57 results on '"Kaipparettu BA"'
Search Results
2. Abstract P2-02-14: Metabolic regulation and drug resistance in c-Src activated triple negative breast cancer
- Author
-
Jung, KH, primary, Park, JH, additional, Sirupangi, T, additional, Jia, D, additional, Gandhi, N, additional, Pudakalakatti, S, additional, Elswood, J, additional, Porter, W, additional, Putluri, N, additional, Zhang, XH-F, additional, Chen, X, additional, Bhattacharya, PK, additional, Creighton, CJ, additional, Lewis, MT, additional, Rosen, JM, additional, Wong, L-JC, additional, Das, GM, additional, Osborne, CK, additional, Rimawi, MF, additional, and Kaipparettu, BA, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P2-02-11: Combinational treatment of biguanides and fatty acid β-oxidation inhibitor in triple-negative breast cancers
- Author
-
Park, JH, primary, Jung, KH, additional, Vithayathil, S, additional, Jia, D, additional, and Kaipparettu, BA, additional
- Published
- 2019
- Full Text
- View/download PDF
4. Abstract P6-06-04: Targeting replication stress in triple negative breast cancer treatment regimen: An emerging approach
- Author
-
Rajamanickam, S, primary, Park, JH, additional, Bates, K, additional, Timilsina, S, additional, Eedunuri, VK, additional, Onyeagucha, B, additional, Subbarayalu, P, additional, Abdelfattah, N, additional, Jung, KH, additional, Favours, E, additional, Mohammad, TA, additional, Chen, H-IH, additional, Vadlamudi, RK, additional, Chen, Y, additional, Kaipparettu, BA, additional, Arbiser, JL, additional, and Rao, MK, additional
- Published
- 2018
- Full Text
- View/download PDF
5. Abstract P6-01-07: Mitochondria-nuclear communication regulates epithelial-mesenchymal transition and metastasis in triple negative breast cancer
- Author
-
Park, JH, primary, Jung, KH, additional, Sirupangi, T, additional, Vithayathil, S, additional, Jin, F, additional, Putluri, V, additional, Piyarathna, DWB, additional, Yotnda, P, additional, Bhat, VB, additional, Sreekumar, A, additional, Lewis, MT, additional, Coarfa, C, additional, Putluri, N, additional, Creighton, CJ, additional, Wong, L-JC, additional, and Kaipparettu, BA, additional
- Published
- 2017
- Full Text
- View/download PDF
6. Abstract BS3-1: Dominance of mitochondria in determining the malignant phenotype
- Author
-
Kaipparettu, BA, primary
- Published
- 2016
- Full Text
- View/download PDF
7. Abstract P5-03-07: CD24 epigenetic regulation in breast cancer tissues and tumor initiating cells: Promoter specific analysis using next generation sequencing
- Author
-
Park, JH, primary, Cui, H, additional, Vithayathil, SA, additional, Sung, P-L, additional, Zhang, V, additional, Wong, L-JC, additional, and Kaipparettu, BA, additional
- Published
- 2013
- Full Text
- View/download PDF
8. Novel egg white based 3D cell culture system for breast cancer research.
- Author
-
Kaipparettu, BA, primary, Kuiatse, I, additional, Tak-Yee Chan, B, additional, Kaipparettu, MB, additional, Lee, AV, additional, and Oesterreich, S, additional
- Published
- 2009
- Full Text
- View/download PDF
9. Mitochondrial reprogramming by activating OXPHOS via glutamine metabolism in African American patients with bladder cancer.
- Author
-
Kami Reddy KR, Piyarathna DWB, Park JH, Putluri V, Amara CS, Kamal AHM, Xu J, Kraushaar D, Huang S, Jung SY, Eberlin LS, Johnson JR, Kittles RA, Ballester LY, Parsawar K, Siddiqui MM, Gao J, Langer Gramer A, Bollag RJ, Terris MK, Lotan Y, Creighton CJ, Lerner SP, Sreekumar A, Kaipparettu BA, and Putluri N
- Subjects
- Animals, Female, Humans, Male, Mice, Middle Aged, Cell Line, Tumor, Cell Proliferation, Electron Transport Complex I metabolism, Electron Transport Complex I genetics, Metabolomics methods, Black or African American genetics, Glutaminase metabolism, Glutaminase genetics, Glutamine metabolism, Mitochondria metabolism, Oxidative Phosphorylation, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms genetics
- Abstract
Bladder cancer (BLCA) mortality is higher in African American (AA) patients compared with European American (EA) patients, but the molecular mechanism underlying race-specific differences are unknown. To address this gap, we conducted comprehensive RNA-Seq, proteomics, and metabolomics analysis of BLCA tumors from AA and EA. Our findings reveal a distinct metabolic phenotype in AA BLCA characterized by elevated mitochondrial oxidative phosphorylation (OXPHOS), particularly through the activation of complex I. The results provide insight into the complex I activation-driven higher OXPHOS activity resulting in glutamine-mediated metabolic rewiring and increased disease progression, which was also confirmed by [U]13C-glutamine tracing. Mechanistic studies further demonstrate that knockdown of NDUFB8, one of the components of complex I in AA BLCA cells, resulted in reduced basal respiration, ATP production, GLS1 expression, and proliferation. Moreover, preclinical studies demonstrate the therapeutic potential of targeting complex I, as evidenced by decreased tumor growth in NDUFB8-depleted AA BLCA tumors. Additionally, genetic and pharmacological inhibition of GLS1 attenuated mitochondrial respiration rates and tumor growth potential in AA BLCA. Taken together, these findings provide insight into BLCA disparity for targeting GLS1-Complex I for future therapy.
- Published
- 2024
- Full Text
- View/download PDF
10. EpCAM-targeted betulinic acid analogue nanotherapy improves therapeutic efficacy and induces anti-tumorigenic immune response in colorectal cancer tumor microenvironment.
- Author
-
Dutta D, Al Hoque A, Paul B, Park JH, Chowdhury C, Quadir M, Banerjee S, Choudhury A, Laha S, Sepay N, Boro P, Kaipparettu BA, and Mukherjee B
- Subjects
- Animals, Mice, Humans, Nanoparticles chemistry, Cell Line, Tumor, Rats, Colorectal Neoplasms drug therapy, Tumor Microenvironment drug effects, Betulinic Acid, Pentacyclic Triterpenes pharmacology, Epithelial Cell Adhesion Molecule metabolism
- Abstract
Background: Betulinic acid (BA) has been well investigated for its antiproliferative and mitochondrial pathway-mediated apoptosis-inducing effects on various cancers. However, its poor solubility and off-target activity have limited its utility in clinical trials. Additionally, the immune modulatory role of betulinic acid analogue in the tumor microenvironment (TME) is largely unknown. Here, we designed a potential nanotherapy for colorectal cancer (CRC) with a lead betulinic acid analogue, named as 2c, carrying a 1,2,3-triazole-moiety attached to BA through a linker, found more effective than BA for inhibiting CRC cell lines, and was chosen here for this investigation. Epithelial cell adhesion molecule (EpCAM) is highly overexpressed on the CRC cell membrane. A single-stranded short oligonucleotide sequence, aptamer (Apt), that folds into a 3D-defined architecture can be used as a targeting ligand for its specific binding to a target protein. EpCAM targeting aptamer was designed for site-specific homing of aptamer-conjugated-2c-loaded nanoparticles (Apt-2cNP) at the CRC tumor site to enhance therapeutic potential and reduce off-target toxicity in normal cells. We investigated the in vitro and in vivo therapeutic efficacy and anti-tumorigenic immune response of aptamer conjugated nanotherapy in CRC-TME., Methods: After the characterization of nanoengineered aptamer conjugated betulinic acid nanotherapy, we evaluated therapeutic efficacy, tumor targeting efficiency, and anti-tumorigenic immune response using cell-based assays and mouse and rat models., Results: We found that Apt-2cNP improved drug bioavailability, enhanced its biological half-life, improved antiproliferative activity, and minimized off-target cytotoxicity. Importantly, in an in vivo TME, Apt-2cNP showed promising signs of anti-tumorigenic immune response (increased mDC/pDC ratio, enhanced M1 macrophage population, and CD8 T-cells). Furthermore, in vivo upregulation of pro-apoptotic while downregulation of anti-apoptotic genes and significant healing efficacy on cancer tissue histopathology suggest that Apt-2cNP had predominantly greater therapeutic potential than the non-aptamer-conjugated nanoparticles and free drug. Moreover, we observed greater tumor accumulation of the radiolabeled Apt-2cNP by live imaging in the CRC rat model., Conclusions: Enhanced therapeutic efficacy and robust anti-tumorigenic immune response of Apt-2cNP in the CRC-TME are promising indicators of its potential as a prospective therapeutic agent for managing CRC. However, further studies are warranted., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. Retraction: Tobacco-specific Carcinogens Induce Hypermethylation, DNA Adducts, and DNA Damage in Bladder Cancer.
- Author
-
Jin F, Thaiparambil J, Donepudi SR, Vantaku V, Piyarathna DWB, Maity S, Krishnapuram R, Putluri V, Gu F, Purwaha P, Bhowmik SK, Ambati CR, von Rundstedt FC, Roghmann F, Berg S, Noldus J, Rajapakshe K, Gödde D, Roth S, Störkel S, Degener S, Michailidis G, Kaipparettu BA, Karanam B, Terris MK, Kavuri SM, Lerner SP, Kheradmand F, Coarfa C, Sreekumar A, Lotan Y, El-Zein R, and Putluri N
- Published
- 2024
- Full Text
- View/download PDF
12. ESR1 and p53 interactome alteration defines mechanisms of tamoxifen response in luminal breast cancer.
- Author
-
Oturkar CC, Rosario SR, Hutson AD, Groman A, Edge SB, Morrison CD, Swetzig WM, Wang J, Park JH, Kaipparettu BA, Singh PK, Kumar S, Cappuccino HH, Ranjan M, Adjei A, Ghasemi M, Goey AKL, Kulkarni S, and Das GM
- Abstract
The canonical mechanism behind tamoxifen's therapeutic effect on estrogen receptor α/ESR1+ breast cancers is inhibition of ESR1-dependent estrogen signaling. Although ESR1+ tumors expressing wild-type p53 were reported to be more responsive to tamoxifen (Tam) therapy, p53 has not been factored into choice of this therapy and the mechanism underlying the role of p53 in Tam response remains unclear. In a window-of-opportunity trial on patients with newly diagnosed stage I-III ESR1+/HER2/wild-type p53 breast cancer who were randomized to arms with or without Tam prior to surgery, we reveal that the ESR1-p53 interaction in tumors was inhibited by Tam. This resulted in functional reactivation of p53 leading to transcriptional reprogramming that favors tumor-suppressive signaling, as well as downregulation of oncogenic pathways. These findings illustrating the convergence of ESR1 and p53 signaling during Tam therapy enrich mechanistic understanding of the impact of p53 on the response to Tam therapy., Competing Interests: The authors declare no competing interests., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
13. CD24 negativity reprograms mitochondrial metabolism to PPARα and NF-κB-driven fatty acid β-oxidation in triple-negative breast cancer.
