74 results on '"Putluri V"'
Search Results
2. Androgen-regulated metabolome in prostate cancer.
- Author
-
Putluri, N., primary, Zhang, Y., additional, Putluri, V., additional, Vareed, S., additional, Vasu, V. T., additional, Fischer, S. M., additional, Chad, C., additional, and Sreekumar, A., additional
- Published
- 2011
- Full Text
- View/download PDF
3. Use of metabolomic profiling to identify potential markers and mechanism for bladder cancer progression.
- Author
-
Putluri, N., primary, Vasu, V. T., additional, Shojaie, A., additional, Thangjam, G. S., additional, Vareed, S., additional, Tallman, C. T., additional, Putluri, V., additional, Butler, C., additional, Giri, J. G., additional, and Sana, T. R., additional
- Published
- 2011
- Full Text
- View/download PDF
4. Multiplex panel of metabolomic markers for early detection of prostate cancer.
- Author
-
Vareed, S. K., primary, Siddiqui, J., additional, Putluri, V., additional, Putluri, N., additional, Rajendiran, T. M., additional, Wei, J. T., additional, Terris, M. K., additional, Chinnaiyan, A., additional, Michailidis, G., additional, and Sreekumar, A., additional
- Published
- 2011
- Full Text
- View/download PDF
5. Tobacco smoke exposure is a driver of altered oxidative stress response and immunity in head and neck cancer.
- Author
-
Li Y, Yadollahi P, Essien F, Putluri V, Chandra S, Kami Reddy KR, Kamal A, Putluri N, Abdurrahman LM, Ruiz-Echartea E, Ernste K, Trivedi A, Vazquez-Perez J, Hudson WH, Decker W, Patel R, Osman AA, Kheradmand F, Lai SY, Myers JN, Skinner HD, Coarfa C, Lee K, Jain A, Malovannaya A, Frederick MJ, and Sandulache VC
- Abstract
Purpose: Exposomes are critical drivers of carcinogenesis. However, how they modulate tumor behavior remains unclear. Extensive clinical data link cigarette smoke as a key exposome that promotes aggressive tumors, higher rates of metastasis, reduced response to chemoradiotherapy, and suppressed anti-tumor immunity. We sought to determine whether smoke itself can modulate aggressive tumor behavior in head and neck squamous cell carcinoma (HNSCC) through reprogramming the cellular reductive state., Experimental Design: Using established human and murine HNSCC cell lines and syngeneic mouse models, we utilized conventional western blotting, steady state and flux metabolomics, RNA sequencing, quantitative proteomics and flow cytometry to analyze the impact of smoke exposure on HNSCC tumor biology., Results: Cigarette smoke persistently activated Nrf2 target genes essential for maintenance of the cellular reductive state and survival under conditions of increased oxidative stress in HNSCC regardless of HPV status. In contrast to e-cigarette vapor, conventional cigarette smoke mobilizes cellular metabolism toward oxidative stress adaptation, resulting in development of cross-resistance to cisplatin. In parallel, smoke exposure modulates both expression of PDL1 and the secretory phenotype of HNSCC cells through activation of NF-κB resulting in an altered tumor immune microenvironment (TIME) in syngeneic mouse models and altered PBMC differentiation that includes downregulated expression of antigen presentation and costimulatory genes in myeloid cells., Conclusion: Cigarette smoke exposome is a potent activator of the Nrf2 pathway and is a likely primary trigger for the tripartite phenotype of aggressive HNSCC consisting of: 1) reduced chemotherapy sensitivity, 2) enhanced metastatic potential and 3) suppressed anti-tumor immunity., Statement of Significance: The smoke exposome drives aggressive tumor behavior, treatment resistance and suppressed immunity through coordinated metabolic reprogramming. Successfully targeting this adaptation is critical to improving survival in smokers with head and neck cancer.
- Published
- 2024
- Full Text
- View/download PDF
6. 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
7. Metabolomic Profiles in Jamaican Children With and Without Autism Spectrum Disorder.
- Author
-
Yazdani A, Samms-Vaughan M, Saroukhani S, Bressler J, Hessabi M, Tahanan A, Grove ML, Gangnus T, Putluri V, Kamal AHM, Putluri N, Loveland KA, and Rahbar MH
- Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with a wide range of behavioral and cognitive impairments. While genetic and environmental factors are known to contribute to its etiology, metabolic perturbations associated with ASD, which can potentially connect genetic and environmental factors, remain poorly understood. Therefore, we conducted a metabolomic case-control study and performed a comprehensive analysis to identify significant alterations in metabolite profiles between children with ASD and typically developing (TD) controls in order to identify specific metabolites that may serve as biomarkers for the disorder. We conducted metabolomic profiling on plasma samples from participants in the second phase of Epidemiological Research on Autism in Jamaica, an age and sex-matched cohort of 200 children with ASD and 200 TD controls (2-8 years old). Using high-throughput liquid chromatography-mass spectrometry techniques, we performed a targeted metabolite analysis, encompassing amino acids, lipids, carbohydrates, and other key metabolic compounds. After quality control and missing data imputation, we performed univariable and multivariable analysis using normalized metabolites while adjusting for covariates, age, sex, socioeconomic status, and child's parish of birth. Our findings revealed unique metabolic patterns in children with ASD for four metabolites compared to TD controls. Notably, three metabolites were fatty acids, including myristoleic acid, eicosatetraenoic acid, and octadecenoic acid. The amino acid sarcosine exhibited a significant association with ASD. These findings highlight the role of metabolites in the etiology of ASD and suggest opportunities for the development of targeted interventions., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
8. Phosphoglycerate mutase regulates Treg differentiation through control of serine synthesis and one-carbon metabolism.
- Author
-
Godfrey WH, Cho K, Deng X, Ambati CSR, Putluri V, Mostafa Kamal AH, Putluri N, and Kornberg MD
- Abstract
The differentiation and suppressive functions of regulatory CD4 T cells (Tregs) are supported by a broad array of metabolic changes, providing potential therapeutic targets for immune modulation. In this study, we focused on the regulatory role of glycolytic enzymes in Tregs and identified phosphoglycerate mutase (PGAM) as being differentially overexpressed in Tregs and associated with a highly suppressive phenotype. Pharmacologic or genetic inhibition of PGAM reduced Treg differentiation and suppressive function while reciprocally inducing markers of a pro-inflammatory, T helper 17 (Th17)-like state. The regulatory role of PGAM was dependent on the contribution of 3-phosphoglycerate (3PG), the PGAM substrate, to de novo serine synthesis. Blocking de novo serine synthesis from 3PG reversed the effect of PGAM inhibition on Treg polarization, while exogenous serine directly inhibited Treg polarization. Additionally, altering serine levels in vivo with a serine/glycine-free diet increased peripheral Tregs and attenuated autoimmunity in a murine model of multiple sclerosis. Mechanistically, we found that serine limits Treg polarization by contributing to one-carbon metabolism and methylation of Treg-associated genes. Inhibiting one-carbon metabolism increased Treg polarization and suppressive function both in vitro and in vivo in a murine model of autoimmune colitis. Our study identifies a novel physiologic role for PGAM and highlights the metabolic interconnectivity between glycolysis, serine synthesis, one-carbon metabolism, and epigenetic regulation of Treg differentiation and suppressive function.
- Published
- 2024
- Full Text
- View/download PDF
9. 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
10. 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
11. 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
12. Role of adenosine deaminase in prostate cancer progression.
- Author
-
Charles C, Lloyd SM, Piyarathna DWB, Gohlke J, Rasaily U, Putluri V, Simons BW, Zaslavsky A, Nallandhighal S, Michailidis G, Palanisamy N, Navone N, Jones JA, Ittmann MM, Putluri N, Rowley DR, Salami SS, Palapattu GS, and Sreekumar A
- Abstract
Prostate cancer (PCa) is the second most common cancer and constitutes about 14.7% of total cancer cases. PCa is highly prevalent and more aggressive in African-American (AA) men than in European-American (EA) men. PCa tends to be highly heterogeneous, and its complex biology is not fully understood. We use metabolomics to better understand the mechanisms behind PCa progression and disparities in its clinical outcome. Adenosine deaminase (ADA) is a key enzyme in the purine metabolic pathway; it was found to be upregulated in PCa and is associated with higher-grade PCa and poor disease-free survival. The inosine-to-adenosine ratio, which is a surrogate for ADA activity was high in PCa patient urine and higher in AA PCa compared to EA PCa. To understand the significance of high ADA in PCa, we established ADA overexpression models and performed various in vitro and in vivo studies. Our studies have revealed that an acute increase in ADA expression during later stages of tumor development enhances in vivo growth in multiple pre-clinical models. Further analysis revealed that mTOR signaling activation could be associated with this tumor growth. Chronic ADA overexpression shows alterations in the cells' adhesion machinery and a decrease in cells' ability to adhere to the extracellular matrix in vitro . Losing cell-matrix interaction is critical for metastatic dissemination which suggests that ADA could potentially be involved in promoting metastasis. This is supported by the association of higher ADA expression with higher-grade tumors and poor patient survival. Overall, our findings suggest that increased ADA expression may promote PCa progression, specifically tumor growth and metastatic dissemination., Competing Interests: None., (AJCEU Copyright © 2023.)
- Published
- 2023
13. 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
14. Evolution of cisplatin resistance through coordinated metabolic reprogramming of the cellular reductive state.
- Author
-
Yu W, Chen Y, Putluri N, Osman A, Coarfa C, Putluri V, Kamal AHM, Asmussen JK, Katsonis P, Myers JN, Lai SY, Lu W, Stephan CC, Powell RT, Johnson FM, Skinner HD, Kazi J, Ahmed KM, Hu L, Threet A, Meyer MD, Bankson JA, Wang T, Davis J, Parker KR, Harris MA, Baek ML, Echeverria GV, Qi X, Wang J, Frederick AI, Walsh AJ, Lichtarge O, Frederick MJ, and Sandulache VC
- Subjects
- Humans, Cisplatin metabolism, Squamous Cell Carcinoma of Head and Neck, Kelch-Like ECH-Associated Protein 1 genetics, NF-E2-Related Factor 2 genetics, Drug Resistance, Neoplasm genetics, Cell Line, Tumor, Glucose, Head and Neck Neoplasms, Antineoplastic Agents pharmacology
- Abstract
Background: Cisplatin (CDDP) is a mainstay treatment for advanced head and neck squamous cell carcinomas (HNSCC) despite a high frequency of innate and acquired resistance. We hypothesised that tumours acquire CDDP resistance through an enhanced reductive state dependent on metabolic rewiring., Methods: To validate this model and understand how an adaptive metabolic programme might be imprinted, we performed an integrated analysis of CDDP-resistant HNSCC clones from multiple genomic backgrounds by whole-exome sequencing, RNA-seq, mass spectrometry, steady state and flux metabolomics., Results: Inactivating KEAP1 mutations or reductions in KEAP1 RNA correlated with Nrf2 activation in CDDP-resistant cells, which functionally contributed to resistance. Proteomics identified elevation of downstream Nrf2 targets and the enrichment of enzymes involved in generation of biomass and reducing equivalents, metabolism of glucose, glutathione, NAD(P), and oxoacids. This was accompanied by biochemical and metabolic evidence of an enhanced reductive state dependent on coordinated glucose and glutamine catabolism, associated with reduced energy production and proliferation, despite normal mitochondrial structure and function., Conclusions: Our analysis identified coordinated metabolic changes associated with CDDP resistance that may provide new therapeutic avenues through targeting of these convergent pathways., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
15. Conjugated bile acids are nutritionally re-programmable antihypertensive metabolites.
- Author
-
Chakraborty S, Lulla A, Cheng X, Yeo JY, Mandal J, Yang T, Mei X, Saha P, Golonka RM, Yeoh BS, Mell B, Jia W, Putluri V, Piyarathna DWB, Putluri N, Sreekumar A, Meyer K, Vijay-Kumar M, and Joe B
- Subjects
- Rats, Humans, Animals, Bile Acids and Salts metabolism, Liver, Taurine metabolism, Antihypertensive Agents pharmacology, Antihypertensive Agents therapeutic use, Hypertension drug therapy, Hypertension metabolism
- Abstract
Background: Hypertension is the largest risk factor affecting global mortality. Despite available medications, uncontrolled hypertension is on the rise, whereby there is an urgent need to develop novel and sustainable therapeutics. Because gut microbiota is now recognized as an important entity in blood pressure regulation, one such new avenue is to target the gut-liver axis wherein metabolites are transacted via host-microbiota interactions. Knowledge on which metabolites within the gut-liver axis regulate blood pressure is largely unknown., Method: To address this, we analyzed bile acid profiles of human, hypertensive and germ-free rat models and report that conjugated bile acids are inversely correlated with blood pressure in humans and rats., Results: Notably intervening with taurine or tauro-cholic acid rescued bile acid conjugation and reduced blood pressure in hypertensive rats. Subsequently, untargeted metabolomics uncovered altered energy metabolism following conjugation of bile acids as a mechanism alleviating high blood pressure., Conclusion: Together this work reveals conjugated bile acids as nutritionally re-programmable anti-hypertensive metabolites., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
16. Integrative metabolomics and transcriptomics analysis reveals novel therapeutic vulnerabilities in lung cancer.
- Author
-
Thaiparambil J, Dong J, Grimm SL, Perera D, Ambati CSR, Putluri V, Robertson MJ, Patel TD, Mistretta B, Gunaratne PH, Kim MP, Yustein JT, Putluri N, Coarfa C, and El-Zein R
- Subjects
- Humans, Transcriptome, Gene Expression Profiling, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms metabolism, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Adenocarcinoma of Lung genetics
- Abstract
Background: Non-small cell lung cancer (NSCLC) comprises the majority (~85%) of all lung tumors, with lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) being the most frequently diagnosed histological subtypes. Multi-modal omics profiling has been carried out in NSCLC, but no studies have yet reported a unique metabolite-related gene signature and altered metabolic pathways associated with LUAD and LUSC., Methods: We integrated transcriptomics and metabolomics to analyze 30 human lung tumors and adjacent noncancerous tissues. Differential co-expression was used to identify modules of metabolites that were altered between normal and tumor., Results: We identified unique metabolite-related gene signatures specific for LUAD and LUSC and key pathways aberrantly regulated at both transcriptional and metabolic levels. Differential co-expression analysis revealed that loss of coherence between metabolites in tumors is a major characteristic in both LUAD and LUSC. We identified one metabolic onco-module gained in LUAD, characterized by nine metabolites and 57 metabolic genes. Multi-omics integrative analysis revealed a 28 metabolic gene signature associated with poor survival in LUAD, with six metabolite-related genes as individual prognostic markers., Conclusions: We demonstrated the clinical utility of this integrated metabolic gene signature in LUAD by using it to guide repurposing of AZD-6482, a PI3Kβ inhibitor which significantly inhibited three genes from the 28-gene signature. Overall, we have integrated metabolomics and transcriptomics analyses to show that LUAD and LUSC have distinct profiles, inferred gene signatures with prognostic value for patient survival, and identified therapeutic targets and repurposed drugs for potential use in NSCLC treatment., (© 2022 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
17. Early Systemic Glycolytic Shift After Aneurysmal Subarachnoid Hemorrhage is Associated with Functional Outcomes.
- Author
-
Gusdon AM, Fu C, Putluri V, Paz AS, Chen H, Ren X, Hassan MK, Dash P, Coarfa C, Putluri N, Choi HA, and Savarraj JPJ
- Subjects
- Humans, Glutamine, Ketoglutaric Acids, Glycolysis, Fumarates, Citrates, Subarachnoid Hemorrhage complications
- Abstract
Background: Aneurysmal subarachnoid hemorrhage (aSAH) leads to a robust systemic inflammatory response. We hypothesized that an early systemic glycolytic shift occurs after aSAH, resulting in a unique metabolic signature and affecting systemic inflammation., Methods: Control patients and patients with aSAH were analyzed. Samples from patients with aSAH were collected within 24 h of aneurysmal rupture. Mass spectrometry-based metabolomics was performed to assess relative abundance of 16 metabolites involved in the tricarboxylic acid cycle, glycolysis, and pentose phosphate pathway. Principal component analysis was used to segregate control patients from patients with aSAH. Dendrograms were developed to depict correlations between metabolites and cytokines. Analytic models predicting functional outcomes were developed, and receiver operating curves were compared., Results: A total of 122 patients with aSAH and 38 control patients were included. Patients with aSAH had higher levels of glycolytic metabolites (3-phosphoglycerate/2-phosphoglycerate, lactate) but lower levels of oxidative metabolites (succinate, malate, fumarate, and oxalate). Patients with higher clinical severity (Hunt-Hess Scale score ≥ 4) had higher levels of glyceraldehyde 3-phosphate and citrate but lower levels of α-ketoglutarate and glutamine. Principal component analysis readily segregated control patients from patients with aSAH. Correlation analysis revealed distinct clusters in control patients that were not observed in patients with aSAH. Higher levels of fumarate were associated with good functional outcomes at discharge (odds ratio [OR] 1.76, 95% confidence interval [CI] 1.15-2.82) in multivariable models, whereas higher levels of citrate were associated with poor functional outcomes at discharge (OR 0.36, 95% CI 0.16-0.73) and at 3 months (OR 0.35, 95% CI 0.14-0.81). No associations were found with delayed cerebral ischemia. Levels of α-ketoglutarate and glutamine correlated with lower levels of interleukin-8, whereas fumarate was associated with lower levels of tumor necrosis factor alpha., Conclusions: Aneurysmal subarachnoid hemorrhage results in a unique pattern of plasma metabolites, indicating a shift toward glycolysis. Higher levels of fumarate and lower levels of citrate were associated with better functional outcomes. These metabolites may represent targets to improve metabolism after aSAH., (© 2022. Springer Science+Business Media, LLC, part of Springer Nature and Neurocritical Care Society.)
- Published
- 2022
- Full Text
- View/download PDF
18. Metabolome and microbiome multi-omics integration from a murine lung inflammation model of bronchopulmonary dysplasia.
- Author
-
El Saie A, Fu C, Grimm SL, Robertson MJ, Hoffman K, Putluri V, Ambati CSR, Putluri N, Shivanna B, Coarfa C, and Pammi M
- Subjects
- Animals, Infant, Newborn, Humans, Mice, Animals, Newborn, Dysbiosis, Lipopolysaccharides metabolism, Multiomics, Infant, Premature, Lung metabolism, Inflammation metabolism, Metabolome, Disease Models, Animal, Bronchopulmonary Dysplasia etiology, Hyperoxia complications, Hyperoxia metabolism, Pneumonia metabolism, Microbiota
- Abstract
Background: Respiratory tract microbial dysbiosis can exacerbate inflammation and conversely inflammation may cause dysbiosis. Dysbiotic microbiome metabolites may lead to bronchopulmonary dysplasia (BPD). Hyperoxia and lipopolysaccharide (LPS) interaction alters lung microbiome and metabolome, mediating BPD lung injury sequence., Methods: C57BL6/J mice were exposed to 21% (normoxia) or 70% (hyperoxia) oxygen during postnatal days (PND) 1-14. Pups were injected with LPS (6 mg/kg) or equal PBS volume, intraperitoneally on PND 3, 5, and 7. At PND14, the lungs were collected for microbiome and metabolomic analyses (n = 5/group)., Results: Microbiome alpha and beta diversity were similar between groups. