxygen Species ,NADP - Abstract
Ovarian cancer (OVCA) is among the most lethal gynecological cancers leading to high mortality rates among women. Increasing evidence indicate that cancer cells undergo metabolic transformation during tumorigenesis and growth through nutrients and growth factors available in tumor microenvironment. This altered metabolic rewiring further enhances tumor progression. Recent studies have begun to unravel the role of amino acids in the tumor microenvironment on the proliferation of cancer cells. One critically important, yet often overlooked, component to tumor growth is the metabolic reprogramming of nitric oxide (NO) pathways in cancer cells. Multiple lines of evidence support the link between NO and tumor growth in some cancers, including pancreas, breast and ovarian. However, the multifaceted role of NO in the metabolism of OVCA is unclear and direct demonstration of NO's role in modulating OVCA cells' metabolism is lacking. This study aims at indentifying the mechanistic links between NO and OVCA metabolism. We uncover a role of NO in modulating OVCA metabolism: NO positively regulates the Warburg effect, which postulates increased glycolysis along with reduced mitochondrial activity under aerobic conditions in cancer cells. Through both NO synthesis inhibition (using L-arginine deprivation, arginine is a substrate for NO synthase (NOS), which catalyzes NO synthesis; using L-Name, a NOS inhibitor) and NO donor (using DETA-NONOate) analysis, we show that NO not only positively regulates tumor growth but also inhibits mitochondrial respiration in OVCA cells, shifting these cells towards glycolysis to maintain their ATP production. Additionally, NO led to an increase in TCA cycle flux and glutaminolysis, suggesting that NO decreases ROS levels by increasing NADPH and glutathione levels. Our results place NO as a central player in the metabolism of OVCA cells. Understanding the effects of NO on cancer cell metabolism can lead to the development of NO targeting drugs for OVCAs.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.