- Author
-
Murthy D, Dutta D, Attri KS, Samanta T, Yang S, Jung KH, Latario SG, Putluri V, Huang S, Putluri N, Park JH, and Kaipparettu BA
- Subjects
- Humans, PPAR alpha genetics, Cell Line, Tumor, Fatty Acids metabolism, CD24 Antigen genetics, CD24 Antigen metabolism, NF-kappa B, Triple Negative Breast Neoplasms pathology
- Abstract
CD24 is a well-characterized breast cancer (BC) stem cell (BCSC) marker. Primary breast tumor cells having CD24-negativity together with CD44-positivity is known to maintain high metastatic potential. However, the functional role of CD24 gene in triple-negative BC (TNBC), an aggressive subtype of BC, is not well understood. While the significance of CD24 in regulating immune pathways is well recognized in previous studies, the significance of CD24 low expression in onco-signaling and metabolic rewiring is largely unknown. Using CD24 knock-down and over-expression TNBC models, our in vitro and in vivo analysis suggest that CD24 is a tumor suppressor in metastatic TNBC. Comprehensive in silico gene expression analysis of breast tumors followed by lipidomic and metabolomic analyses of CD24-modulated cells revealed that CD24 negativity induces mitochondrial oxidative phosphorylation and reprograms TNBC metabolism toward the fatty acid beta-oxidation (FAO) pathway. CD24 silencing activates PPARα-mediated regulation of FAO in TNBC cells. Further analysis using reverse-phase protein array and its validation using CD24-modulated TNBC cells and xenograft models nominated CD24-NF-κB-CPT1A signaling pathway as the central regulatory mechanism of CD24-mediated FAO activity. Overall, our study proposes a novel role of CD24 in metabolic reprogramming that can open new avenues for the treatment strategies for patients with metastatic TNBC., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Metabolomic Rewiring Promotes Endocrine Therapy Resistance in Breast Cancer.
- Author
-
Ahn S, Park JH, Grimm SL, Piyarathna DWB, Samanta T, Putluri V, Mezquita D, Fuqua SAW, Putluri N, Coarfa C, and Kaipparettu BA
- Subjects
- Humans, Female, Cell Line, Tumor, Phosphorylation, Signal Transduction, Fatty Acids metabolism, Drug Resistance, Neoplasm genetics, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms metabolism, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- Abstract
Approximately one-third of endocrine-treated women with estrogen receptor alpha-positive (ER+) breast cancers are at risk of recurrence due to intrinsic or acquired resistance. Thus, it is vital to understand the mechanisms underlying endocrine therapy resistance in ER+ breast cancer to improve patient treatment. Mitochondrial fatty acid β-oxidation (FAO) has been shown to be a major metabolic pathway in triple-negative breast cancer (TNBC) that can activate Src signaling. Here, we found metabolic reprogramming that increases FAO in ER+ breast cancer as a mechanism of resistance to endocrine therapy. A metabolically relevant, integrated gene signature was derived from transcriptomic, metabolomic, and lipidomic analyses in TNBC cells following inhibition of the FAO rate-limiting enzyme carnitine palmitoyl transferase 1 (CPT1), and this TNBC-derived signature was significantly associated with endocrine resistance in patients with ER+ breast cancer. Molecular, genetic, and metabolomic experiments identified activation of AMPK-FAO-oxidative phosphorylation (OXPHOS) signaling in endocrine-resistant ER+ breast cancer. CPT1 knockdown or treatment with FAO inhibitors in vitro and in vivo significantly enhanced the response of ER+ breast cancer cells to endocrine therapy. Consistent with the previous findings in TNBC, endocrine therapy-induced FAO activated the Src pathway in ER+ breast cancer. Src inhibitors suppressed the growth of endocrine-resistant tumors, and the efficacy could be further enhanced by metabolic priming with CPT1 inhibition. Collectively, this study developed and applied a TNBC-derived signature to reveal that metabolic reprogramming to FAO activates the Src pathway to drive endocrine resistance in ER+ breast cancer., Significance: Increased fatty acid oxidation induced by endocrine therapy activates Src signaling to promote endocrine resistance in breast cancer, which can be overcome using clinically approved therapies targeting FAO and Src., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
15. Interplay of gut microbiota and host epithelial mitochondrial dysfunction is necessary for the development of spontaneous intestinal inflammation in mice.
- Author
-
Alula KM, Dowdell AS, LeBere B, Lee JS, Levens CL, Kuhn KA, Kaipparettu BA, Thompson WE, Blumberg RS, Colgan SP, and Theiss AL
- Subjects
- Humans, Animals, Mice, Inflammation metabolism, Paneth Cells, Butyrates metabolism, Mitochondria metabolism, Intestinal Mucosa metabolism, Crohn Disease, Gastrointestinal Microbiome, Ileitis metabolism, Inflammatory Bowel Diseases metabolism
- Abstract
Background: Intestinal epithelial cell (IEC) mitochondrial dysfunction involvement in inflammatory bowel diseases (IBD), including Crohn's disease affecting the small intestine, is emerging in recent studies. As the interface between the self and the gut microbiota, IECs serve as hubs of bidirectional cross-talk between host and luminal microbiota. However, the role of mitochondrial-microbiota interaction in the ileum is largely unexplored. Prohibitin 1 (PHB1), a chaperone protein of the inner mitochondrial membrane required for optimal electron transport chain function, is decreased during IBD. We previously demonstrated that mice deficient in PHB1 specifically in IECs (Phb1
i∆IEC ) exhibited mitochondrial impairment, Paneth cell defects, gut microbiota dysbiosis, and spontaneous inflammation in the ileum (ileitis). Mice deficient in PHB1 in Paneth cells (epithelial secretory cells of the small intestine; Phb1∆PC ) also exhibited mitochondrial impairment, Paneth cell defects, and spontaneous ileitis. Here, we determined whether this phenotype is driven by Phb1 deficiency-associated ileal microbiota alterations or direct effects of loss of PHB1 in host IECs., Results: Depletion of gut microbiota by broad-spectrum antibiotic treatment in Phb1∆PC or Phb1i∆IEC mice revealed a necessary role of microbiota to cause ileitis. Using germ-free mice colonized with ileal microbiota from Phb1-deficient mice, we show that this microbiota could not independently induce ileitis without host mitochondrial dysfunction. The luminal microbiota phenotype of Phb1i∆IEC mice included a loss of the short-chain fatty acid butyrate. Supplementation of butyrate in Phb1-deficient mice ameliorated Paneth cell abnormalities and ileitis. Phb1-deficient ileal enteroid models suggest deleterious epithelial-intrinsic responses to ileal microbiota that were protected by butyrate., Conclusions: These results suggest a mutual and essential reinforcing interplay of gut microbiota and host IEC, including Paneth cell, mitochondrial health in influencing ileitis. Restoration of butyrate is a potential therapeutic option in Crohn's disease patients harboring epithelial cell mitochondrial dysfunction. Video Abstract., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
16. A defect in mitochondrial fatty acid synthesis impairs iron metabolism and causes elevated ceramide levels.
- Author
-
Dutta D, Kanca O, Byeon SK, Marcogliese PC, Zuo Z, Shridharan RV, Park JH, Lin G, Ge M, Heimer G, Kohler JN, Wheeler MT, Kaipparettu BA, Pandey A, and Bellen HJ
- Subjects
- Child, Animals, Humans, Ceramides, Drosophila, Iron, Fatty Acids, Adipogenesis, Mitochondria
- Abstract
In most eukaryotic cells, fatty acid synthesis (FAS) occurs in the cytoplasm and in mitochondria. However, the relative contribution of mitochondrial FAS (mtFAS) to the cellular lipidome is not well defined. Here we show that loss of function of Drosophila mitochondrial enoyl coenzyme A reductase (Mecr), which is the enzyme required for the last step of mtFAS, causes lethality, while neuronal loss of Mecr leads to progressive neurodegeneration. We observe a defect in Fe-S cluster biogenesis and increased iron levels in flies lacking mecr, leading to elevated ceramide levels. Reducing the levels of either iron or ceramide suppresses the neurodegenerative phenotypes, indicating an interplay between ceramide and iron metabolism. Mutations in human MECR cause pediatric-onset neurodegeneration, and we show that human-derived fibroblasts display similar elevated ceramide levels and impaired iron homeostasis. In summary, this study identifies a role of mecr/MECR in ceramide and iron metabolism, providing a mechanistic link between mtFAS and neurodegeneration., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
17. Vitamin B2 enables regulation of fasting glucose availability.
- Author
-
Masschelin PM, Saha P, Ochsner SA, Cox AR, Kim KH, Felix JB, Sharp R, Li X, Tan L, Park JH, Wang L, Putluri V, Lorenzi PL, Nuotio-Antar AM, Sun Z, Kaipparettu BA, Putluri N, Moore DD, Summers SA, McKenna NJ, and Hartig SM
- Subjects
- Mice, Animals, PPAR alpha genetics, PPAR alpha metabolism, Flavin-Adenine Dinucleotide metabolism, Fatty Acids metabolism, Liver metabolism, Fasting metabolism, Oxidation-Reduction, Flavoproteins metabolism, Glucose metabolism, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Flavin adenine dinucleotide (FAD) interacts with flavoproteins to mediate oxidation-reduction reactions required for cellular energy demands. Not surprisingly, mutations that alter FAD binding to flavoproteins cause rare inborn errors of metabolism (IEMs) that disrupt liver function and render fasting intolerance, hepatic steatosis, and lipodystrophy. In our study, depleting FAD pools in mice with a vitamin B2-deficient diet (B2D) caused phenotypes associated with organic acidemias and other IEMs, including reduced body weight, hypoglycemia, and fatty liver disease. Integrated discovery approaches revealed B2D tempered fasting activation of target genes for the nuclear receptor PPARα, including those required for gluconeogenesis. We also found PPARα knockdown in the liver recapitulated B2D effects on glucose excursion and fatty liver disease in mice. Finally, treatment with the PPARα agonist fenofibrate activated the integrated stress response and refilled amino acid substrates to rescue fasting glucose availability and overcome B2D phenotypes. These findings identify metabolic responses to FAD availability and nominate strategies for the management of organic acidemias and other rare IEMs., Competing Interests: PM, PS, SO, AC, KK, JF, RS, XL, LT, JP, LW, VP, PL, AN, ZS, BK, NP, DM, NM, SH No competing interests declared, SS Scott Summers is a co-founder and shareholder of Centaurus Therapeutics. There are no competing interests otherwise related to this article, (© 2023, Masschelin et al.)
- Published
- 2023
- Full Text
- View/download PDF
18. Screening of GPCR drugs for repurposing in breast cancer.
- Author
-
Abdulkareem NM, Bhat R, Powell RT, Chikermane S, Yande S, Trinh L, Abdelnasser HY, Tabassum M, Ruiz A, Sobieski M, Nguyen ND, Park JH, Johnson CA, Kaipparettu BA, Bond RA, Johnson M, Stephan C, and Trivedi MV
- Abstract
Drug repurposing can overcome both substantial costs and the lengthy process of new drug discovery and development in cancer treatment. Some Food and Drug Administration (FDA)-approved drugs have been found to have the potential to be repurposed as anti-cancer drugs. However, the progress is slow due to only a handful of strategies employed to identify drugs with repurposing potential. In this study, we evaluated GPCR-targeting drugs by high throughput screening (HTS) for their repurposing potential in triple-negative breast cancer (TNBC) and drug-resistant human epidermal growth factor receptor-2-positive (HER2+) breast cancer (BC), due to the dire need to discover novel targets and drugs in these subtypes. We assessed the efficacy and potency of drugs/compounds targeting different GPCRs for the growth rate inhibition in the following models: two TNBC cell lines (MDA-MB-231 and MDA-MB-468) and two HER2+ BC cell lines (BT474 and SKBR3), sensitive or resistant to lapatinib + trastuzumab, an effective combination of HER2-targeting therapies. We identified six drugs/compounds as potential hits, of which 4 were FDA-approved drugs. We focused on β-adrenergic receptor-targeting nebivolol as a candidate, primarily because of the potential role of these receptors in BC and its excellent long-term safety profile. The effects of nebivolol were validated in an independent assay in all the cell line models. The effects of nebivolol were independent of its activation of β3 receptors and nitric oxide production. Nebivolol reduced invasion and migration potentials which also suggests its inhibitory role in metastasis. Analysis of the Surveillance, Epidemiology and End Results (SEER)-Medicare dataset found numerically but not statistically significant reduced risk of all-cause mortality in the nebivolol group. In-depth future analyses, including detailed in vivo studies and real-world data analysis with more patients, are needed to further investigate the potential of nebivolol as a repurposed therapy for BC., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Abdulkareem, Bhat, Powell, Chikermane, Yande, Trinh, Abdelnasser, Tabassum, Ruiz, Sobieski, Nguyen, Park, Johnson, Kaipparettu, Bond, Johnson, Stephan and Trivedi.)
- Published
- 2022
- Full Text
- View/download PDF
19. Redox regulation of hybrid metabolic state in breast cancer metastasis.
- Author
-
Attri KS, Park JH, and Kaipparettu BA
- Abstract
Competing Interests: Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-22-3730/coif). BAK was supported by R01CA253445, R01CA234479 and W81XWH-18-1-0714. The other authors have no conflicts of interest to declare.
- Published
- 2022
- Full Text
- View/download PDF
20. Targeting aberrant replication and DNA repair events for treating breast cancers.
- Author
-
Rajamanickam S, Park JH, Subbarayalu P, Timilsina S, Bates K, Yadav P, Nirzhor SSR, Eedunuri V, Mohammad TA, Jung KH, Onyeagucha B, Abdelfattah N, Benevides R, Lee G, Chen Y, Vadlamudi R, Brenner A, Kaklamani V, Jatoi I, Kuhn J, Hromas R, Gupta YK, Kaipparettu BA, Arbiser JL, and Rao MK
- Subjects
- Animals, DNA, DNA Repair, Female, Humans, Mice, Receptors, Estrogen metabolism, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
The major limitations of DNA-targeting chemotherapy drugs include life-threatening toxicity, acquired resistance and occurrence of secondary cancers. Here, we report a small molecule, Carbazole Blue (CB), that binds to DNA and inhibits cancer growth and metastasis by targeting DNA-related processes that tumor cells use but not the normal cells. We show that CB inhibits the expression of pro-tumorigenic genes that promote unchecked replication and aberrant DNA repair that cancer cells get addicted to survive. In contrast to chemotherapy drugs, systemic delivery of CB suppressed breast cancer growth and metastasis with no toxicity in pre-clinical mouse models. Using PDX and ex vivo explants from estrogen receptor (ER) positive, ER mutant and TNBC patients, we further demonstrated that CB effectively blocks therapy-sensitive and therapy-resistant breast cancer growth without affecting normal breast tissue. Our data provide a strong rationale to develop CB as a viable therapeutic for treating breast cancers., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. A mechanistic modeling framework reveals the key principles underlying tumor metabolism.
- Author
-
Tripathi S, Park JH, Pudakalakatti S, Bhattacharya PK, Kaipparettu BA, and Levine H
- Subjects
- Adenosine Triphosphate metabolism, Citric Acid Cycle, Humans, Phosphofructokinase-1 metabolism, Glycolysis, Neoplasms metabolism
- Abstract
While aerobic glycolysis, or the Warburg effect, has for a long time been considered a hallmark of tumor metabolism, recent studies have revealed a far more complex picture. Tumor cells exhibit widespread metabolic heterogeneity, not only in their presentation of the Warburg effect but also in the nutrients and the metabolic pathways they are dependent on. Moreover, tumor cells can switch between different metabolic phenotypes in response to environmental cues and therapeutic interventions. A framework to analyze the observed metabolic heterogeneity and plasticity is, however, lacking. Using a mechanistic model that includes the key metabolic pathways active in tumor cells, we show that the inhibition of phosphofructokinase by excess ATP in the cytoplasm can drive a preference for aerobic glycolysis in fast-proliferating tumor cells. The differing rates of ATP utilization by tumor cells can therefore drive heterogeneity with respect to the presentation of the Warburg effect. Building upon this idea, we couple the metabolic phenotype of tumor cells to their migratory phenotype, and show that our model predictions are in agreement with previous experiments. Next, we report that the reliance of proliferating cells on different anaplerotic pathways depends on the relative availability of glucose and glutamine, and can further drive metabolic heterogeneity. Finally, using treatment of melanoma cells with a BRAF inhibitor as an example, we show that our model can be used to predict the metabolic and gene expression changes in cancer cells in response to drug treatment. By making predictions that are far more generalizable and interpretable as compared to previous tumor metabolism modeling approaches, our framework identifies key principles that govern tumor cell metabolism, and the reported heterogeneity and plasticity. These principles could be key to targeting the metabolic vulnerabilities of cancer., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
22. Patient-derived iPSCs link elevated mitochondrial respiratory complex I function to osteosarcoma in Rothmund-Thomson syndrome.
- Author
-
Jewell BE, Xu A, Zhu D, Huang MF, Lu L, Liu M, Underwood EL, Park JH, Fan H, Gingold JA, Zhou R, Tu J, Huo Z, Liu Y, Jin W, Chen YH, Xu Y, Chen SH, Rainusso N, Berg NK, Bazer DA, Vellano C, Jones P, Eltzschig HK, Zhao Z, Kaipparettu BA, Zhao R, Wang LL, and Lee DF
- Subjects
- Adenosine Triphosphate biosynthesis, Cell Proliferation drug effects, Cell Respiration drug effects, Cellular Senescence genetics, Electron Transport Complex I antagonists & inhibitors, Gene Expression Regulation, Developmental genetics, Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells metabolism, Mitogen-Activated Protein Kinase Kinases genetics, Mutation genetics, Osteoblasts drug effects, Osteogenesis genetics, Osteosarcoma complications, Osteosarcoma pathology, Oxadiazoles pharmacology, Oxidative Phosphorylation drug effects, Piperidines pharmacology, Rothmund-Thomson Syndrome complications, Rothmund-Thomson Syndrome pathology, Electron Transport Complex I genetics, Osteosarcoma genetics, RNA, Long Noncoding genetics, RecQ Helicases genetics, Rothmund-Thomson Syndrome genetics
- Abstract
Rothmund-Thomson syndrome (RTS) is an autosomal recessive genetic disorder characterized by poikiloderma, small stature, skeletal anomalies, sparse brows/lashes, cataracts, and predisposition to cancer. Type 2 RTS patients with biallelic RECQL4 pathogenic variants have multiple skeletal anomalies and a significantly increased incidence of osteosarcoma. Here, we generated RTS patient-derived induced pluripotent stem cells (iPSCs) to dissect the pathological signaling leading to RTS patient-associated osteosarcoma. RTS iPSC-derived osteoblasts showed defective osteogenic differentiation and gain of in vitro tumorigenic ability. Transcriptome analysis of RTS osteoblasts validated decreased bone morphogenesis while revealing aberrantly upregulated mitochondrial respiratory complex I gene expression. RTS osteoblast metabolic assays demonstrated elevated mitochondrial respiratory complex I function, increased oxidative phosphorylation (OXPHOS), and increased ATP production. Inhibition of mitochondrial respiratory complex I activity by IACS-010759 selectively suppressed cellular respiration and cell proliferation of RTS osteoblasts. Furthermore, systems analysis of IACS-010759-induced changes in RTS osteoblasts revealed that chemical inhibition of mitochondrial respiratory complex I impaired cell proliferation, induced senescence, and decreased MAPK signaling and cell cycle associated genes, but increased H19 and ribosomal protein genes. In summary, our study suggests that mitochondrial respiratory complex I is a potential therapeutic target for RTS-associated osteosarcoma and provides future insights for clinical treatment strategies., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
23. Towards decoding the coupled decision-making of metabolism and epithelial-to-mesenchymal transition in cancer.
- Author
-
Jia D, Park JH, Kaur H, Jung KH, Yang S, Tripathi S, Galbraith M, Deng Y, Jolly MK, Kaipparettu BA, Onuchic JN, and Levine H
- Subjects
- Animals, Cell Differentiation, Epithelial-Mesenchymal Transition genetics, Humans, Neoplasm Metastasis, Neoplasms metabolism, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells physiology, Energy Metabolism physiology, Epithelial-Mesenchymal Transition physiology, Neoplasms pathology
- Abstract
Cancer cells have the plasticity to adjust their metabolic phenotypes for survival and metastasis. A developmental programme known as epithelial-to-mesenchymal transition (EMT) plays a critical role during metastasis, promoting the loss of polarity and cell-cell adhesion and the acquisition of motile, stem-cell characteristics. Cells undergoing EMT or the reverse mesenchymal-to-epithelial transition (MET) are often associated with metabolic changes, as the change in phenotype often correlates with a different balance of proliferation versus energy-intensive migration. Extensive crosstalk occurs between metabolism and EMT, but how this crosstalk leads to coordinated physiological changes is still uncertain. The elusive connection between metabolism and EMT compromises the efficacy of metabolic therapies targeting metastasis. In this review, we aim to clarify the causation between metabolism and EMT on the basis of experimental studies, and propose integrated theoretical-experimental efforts to better understand the coupled decision-making of metabolism and EMT.
- Published
- 2021
- Full Text
- View/download PDF
24. Targeting Mitochondrial Damage as a Therapeutic for Ileal Crohn's Disease.
- Author
-
Alula KM, Jackson DN, Smith AD, Kim DS, Turner K, Odstrcil E, Kaipparettu BA, Dassopoulos T, Venuprasad K, Feagins LA, and Theiss AL
- Subjects
- Biopsy methods, Enterocytes cytology, Epithelium drug effects, Epithelium pathology, Humans, Lipid Metabolism physiology, Paneth Cells pathology, Phenotype, Antioxidants therapeutic use, Crohn Disease drug therapy, Crohn Disease metabolism, Inflammation drug therapy, Mitochondria metabolism
- Abstract
Paneth cell defects in Crohn's disease (CD) patients (called the Type I phenotype) are associated with worse clinical outcomes. Recent studies have implicated mitochondrial dysfunction in Paneth cells as a mediator of ileitis in mice. We hypothesized that CD Paneth cells exhibit impaired mitochondrial health and that mitochondrial-targeted therapeutics may provide a novel strategy for ileal CD. Terminal ileal mucosal biopsies from adult CD and non-IBD patients were characterized for Paneth cell phenotyping and mitochondrial damage. To demonstrate the response of mitochondrial-targeted therapeutics in CD, biopsies were treated with vehicle or Mito-Tempo, a mitochondrial-targeted antioxidant, and RNA transcriptome was analyzed. During active CD inflammation, the epithelium exhibited mitochondrial damage evident in Paneth cells, goblet cells, and enterocytes. Independent of inflammation, Paneth cells in Type I CD patients exhibited mitochondrial damage. Mito-Tempo normalized the expression of interleukin (IL)-17/IL-23, lipid metabolism, and apoptotic gene signatures in CD patients to non-IBD levels. When stratified by Paneth cell phenotype, the global tissue response to Mito-Tempo in Type I patients was associated with innate immune, lipid metabolism, and G protein-coupled receptor (GPCR) gene signatures. Targeting impaired mitochondria as an underlying contributor to inflammation provides a novel treatment approach for CD.
- Published
- 2021
- Full Text
- View/download PDF
25. Ataxia-telangiectasia mutated interacts with Parkin and induces mitophagy independent of kinase activity. Evidence from mantle cell lymphoma.
- Author
-
Sarkar A, Stellrecht CM, Vangapandu HV, Ayres M, Kaipparettu BA, Park JH, Balakrishnan K, Burks JK, Pandita TK, Hittelman WN, Neelapu SS, and Gandhi V
- Subjects
- Adult, Animals, HeLa Cells, Humans, Mice, Mitophagy genetics, Phosphorylation, Protein Kinases genetics, Protein Kinases metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Ataxia Telangiectasia, Ataxia Telangiectasia Mutated Proteins, Lymphoma, Mantle-Cell genetics
- Abstract
Ataxia telangiectasia mutated (ATM), a critical DNA damage sensor with protein kinase activity,is frequently altered in human cancers including mantle cell lymphoma (MCL). Loss of ATM protein is linked to accumulation of nonfunctional mitochondria and defective mitophagy, in both murine thymocytes and in A-T cells. However, the mechanistic role of ATM kinase in cancer cell mitophagy is unknown. Here, we provide evidence that FCCP-induced mitophagy in MCL and other cancer cell lines is dependent on ATM but independent of its kinase function. While Granta-519 MCL cells possess single copy and kinase dead ATM and are resistant to FCCP-induced mitophagy, both Jeko-1 and Mino cells are ATM proficient and induce mitophagy. Stable knockdown of ATM in Jeko-1 and Mino cells conferred resistance to mitophagy and was associated with reduced ATP production, oxygen consumption, and increased mROS. ATM interacts with the E3 ubiquitin ligase Parkin in a kinase-independent manner. Knockdown of ATM in HeLa cells resulted in proteasomal degradation of GFP-Parkin which was rescued by the proteasome inhibitor, MG132 suggesting that ATM-Parkin interaction is important for Parkin stability. Neither loss of ATM kinase activity in primary B cell lymphomas nor inhibition of ATM kinase in MCL, A-T and HeLa cell lines mitigated FCCP or CCCP-induced mitophagy suggesting that ATM kinase activity is dispensable for mitophagy. Malignant B-cell lymphomas without detectable ATM, Parkin, Pink1, and Parkin-Ub ser65 phosphorylation were resistant to mitophagy, providing the first molecular evidence of ATM's role in mitophagy in MCL and other B-cell lymphomas.
- Published
- 2021
- Full Text
- View/download PDF
26. TP53 Status as a Determinant of Pro- vs Anti-Tumorigenic Effects of Estrogen Receptor-Beta in Breast Cancer.
- Author
-
Mukhopadhyay UK, Oturkar CC, Adams C, Wickramasekera N, Bansal S, Medisetty R, Miller A, Swetzig WM, Silwal-Pandit L, Børresen-Dale AL, Creighton CJ, Park JH, Konduri SD, Mukhopadhyay A, Caradori A, Omilian A, Bshara W, Kaipparettu BA, and Das GM
- Subjects
- Biomarkers, Tumor genetics, Carcinogenesis genetics, Carcinogenesis metabolism, Cell Proliferation, Cohort Studies, Estrogen Receptor beta genetics, Female, Humans, Mutant Proteins genetics, Prognosis, Survival Rate, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Biomarkers, Tumor metabolism, Carcinogenesis pathology, Estrogen Receptor beta metabolism, Mutant Proteins metabolism, Mutation, Triple Negative Breast Neoplasms pathology, Tumor Suppressor Protein p53 metabolism
- Abstract
Background: Anti-tumorigenic vs pro-tumorigenic roles of estrogen receptor-beta (ESR2) in breast cancer remain unsettled. We investigated the potential of TP53 status to be a determinant of the bi-faceted role of ESR2 and associated therapeutic implications for triple negative breast cancer (TNBC)., Methods: ESR2-TP53 interaction was analyzed with multiple assays including the in situ proximity ligation assay. Transcriptional effects on TP53-target genes and cell proliferation in response to knocking down or overexpressing ESR2 were determined. Patient survival according to ESR2 expression levels and TP53 mutation status was analyzed in the basal-like TNBC subgroup in the Molecular Taxonomy of Breast Cancer International Consortium (n = 308) and Roswell Park Comprehensive Cancer Center (n = 46) patient cohorts by univariate Cox regression and log-rank test. All statistical tests are two-sided., Results: ESR2 interaction with wild-type and mutant TP53 caused pro-proliferative and anti-proliferative effects, respectively. Depleting ESR2 in cells expressing wild-type TP53 resulted in increased expression of TP53-target genes CDKN1A (control group mean [SD] = 1 [0.13] vs ESR2 depletion group mean [SD] = 2.08 [0.24], P = .003) and BBC3 (control group mean [SD] = 1 [0.06] vs ESR2 depleted group mean [SD] = 1.92 [0.25], P = .003); however, expression of CDKN1A (control group mean [SD] = 1 [0.21] vs ESR2 depleted group mean [SD] = 0.56 [0.12], P = .02) and BBC3 (control group mean [SD] = 1 [0.03] vs ESR2 depleted group mean [SD] = 0.55 [0.09], P = .008) was decreased in cells expressing mutant TP53. Overexpressing ESR2 had opposite effects. Tamoxifen increased ESR2-mutant TP53 interaction, leading to reactivation of TP73 and apoptosis. High levels of ESR2 expression in mutant TP53-expressing basal-like tumors is associated with better prognosis (Molecular Taxonomy of Breast Cancer International Consortium cohort: log-rank P = .001; hazard ratio = 0.26, 95% confidence interval = 0.08 to 0.84, univariate Cox P = .02)., Conclusions: TP53 status is a determinant of the functional duality of ESR2. Our study suggests that ESR2-mutant TP53 combination prognosticates survival in TNBC revealing a novel strategy to stratify TNBC for therapeutic intervention potentially by repurposing tamoxifen., (© The Author(s) 2019. Published by Oxford University Press.)
- Published
- 2019
- Full Text
- View/download PDF
27. Therapeutic inhibition of mTORC2 rescues the behavioral and neurophysiological abnormalities associated with Pten-deficiency.
- Author
-
Chen CJ, Sgritta M, Mays J, Zhou H, Lucero R, Park J, Wang IC, Park JH, Kaipparettu BA, Stoica L, Jafar-Nejad P, Rigo F, Chin J, Noebels JL, and Costa-Mattioli M
- Subjects
- Animals, Brain metabolism, Brain pathology, Disease Models, Animal, Humans, Loss of Function Mutation genetics, Mechanistic Target of Rapamycin Complex 1 genetics, Mice, Mice, Knockout, Nervous System Diseases metabolism, Nervous System Diseases pathology, Oligonucleotides, Antisense genetics, Oligonucleotides, Antisense pharmacology, PTEN Phosphohydrolase deficiency, Rapamycin-Insensitive Companion of mTOR Protein antagonists & inhibitors, Rapamycin-Insensitive Companion of mTOR Protein genetics, Tuberous Sclerosis Complex 1 Protein genetics, Mechanistic Target of Rapamycin Complex 2 genetics, Nervous System Diseases genetics, PTEN Phosphohydrolase genetics, TOR Serine-Threonine Kinases genetics
- Abstract
Dysregulation of the mammalian target of rapamycin (mTOR) signaling, which is mediated by two structurally and functionally distinct complexes, mTORC1 and mTORC2, has been implicated in several neurological disorders
1-3 . Individuals carrying loss-of-function mutations in the phosphatase and tensin homolog (PTEN) gene, a negative regulator of mTOR signaling, are prone to developing macrocephaly, autism spectrum disorder (ASD), seizures and intellectual disability2,4,5 . It is generally believed that the neurological symptoms associated with loss of PTEN and other mTORopathies (for example, mutations in the tuberous sclerosis genes TSC1 or TSC2) are due to hyperactivation of mTORC1-mediated protein synthesis1,2,4,6,7 . Using molecular genetics, we unexpectedly found that genetic deletion of mTORC2 (but not mTORC1) activity prolonged lifespan, suppressed seizures, rescued ASD-like behaviors and long-term memory, and normalized metabolic changes in the brain of mice lacking Pten. In a more therapeutically oriented approach, we found that administration of an antisense oligonucleotide (ASO) targeting mTORC2's defining component Rictor specifically inhibits mTORC2 activity and reverses the behavioral and neurophysiological abnormalities in adolescent Pten-deficient mice. Collectively, our findings indicate that mTORC2 is the major driver underlying the neuropathophysiology associated with Pten-deficiency, and its therapeutic reduction could represent a promising and broadly effective translational therapy for neurological disorders where mTOR signaling is dysregulated.- Published
- 2019
- Full Text
- View/download PDF
28. Correction: Crosstalk from Non-Cancerous Mitochondria Can Inhibit Tumor Properties of Metastatic Cells by Suppressing Oncogenic Pathways.
- Author
-
Kaipparettu BA, Ma Y, Park JH, Lee TL, Zhang Y, Yotnda P, Creighton CJ, Chan WY, and Wong LC
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0061747.].
- Published
- 2019
- Full Text
- View/download PDF
29. Aerobic Plus Resistance Exercise in Obese Older Adults Improves Muscle Protein Synthesis and Preserves Myocellular Quality Despite Weight Loss.
- Author
-
Colleluori G, Aguirre L, Phadnis U, Fowler K, Armamento-Villareal R, Sun Z, Brunetti L, Hyoung Park J, Kaipparettu BA, Putluri N, Auetumrongsawat V, Yarasheski K, Qualls C, and Villareal DT
- Subjects
- Aged, Humans, Weight Loss, Exercise, Muscle Proteins biosynthesis, Muscle, Skeletal metabolism, Obesity metabolism, Resistance Training
- Abstract
Anabolic resistance and impaired myocellular quality contribute to age-related sarcopenia, which exacerbates with obesity. Diet-induced muscle mass loss is attenuated by resistance or aerobic plus resistance exercise compared to aerobic exercise in obese elderly. We assessed chronic effects of weight loss plus different exercise modalities on muscle protein synthesis response to feeding and myocellular quality. Obese older adults were randomized to a weight-management program plus aerobic, resistance, or combined aerobic and resistance exercise or to control. Participants underwent vastus lateralis biopsies at baseline and 6 months. Muscle protein synthesis rate increased more in resistance and combined than in control. Autophagy mediators' expression decreased more in combined than in aerobic, which experienced a higher increase in inflammation and mitochondrial regulators' expression. In obese elderly, combined aerobic and resistance exercise is superior to either mode independently for improving muscle protein synthesis and myocellular quality, thereby maintaining muscle mass during weight-loss therapy., (Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
30. Assessing Therapeutic Efficacy in Real-time by Hyperpolarized Magnetic Resonance Metabolic Imaging.
- Author
-
Dutta P, Salzillo TC, Pudakalakatti S, Gammon ST, Kaipparettu BA, McAllister F, Wagner S, Frigo DE, Logothetis CJ, Zacharias NM, and Bhattacharya PK
- Subjects
- Animals, Cell Line, Humans, Mice, Rats, Carbon Isotopes metabolism, Magnetic Resonance Imaging methods, Neoplasms metabolism, Neoplasms therapy, Outcome and Process Assessment, Health Care, Pyruvic Acid metabolism
- Abstract
Precisely measuring tumor-associated alterations in metabolism clinically will enable the efficient assessment of therapeutic responses. Advances in imaging technologies can exploit the differences in cancer-associated cell metabolism as compared to normal tissue metabolism, linking changes in target metabolism to therapeutic efficacy. Metabolic imaging by Positron Emission Tomography (PET) employing 2-fluoro-deoxy-glucose ([
18 F]FDG) has been used as a routine diagnostic tool in the clinic. Recently developed hyperpolarized Magnetic Resonance (HP-MR), which radically increases the sensitivity of conventional MRI, has created a renewed interest in functional and metabolic imaging. The successful translation of this technique to the clinic was achieved recently with measurements of13 C-pyruvate metabolism. Here, we review the potential clinical roles for metabolic imaging with hyperpolarized MRI as applied in assessing therapeutic intervention in different cancer systems.- Published
- 2019
- Full Text
- View/download PDF
31. Elucidating cancer metabolic plasticity by coupling gene regulation with metabolic pathways.
- Author
-
Jia D, Lu M, Jung KH, Park JH, Yu L, Onuchic JN, Kaipparettu BA, and Levine H
- Subjects
- AMP-Activated Protein Kinases metabolism, Cell Line, Tumor, Fatty Acids metabolism, Female, Glucose metabolism, Glycolysis genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mitochondria metabolism, Models, Theoretical, Oxidative Phosphorylation, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, AMP-Activated Protein Kinases genetics, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Metabolic Networks and Pathways genetics, Triple Negative Breast Neoplasms genetics
- Abstract
Metabolic plasticity enables cancer cells to switch their metabolism phenotypes between glycolysis and oxidative phosphorylation (OXPHOS) during tumorigenesis and metastasis. However, it is still largely unknown how cancer cells orchestrate gene regulation to balance their glycolysis and OXPHOS activities. Previously, by modeling the gene regulation of cancer metabolism we have reported that cancer cells can acquire a stable hybrid metabolic state in which both glycolysis and OXPHOS can be used. Here, to comprehensively characterize cancer metabolic activity, we establish a theoretical framework by coupling gene regulation with metabolic pathways. Our modeling results demonstrate a direct association between the activities of AMPK and HIF-1, master regulators of OXPHOS and glycolysis, respectively, with the activities of three major metabolic pathways: glucose oxidation, glycolysis, and fatty acid oxidation. Our model further characterizes the hybrid metabolic state and a metabolically inactive state where cells have low activity of both glycolysis and OXPHOS. We verify the model prediction using metabolomics and transcriptomics data from paired tumor and adjacent benign tissue samples from a cohort of breast cancer patients and RNA-sequencing data from The Cancer Genome Atlas. We further validate the model prediction by in vitro studies of aggressive triple-negative breast cancer (TNBC) cells. The experimental results confirm that TNBC cells can maintain a hybrid metabolic phenotype and targeting both glycolysis and OXPHOS is necessary to eliminate their metabolic plasticity. In summary, our work serves as a platform to symmetrically study how tuning gene activity modulates metabolic pathway activity, and vice versa., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
32. Pharmacological targeting of MYC-regulated IRE1/XBP1 pathway suppresses MYC-driven breast cancer.
- Author
-
Zhao N, Cao J, Xu L, Tang Q, Dobrolecki LE, Lv X, Talukdar M, Lu Y, Wang X, Hu DZ, Shi Q, Xiang Y, Wang Y, Liu X, Bu W, Jiang Y, Li M, Gong Y, Sun Z, Ying H, Yuan B, Lin X, Feng XH, Hartig SM, Li F, Shen H, Chen Y, Han L, Zeng Q, Patterson JB, Kaipparettu BA, Putluri N, Sicheri F, Rosen JM, Lewis MT, and Chen X
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Endoribonucleases genetics, Female, Humans, Mice, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins c-myc genetics, Saccharomyces cerevisiae, Signal Transduction genetics, Unfolded Protein Response drug effects, Unfolded Protein Response genetics, X-Box Binding Protein 1 genetics, Xenograft Model Antitumor Assays, Breast Neoplasms drug therapy, Docetaxel pharmacology, Drug Delivery Systems, Endoribonucleases metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-myc metabolism, Response Elements, Signal Transduction drug effects, X-Box Binding Protein 1 metabolism
- Abstract
The unfolded protein response (UPR) is a cellular homeostatic mechanism that is activated in many human cancers and plays pivotal roles in tumor progression and therapy resistance. However, the molecular mechanisms for UPR activation and regulation in cancer cells remain elusive. Here, we show that oncogenic MYC regulates the inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (XBP1) branch of the UPR in breast cancer via multiple mechanisms. We found that MYC directly controls IRE1 transcription by binding to its promoter and enhancer. Furthermore, MYC forms a transcriptional complex with XBP1, a target of IRE1, and enhances its transcriptional activity. Importantly, we demonstrate that XBP1 is a synthetic lethal partner of MYC. Silencing of XBP1 selectively blocked the growth of MYC-hyperactivated cells. Pharmacological inhibition of IRE1 RNase activity with small molecule inhibitor 8866 selectively restrained the MYC-overexpressing tumor growth in vivo in a cohort of preclinical patient-derived xenograft models and genetically engineered mouse models. Strikingly, 8866 substantially enhanced the efficacy of docetaxel chemotherapy, resulting in rapid regression of MYC-overexpressing tumors. Collectively, these data establish the synthetic lethal interaction of the IRE1/XBP1 pathway with MYC hyperactivation and provide a potential therapy for MYC-driven human breast cancers.
- Published
- 2018
- Full Text
- View/download PDF
33. GLUT12 promotes prostate cancer cell growth and is regulated by androgens and CaMKK2 signaling.
- Author
-
White MA, Tsouko E, Lin C, Rajapakshe K, Spencer JM, Wilkenfeld SR, Vakili SS, Pulliam TL, Awad D, Nikolos F, Katreddy RR, Kaipparettu BA, Sreekumar A, Zhang X, Cheung E, Coarfa C, and Frigo DE
- Subjects
- Androgens pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Glucose Transport Proteins, Facilitative genetics, Humans, Male, Metribolone pharmacology, Phosphorylation drug effects, Prostate drug effects, Prostate pathology, Prostatic Neoplasms pathology, RNA, Small Interfering, Signal Transduction drug effects, Calcium-Calmodulin-Dependent Protein Kinase Kinase metabolism, Glucose Transport Proteins, Facilitative metabolism, Prostate metabolism, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Signal Transduction physiology
- Abstract
Despite altered metabolism being an accepted hallmark of cancer, it is still not completely understood which signaling pathways regulate these processes. Given the central role of androgen receptor (AR) signaling in prostate cancer, we hypothesized that AR could promote prostate cancer cell growth in part through increasing glucose uptake via the expression of distinct glucose transporters. Here, we determined that AR directly increased the expression of SLC2A12 , the gene that encodes the glucose transporter GLUT12. In support of these findings, gene signatures of AR activity correlated with SLC2A12 expression in multiple clinical cohorts. Functionally, GLUT12 was required for maximal androgen-mediated glucose uptake and cell growth in LNCaP and VCaP cells. Knockdown of GLUT12 also decreased the growth of C4-2, 22Rv1 and AR-negative PC-3 cells. This latter observation corresponded with a significant reduction in glucose uptake, indicating that additional signaling mechanisms could augment GLUT12 function in an AR-independent manner. Interestingly, GLUT12 trafficking to the plasma membrane was modulated by calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-5'-AMP-activated protein kinase (AMPK) signaling, a pathway we previously demonstrated to be a downstream effector of AR. Inhibition of CaMKK2-AMPK signaling decreased GLUT12 translocation to the plasma membrane by inhibiting the phosphorylation of TBC1D4, a known regulator of glucose transport. Further, AR increased TBC1D4 expression. Correspondingly, expression of TBC1D4 correlated with AR activity in prostate cancer patient samples. Taken together, these data demonstrate that prostate cancer cells can increase the functional levels of GLUT12 through multiple mechanisms to promote glucose uptake and subsequent cell growth., (© 2018 Society for Endocrinology.)
- Published
- 2018
- Full Text
- View/download PDF
34. Elucidating the Metabolic Plasticity of Cancer: Mitochondrial Reprogramming and Hybrid Metabolic States.
- Author
-
Jia D, Park JH, Jung KH, Levine H, and Kaipparettu BA
- Abstract
Aerobic glycolysis, also referred to as the Warburg effect, has been regarded as the dominant metabolic phenotype in cancer cells for a long time. More recently, it has been shown that mitochondria in most tumors are not defective in their ability to carry out oxidative phosphorylation (OXPHOS). Instead, in highly aggressive cancer cells, mitochondrial energy pathways are reprogrammed to meet the challenges of high energy demand, better utilization of available fuels and macromolecular synthesis for rapid cell division and migration. Mitochondrial energy reprogramming is also involved in the regulation of oncogenic pathways via mitochondria-to-nucleus retrograde signaling and post-translational modification of oncoproteins. In addition, neoplastic mitochondria can engage in crosstalk with the tumor microenvironment. For example, signals from cancer-associated fibroblasts can drive tumor mitochondria to utilize OXPHOS, a process known as the reverse Warburg effect. Emerging evidence shows that cancer cells can acquire a hybrid glycolysis/OXPHOS phenotype in which both glycolysis and OXPHOS can be utilized for energy production and biomass synthesis. The hybrid glycolysis/OXPHOS phenotype facilitates metabolic plasticity of cancer cells and may be specifically associated with metastasis and therapy-resistance. Moreover, cancer cells can switch their metabolism phenotypes in response to external stimuli for better survival. Taking into account the metabolic heterogeneity and plasticity of cancer cells, therapies targeting cancer metabolic dependency in principle can be made more effective., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
35. B-cell Receptor Signaling Regulates Metabolism in Chronic Lymphocytic Leukemia.
- Author
-
Vangapandu HV, Havranek O, Ayres ML, Kaipparettu BA, Balakrishnan K, Wierda WG, Keating MJ, Davis RE, Stellrecht CM, and Gandhi V
- Subjects
- Adenosine Triphosphate genetics, Adenosine Triphosphate metabolism, Apoptosis genetics, Cell Line, Tumor, Cell Proliferation genetics, Class I Phosphatidylinositol 3-Kinases genetics, Humans, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Metabolic Networks and Pathways genetics, Oxidative Phosphorylation, Oxygen Consumption genetics, ZAP-70 Protein-Tyrosine Kinase genetics, Glycolysis genetics, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Prognosis, Receptors, Antigen, B-Cell genetics
- Abstract
Peripheral blood chronic lymphocytic leukemia (CLL) cells are quiescent but have active transcription and translation processes, suggesting that these lymphocytes are metabolically active. Based on this premise, the metabolic phenotype of CLL lymphocytes was investigated by evaluating the two intracellular ATP-generating pathways. Metabolic flux was assessed by measuring glycolysis as extracellular acidification rate (ECAR) and mitochondrial oxidative phosphorylation as oxygen consumption rate (OCR) and then correlated with prognostic factors. Further, the impact of B-cell receptor signaling (BCR) on metabolism was determined by genetic ablation and pharmacological inhibitors. Compared with proliferative B-cell lines, metabolic fluxes of oxygen and lactate were low in CLL cells. ECAR was consistently low, but OCR varied considerably in human patient samples ( n = 45). Higher OCR was associated with poor prognostic factors such as ZAP 70 positivity, unmutated IGHV, high β2M levels, and higher Rai stage. Consistent with the association of ZAP 70 and IGHV unmutated status with active BCR signaling, genetic ablation of BCR mitigated OCR in malignant B cells. Similarly, knocking out PI3Kδ, a critical component of the BCR pathway, decreased OCR and ECAR. In concert, PI3K pathway inhibitors dramatically reduced OCR and ECAR. In harmony with a decline in metabolic activity, the ribonucleotide pools in CLL cells were reduced with duvelisib treatment. Collectively, these data demonstrate that CLL metabolism, especially OCR, is linked to prognostic factors and is curbed by BCR and PI3K pathway inhibition. Implications: This study identifies a relationship between oxidative phosphorylation in CLL and prognostic factors providing a rationale to therapeutically target these processes. Mol Cancer Res; 15(12); 1692-703. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
36. Tobacco-Specific Carcinogens Induce Hypermethylation, DNA Adducts, and DNA Damage in Bladder Cancer.
- Author
-
Jin F, Thaiparambil J, Donepudi SR, Vantaku V, Piyarathna DWB, Maity S, Krishnapuram R, Putluri V, Gu F, Purwaha P, Bhowmik SK, Ambati CR, von Rundstedt FC, Roghmann F, Berg S, Noldus J, Rajapakshe K, Gödde D, Roth S, Störkel S, Degener S, Michailidis G, Kaipparettu BA, Karanam B, Terris MK, Kavuri SM, Lerner SP, Kheradmand F, Coarfa C, Sreekumar A, Lotan Y, El-Zein R, and Putluri N
- Subjects
- Azacitidine analogs & derivatives, Azacitidine pharmacology, Benzo(a)pyrene toxicity, Butanones blood, Carcinogens analysis, Cell Line, Tumor, Cohort Studies, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, DNA Adducts blood, Decitabine, Early Detection of Cancer methods, Female, Humans, Male, Metabolome drug effects, Metabolomics methods, Mutagens analysis, Neoplasm Grading, Nitrosamines toxicity, Polycyclic Aromatic Hydrocarbons blood, Polycyclic Aromatic Hydrocarbons urine, Risk Assessment methods, Smoking blood, Smoking urine, Nicotiana chemistry, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms therapy, Urinary Bladder Neoplasms urine, Biomarkers, Tumor urine, Carcinogens toxicity, DNA Damage drug effects, DNA Methylation drug effects, Mutagens toxicity, Smoking adverse effects, Nicotiana toxicity, Tobacco Products toxicity, Urinary Bladder Neoplasms metabolism
- Abstract
Smoking is a major risk factor for the development of bladder cancer; however, the functional consequences of the carcinogens in tobacco smoke and bladder cancer-associated metabolic alterations remain poorly defined. We assessed the metabolic profiles in bladder cancer smokers and non-smokers and identified the key alterations in their metabolism. LC/MS and bioinformatic analysis were performed to determine the metabolome associated with bladder cancer smokers and were further validated in cell line models. Smokers with bladder cancer were found to have elevated levels of methylated metabolites, polycyclic aromatic hydrocarbons, DNA adducts, and DNA damage. DNA methyltransferase 1 (DNMT1) expression was significantly higher in smokers than non-smokers with bladder cancer. An integromics approach, using multiple patient cohorts, revealed strong associations between smokers and high-grade bladder cancer. In vitro exposure to the tobacco smoke carcinogens, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and benzo[a]pyrene (BaP) led to increase in levels of methylated metabolites, DNA adducts, and extensive DNA damage in bladder cancer cells. Cotreatment of bladder cancer cells with these carcinogens and the methylation inhibitor 5-aza-2'-deoxycytidine rewired the methylated metabolites, DNA adducts, and DNA damage. These findings were confirmed through the isotopic-labeled metabolic flux analysis. Screens using smoke-associated metabolites and DNA adducts could provide robust biomarkers and improve individual risk prediction in bladder cancer smokers. Noninvasive predictive biomarkers that can stratify the risk of developing bladder cancer in smokers could aid in early detection and treatment. Cancer Prev Res; 10(10); 588-97. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
37. Modeling the Genetic Regulation of Cancer Metabolism: Interplay between Glycolysis and Oxidative Phosphorylation.
- Author
-
Yu L, Lu M, Jia D, Ma J, Ben-Jacob E, Levine H, Kaipparettu BA, and Onuchic JN
- Subjects
- AMP-Activated Protein Kinases physiology, Humans, Hypoxia-Inducible Factor 1 physiology, Models, Biological, Neoplasms genetics, Neoplasms therapy, Proto-Oncogene Proteins c-myc physiology, Reactive Oxygen Species metabolism, Tumor Microenvironment, Glycolysis, Neoplasms metabolism, Oxidative Phosphorylation
- Abstract
Abnormal metabolism is a hallmark of cancer, yet its regulation remains poorly understood. Cancer cells were considered to utilize primarily glycolysis for ATP production, referred to as the Warburg effect. However, recent evidence suggests that oxidative phosphorylation (OXPHOS) plays a crucial role during cancer progression. Here we utilized a systems biology approach to decipher the regulatory principle of glycolysis and OXPHOS. Integrating information from literature, we constructed a regulatory network of genes and metabolites, from which we extracted a core circuit containing HIF-1, AMPK, and ROS. Our circuit analysis showed that while normal cells have an oxidative state and a glycolytic state, cancer cells can access a hybrid state with both metabolic modes coexisting. This was due to higher ROS production and/or oncogene activation, such as RAS, MYC, and c-SRC. Guided by the model, we developed two signatures consisting of AMPK and HIF-1 downstream genes, respectively, to quantify the activity of glycolysis and OXPHOS. By applying the AMPK and HIF-1 signatures to The Cancer Genome Atlas patient transcriptomics data of multiple cancer types and single-cell RNA-seq data of lung adenocarcinoma, we confirmed an anticorrelation between AMPK and HIF-1 activities and the association of metabolic states with oncogenes. We propose that the hybrid phenotype contributes to metabolic plasticity, allowing cancer cells to adapt to various microenvironments. Using model simulations, our theoretical framework of metabolism can serve as a platform to decode cancer metabolic plasticity and design cancer therapies targeting metabolism. Cancer Res; 77(7); 1564-74. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
38. Bifunctional Luminomagnetic Rare-Earth Nanorods for High-Contrast Bioimaging Nanoprobes.
- Author
-
Gupta BK, Singh S, Kumar P, Lee Y, Kedawat G, Narayanan TN, Vithayathil SA, Ge L, Zhan X, Gupta S, Martí AA, Vajtai R, Ajayan PM, and Kaipparettu BA
- Subjects
- Cell Line, Tumor, Epithelial Cells ultrastructure, Humans, Luminescent Measurements, Magnets, Nanoparticles chemistry, Nanoparticles ultrastructure, Nanotubes ultrastructure, Europium chemistry, Gadolinium chemistry, Luminescent Agents chemistry, Nanotechnology methods, Nanotubes chemistry, Optical Imaging methods
- Abstract
Nanoparticles exhibiting both magnetic and luminescent properties are need of the hour for many biological applications. A single compound exhibiting this combination of properties is uncommon. Herein, we report a strategy to synthesize a bifunctional luminomagnetic Gd2-xEuxO3 (x = 0.05 to 0.5) nanorod, with a diameter of ~20 nm and length in ~0.6 μm, using hydrothermal method. Gd2O3:Eu(3+) nanorods have been characterized by studying its structural, optical and magnetic properties. The advantage offered by photoluminescent imaging with Gd2O3:Eu(3+) nanorods is that this ultrafine nanorod material exhibits hypersensitive intense red emission (610 nm) with good brightness (quantum yield more than 90%), which is an essential parameter for high-contrast bioimaging, especially for overcoming auto fluorescent background. The utility of luminomagnetic nanorods for biological applications in high-contrast cell imaging capability and cell toxicity to image two human breast cancer cell lines T47D and MDA-MB-231 are also evaluated. Additionally, to understand the significance of shape of the nanostructure, the photoluminescence and paramagnetic characteristic of Gd2O3:Eu(3+) nanorods were compared with the spherical nanoparticles of Gd2O3:Eu(3+).
- Published
- 2016
- Full Text
- View/download PDF
39. The autophagy inhibitor chloroquine targets cancer stem cells in triple negative breast cancer by inducing mitochondrial damage and impairing DNA break repair.
- Author
-
Liang DH, Choi DS, Ensor JE, Kaipparettu BA, Bass BL, and Chang JC
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carboplatin pharmacology, Cell Line, Tumor, Cell Movement drug effects, Dose-Response Relationship, Drug, Electron Transport Complex IV metabolism, Female, Histones metabolism, Humans, Membrane Potential, Mitochondrial drug effects, Mice, SCID, Mitochondria metabolism, Mitochondria ultrastructure, Neoplasm Metastasis, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells ultrastructure, Superoxides metabolism, Time Factors, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms ultrastructure, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Autophagy drug effects, Chloroquine pharmacology, DNA Damage, DNA Repair drug effects, Mitochondria drug effects, Neoplastic Stem Cells drug effects, Triple Negative Breast Neoplasms drug therapy
- Abstract
Triple negative breast cancer (TNBC), characterized by an abundance of treatment-resistant breast cancer stem cells (CSCs), has a poorer prognosis than other types of breast cancers. Despite its aggressiveness, no effective targeted therapy exists for TNBC. Here, we demonstrate that CQ effectively targets CSCs via autophagy inhibition, mitochondrial structural damage, and impairment of double-stranded DNA break repair. Electron microscopy demonstrates CQ-induced mitochondrial cristae damage, which leads to mitochondrial membrane depolarization with a significant reduction in the activity of cytochrome c oxidase and accumulation of superoxide and double-stranded DNA breaks. CQ effectively diminishes the TNBC cells' ability to metastasize in vitro and in a TNBC xenograft model. When administered in combination with carboplatin, CQ effectively inhibits carboplatin-induced autophagy. This combination treatment significantly diminishes the expression of DNA repair proteins in CSC subpopulations, resulting in tumor growth reduction in carboplatin-resistant BRCA1 wild-type TNBC orthotopic xenografts. As TNBC's high treatment failure rate has been attributed to enrichment of CSCs, CQ, an autophagy inhibitor with anti-CSC effects, may be an effective adjunct to current TNBC chemotherapy regimens with carboplatin., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Inhibition of the hexosamine biosynthetic pathway promotes castration-resistant prostate cancer.
- Author
-
Kaushik AK, Shojaie A, Panzitt K, Sonavane R, Venghatakrishnan H, Manikkam M, Zaslavsky A, Putluri V, Vasu VT, Zhang Y, Khan AS, Lloyd S, Szafran AT, Dasgupta S, Bader DA, Stossi F, Li H, Samanta S, Cao X, Tsouko E, Huang S, Frigo DE, Chan L, Edwards DP, Kaipparettu BA, Mitsiades N, Weigel NL, Mancini M, McGuire SE, Mehra R, Ittmann MM, Chinnaiyan AM, Putluri N, Palapattu GS, Michailidis G, and Sreekumar A
- Subjects
- Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Cell Line, Humans, Male, Mice, Mice, SCID, Phosphatidylinositol 3-Kinases metabolism, Prostatic Neoplasms, Castration-Resistant drug therapy, Proto-Oncogene Proteins c-akt metabolism, Hexosamines biosynthesis, Prostatic Neoplasms, Castration-Resistant metabolism
- Abstract
The precise molecular alterations driving castration-resistant prostate cancer (CRPC) are not clearly understood. Using a novel network-based integrative approach, here, we show distinct alterations in the hexosamine biosynthetic pathway (HBP) to be critical for CRPC. Expression of HBP enzyme glucosamine-phosphate N-acetyltransferase 1 (GNPNAT1) is found to be significantly decreased in CRPC compared with localized prostate cancer (PCa). Genetic loss-of-function of GNPNAT1 in CRPC-like cells increases proliferation and aggressiveness, in vitro and in vivo. This is mediated by either activation of the PI3K-AKT pathway in cells expressing full-length androgen receptor (AR) or by specific protein 1 (SP1)-regulated expression of carbohydrate response element-binding protein (ChREBP) in cells containing AR-V7 variant. Strikingly, addition of the HBP metabolite UDP-N-acetylglucosamine (UDP-GlcNAc) to CRPC-like cells significantly decreases cell proliferation, both in-vitro and in animal studies, while also demonstrates additive efficacy when combined with enzalutamide in-vitro. These observations demonstrate the therapeutic value of targeting HBP in CRPC.
- Published
- 2016
- Full Text
- View/download PDF
41. Multilevel Genomics-Based Taxonomy of Renal Cell Carcinoma.
- Author
-
Chen F, Zhang Y, Şenbabaoğlu Y, Ciriello G, Yang L, Reznik E, Shuch B, Micevic G, De Velasco G, Shinbrot E, Noble MS, Lu Y, Covington KR, Xi L, Drummond JA, Muzny D, Kang H, Lee J, Tamboli P, Reuter V, Shelley CS, Kaipparettu BA, Bottaro DP, Godwin AK, Gibbs RA, Getz G, Kucherlapati R, Park PJ, Sander C, Henske EP, Zhou JH, Kwiatkowski DJ, Ho TH, Choueiri TK, Hsieh JJ, Akbani R, Mills GB, Hakimi AA, Wheeler DA, and Creighton CJ
- Subjects
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell mortality, Chromatin metabolism, Gene Expression Profiling, Humans, Kidney Neoplasms genetics, Kidney Neoplasms mortality, MicroRNAs metabolism, Mutation, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, RNA, Messenger metabolism, Signal Transduction genetics, Survival Rate, TOR Serine-Threonine Kinases metabolism, Carcinoma, Renal Cell pathology, Genomics, Kidney Neoplasms pathology
- Abstract
On the basis of multidimensional and comprehensive molecular characterization (including DNA methalylation and copy number, RNA, and protein expression), we classified 894 renal cell carcinomas (RCCs) of various histologic types into nine major genomic subtypes. Site of origin within the nephron was one major determinant in the classification, reflecting differences among clear cell, chromophobe, and papillary RCC. Widespread molecular changes associated with TFE3 gene fusion or chromatin modifier genes were present within a specific subtype and spanned multiple subtypes. Differences in patient survival and in alteration of specific pathways (including hypoxia, metabolism, MAP kinase, NRF2-ARE, Hippo, immune checkpoint, and PI3K/AKT/mTOR) could further distinguish the subtypes. Immune checkpoint markers and molecular signatures of T cell infiltrates were both highest in the subtype associated with aggressive clear cell RCC. Differences between the genomic subtypes suggest that therapeutic strategies could be tailored to each RCC disease subset., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
42. Fatty Acid Oxidation-Driven Src Links Mitochondrial Energy Reprogramming and Oncogenic Properties in Triple-Negative Breast Cancer.
- Author
-
Park JH, Vithayathil S, Kumar S, Sung PL, Dobrolecki LE, Putluri V, Bhat VB, Bhowmik SK, Gupta V, Arora K, Wu D, Tsouko E, Zhang Y, Maity S, Donti TR, Graham BH, Frigo DE, Coarfa C, Yotnda P, Putluri N, Sreekumar A, Lewis MT, Creighton CJ, Wong LC, and Kaipparettu BA
- Subjects
- Animals, Carcinogenesis, Cell Line, Tumor, Female, Humans, Mice, SCID, Neoplasm Transplantation, Oxidation-Reduction, Phosphorylation, Protein Processing, Post-Translational, Triple Negative Breast Neoplasms pathology, Energy Metabolism, Fatty Acids metabolism, Mitochondria metabolism, Triple Negative Breast Neoplasms metabolism, src-Family Kinases metabolism
- Abstract
Transmitochondrial cybrids and multiple OMICs approaches were used to understand mitochondrial reprogramming and mitochondria-regulated cancer pathways in triple-negative breast cancer (TNBC). Analysis of cybrids and established breast cancer (BC) cell lines showed that metastatic TNBC maintains high levels of ATP through fatty acid β oxidation (FAO) and activates Src oncoprotein through autophosphorylation at Y419. Manipulation of FAO including the knocking down of carnitine palmitoyltransferase-1A (CPT1) and 2 (CPT2), the rate-limiting proteins of FAO, and analysis of patient-derived xenograft models confirmed the role of mitochondrial FAO in Src activation and metastasis. Analysis of TCGA and other independent BC clinical data further reaffirmed the role of mitochondrial FAO and CPT genes in Src regulation and their significance in BC metastasis., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
43. Eu 3+ doped α-sodium gadolinium fluoride luminomagnetic nanophosphor as a bimodal nanoprobe for high-contrast in vitro bioimaging and external magnetic field tracking applications.
- Author
-
Singh S, Kumar P, Kaipparettu BA, and Gupta BK
- Abstract
Herein, we introduce a novel strategy for the synthesis of Eu
3+ doped α-sodium gadolinium fluoride (α-NaGd0.88 F4 :Eu0.12 3+ ) based luminomagnetic nanophosphors using hydrothermal route. The synthesized nanophosphor has exceptional luminescent and paramagnetic properties in a single host lattice, which is highly desirable for biomedical applications. This highly luminescent nanophosphor with an average particle size ∼ 5±3 nm enables high-contrast fluorescent imaging with decreased light scattering. In vitro cellular uptake is shown by fluorescent microscopy that envisages the characteristic hypersensitive red emission of Eu3+ doped α-sodium gadolinium fluoride centered at 608 nm (5 D0 -7 F2 ) upon 465 nm excitation wavelength. No apparent cytotoxicity is observed. Furthermore, time- resolved emission spectroscopy and SQUID magnetic measurements successfully demonstrate a photoluminescence decay time in microseconds and enhanced paramagnetic behavior respectively, which promises the applications of nanophosphors in biomedical studies. Hence, the obtained results strongly suggest that this nanophosphor could be potentially used as a bimodal nanoprobe for high-contrast in vitro bio-imaging of HeLa cells and external magnetic field tracking applications of luminomagnetic nanophosphors using permanent magnet.- Published
- 2016
- Full Text
- View/download PDF
44. Probing highly luminescent europium-doped lanthanum orthophosphate nanorods for strategic applications.
- Author
-
Saraf M, Kumar P, Kedawat G, Dwivedi J, Vithayathil SA, Jaiswal N, Kaipparettu BA, and Gupta BK
- Subjects
- Cell Line, Tumor, Humans, Molecular Imaging, Biocompatible Materials chemistry, Drug Design, Europium chemistry, Luminescent Agents chemistry, Nanotubes chemistry, Phosphates chemistry
- Abstract
Herein we have established a strategy for the synthesis of highly luminescent and biocompatible europium-doped lanthanum orthophosphate (La0.85PO4Eu0.15(3+)) nanorods. The structure and morphogenesis of these nanorods have been probed by XRD, SEM, and TEM/HRTEM techniques. The XRD result confirms that the as-synthesized nanorods form in a monazite phase with a monoclinic crystal structure. Furthermore, the surface morphology shows that the synthesized nanorods have an average diameter of ∼90 nm and length of ∼2 μm. The HRTEM images show clear lattice fringes that support the presence of better crystal quality and enhanced photoluminescence hypersensitive red emission at 610 nm ((5)D0-(7)F2) upon 394 nm wavelength excitation. Furthermore, time-resolved spectroscopy and an MTT assay of these luminescent nanorods demonstrate a photoluminescent decay time of milliseconds with nontoxic behavior. Hence, these obtained results suggest that the as-synthesized luminescent nanorods could be potentially used in invisible security ink and high-contrast bioimaging applications.
- Published
- 2015
- Full Text
- View/download PDF
45. The somatic genomic landscape of chromophobe renal cell carcinoma.
- Author
-
Davis CF, Ricketts CJ, Wang M, Yang L, Cherniack AD, Shen H, Buhay C, Kang H, Kim SC, Fahey CC, Hacker KE, Bhanot G, Gordenin DA, Chu A, Gunaratne PH, Biehl M, Seth S, Kaipparettu BA, Bristow CA, Donehower LA, Wallen EM, Smith AB, Tickoo SK, Tamboli P, Reuter V, Schmidt LS, Hsieh JJ, Choueiri TK, Hakimi AA, Chin L, Meyerson M, Kucherlapati R, Park WY, Robertson AG, Laird PW, Henske EP, Kwiatkowski DJ, Park PJ, Morgan M, Shuch B, Muzny D, Wheeler DA, Linehan WM, Gibbs RA, Rathmell WK, and Creighton CJ
- Subjects
- Base Sequence, Chromosome Breakpoints, Chromosome Deletion, Chromosomes, Human genetics, DNA Copy Number Variations, DNA Methylation, DNA Mutational Analysis, DNA, Mitochondrial genetics, Exome, Genome, Human, Humans, Molecular Sequence Data, Promoter Regions, Genetic, Telomerase genetics, Transcriptome, Carcinoma, Renal Cell genetics, Kidney Neoplasms genetics
- Abstract
We describe the landscape of somatic genomic alterations of 66 chromophobe renal cell carcinomas (ChRCCs) on the basis of multidimensional and comprehensive characterization, including mtDNA and whole-genome sequencing. The result is consistent that ChRCC originates from the distal nephron compared with other kidney cancers with more proximal origins. Combined mtDNA and gene expression analysis implicates changes in mitochondrial function as a component of the disease biology, while suggesting alternative roles for mtDNA mutations in cancers relying on oxidative phosphorylation. Genomic rearrangements lead to recurrent structural breakpoints within TERT promoter region, which correlates with highly elevated TERT expression and manifestation of kataegis, representing a mechanism of TERT upregulation in cancer distinct from previously observed amplifications and point mutations., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
46. Crosstalk from non-cancerous mitochondria can inhibit tumor properties of metastatic cells by suppressing oncogenic pathways.
- Author
-
Kaipparettu BA, Ma Y, Park JH, Lee TL, Zhang Y, Yotnda P, Creighton CJ, Chan WY, and Wong LJ
- Subjects
- Adenosine Triphosphate metabolism, Animals, Antibiotics, Antineoplastic pharmacology, Cell Hypoxia, Cell Line, Cell Line, Tumor, Cell Nucleus metabolism, Cell Survival drug effects, Cell Survival genetics, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, DNA, Mitochondrial metabolism, Doxorubicin pharmacology, Female, Heterografts, Humans, Hybrid Cells drug effects, Hybrid Cells metabolism, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Mice, Nude, Mitochondria metabolism, Neoplasm Metastasis, Oligonucleotide Array Sequence Analysis, Reactive Oxygen Species metabolism, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction genetics, Transcriptome drug effects, Transcriptome genetics, Cell Nucleus genetics, DNA, Mitochondrial genetics, Mammary Neoplasms, Experimental genetics, Mitochondria genetics
- Abstract
Mitochondrial-nucleus cross talks and mitochondrial retrograde regulation can play a significant role in cellular properties. Transmitochondrial cybrid systems (cybrids) are an excellent tool to study specific effects of altered mitochondria under a defined nuclear background. The majority of the studies using the cybrid model focused on the significance of specific mitochondrial DNA variations in mitochondrial function or tumor properties. However, most of these variants are benign polymorphisms without known functional significance. From an objective of rectifying mitochondrial defects in cancer cells and to establish mitochondria as a potential anticancer drug target, understanding the role of functional mitochondria in reversing oncogenic properties under a cancer nuclear background is very important. Here we analyzed the potential reversal of oncogenic properties of a highly metastatic cell line with the introduction of non-cancerous mitochondria. Cybrids were established by fusing the mitochondria DNA depleted 143B TK- ρ0 cells from an aggressive osteosarcoma cell line with mitochondria from benign breast epithelial cell line MCF10A, moderately metastatic breast cancer cell line MDA-MB-468 and 143B cells. In spite of the uniform cancerous nuclear background, as observed with the mitochondria donor cells, cybrids with benign mitochondria showed high mitochondrial functional properties including increased ATP synthesis, oxygen consumption and respiratory chain activities compared to cybrids with cancerous mitochondria. Interestingly, benign mitochondria could reverse different oncogenic characteristics of 143B TK(-) cell including cell proliferation, viability under hypoxic condition, anti-apoptotic properties, resistance to anti-cancer drug, invasion, and colony formation in soft agar, and in vivo tumor growth in nude mice. Microarray analysis suggested that several oncogenic pathways observed in cybrids with cancer mitochondria are inhibited in cybrids with non-cancerous mitochondria. These results suggest the critical oncogenic regulation by mitochondrial-nuclear cross talk and highlights rectifying mitochondrial functional properties as a promising target in cancer therapy.
- Published
- 2013
- Full Text
- View/download PDF
47. Highly luminescent-paramagnetic nanophosphor probes for in vitro high-contrast imaging of human breast cancer cells.
- Author
-
Gupta BK, Narayanan TN, Vithayathil SA, Lee Y, Koshy S, Reddy AL, Saha A, Shanker V, Singh VN, Kaipparettu BA, Martí AA, and Ajayan PM
- Subjects
- Cell Line, Tumor, Humans, Microscopy, Electron, Transmission, Optical Imaging, X-Ray Diffraction, Breast Neoplasms pathology, Europium chemistry, Luminescent Agents chemistry, Metal Nanoparticles chemistry, Yttrium chemistry
- Abstract
Highly luminescent-paramagnetic nanophosphors have a seminal role in biotechnology and biomedical research due to their potential applications in biolabeling, bioimaging, and drug delivery. Herein, the synthesis of high-quality, ultrafine, europium-doped yttrium oxide nanophosphors (Y(1.9)O(3):Eu(0.1)(3+)) using a modified sol-gel technique is reported and in vitro fluorescence imaging studies are demonstrated in human breast cancer cells. These highly luminescent nanophosphors with an average particle size of ≈6 nm provide high-contrast optical imaging and decreased light scattering. In vitro cellular uptake is shown by fluorescence microscopy, which visualizes the characteristic intense hypersensitive red emission of Eu(3+) peaking at 610 nm ((5)D(0)-(7)F(2)) upon 246 nm UV light excitation. No apparent cytotoxicity is observed. Subsequently, time-resolved emission spectroscopy and SQUID magnetometry measurements demonstrate a photoluminescence decay time in milliseconds and paramagnetic behavior, which assure applications of the nanophosphors in biomedical studies., (Copyright © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2012
- Full Text
- View/download PDF
48. Hybrid 2D nanomaterials as dual-mode contrast agents in cellular imaging.
- Author
-
Narayanan TN, Gupta BK, Vithayathil SA, Aburto RR, Mani SA, Taha-Tijerina J, Xie B, Kaipparettu BA, Torti SV, and Ajayan PM
- Subjects
- Cell Line, Tumor, Cell Survival drug effects, Contrast Media toxicity, Graphite chemistry, Humans, Hydrogen-Ion Concentration, Luminescent Measurements, Magnetic Resonance Imaging, Nanostructures toxicity, Oxides chemistry, Contrast Media chemistry, Molecular Imaging methods, Nanostructures chemistry
- Published
- 2012
- Full Text
- View/download PDF
49. Graphene quantum dots derived from carbon fibers.
- Author
-
Peng J, Gao W, Gupta BK, Liu Z, Romero-Aburto R, Ge L, Song L, Alemany LB, Zhan X, Gao G, Vithayathil SA, Kaipparettu BA, Marti AA, Hayashi T, Zhu JJ, and Ajayan PM
- Subjects
- Antineoplastic Agents pharmacology, Carbon pharmacology, Carbon Fiber, Cell Line, Tumor, Cell Survival drug effects, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Fluorescence, Graphite pharmacology, Humans, Particle Size, Solubility, Structure-Activity Relationship, Surface Properties, Antineoplastic Agents chemistry, Carbon chemistry, Graphite chemistry, Quantum Dots
- Abstract
Graphene quantum dots (GQDs), which are edge-bound nanometer-size graphene pieces, have fascinating optical and electronic properties. These have been synthesized either by nanolithography or from starting materials such as graphene oxide (GO) by the chemical breakdown of their extended planar structure, both of which are multistep tedious processes. Here, we report that during the acid treatment and chemical exfoliation of traditional pitch-based carbon fibers, that are both cheap and commercially available, the stacked graphitic submicrometer domains of the fibers are easily broken down, leading to the creation of GQDs with different size distribution in scalable amounts. The as-produced GQDs, in the size range of 1-4 nm, show two-dimensional morphology, most of which present zigzag edge structure, and are 1-3 atomic layers thick. The photoluminescence of the GQDs can be tailored through varying the size of the GQDs by changing process parameters. Due to the luminescence stability, nanosecond lifetime, biocompatibility, low toxicity, and high water solubility, these GQDs are demonstrated to be excellent probes for high contrast bioimaging and biosensing applications., (© 2012 American Chemical Society)
- Published
- 2012
- Full Text
- View/download PDF
50. Transmitochondrial cybrids: tools for functional studies of mutant mitochondria.
- Author
-
Vithayathil SA, Ma Y, and Kaipparettu BA
- Subjects
- DNA, Mitochondrial genetics, DNA, Mitochondrial metabolism, Ethidium metabolism, Genetic Markers genetics, Humans, Transfection, Uridine metabolism, Cell Fusion methods, Mitochondria genetics, Mutation
- Abstract
Mitochondrial functions are controlled by both mitochondrial DNA (mtDNA) and nuclear DNA. Hence, it is difficult to identify whether mitochondrial or nuclear genome is responsible for a particular mitochondrial defect. Cybrid is a useful tool to overcome this difficulty, where we can compare mitochondria from different sources in a defined nuclear background. Cybrids are constructed by fusing enucleated cells harboring wild type or altered mtDNA of interest with ρ(0) cells (cells lacking mtDNA) in which the endogenous mtDNA has been depleted. Therefore, cybrids are very useful in studying consequences of mtDNA alterations or other mitochondrial defects at the cellular level by excluding the influence of nuclear DNA mutations.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.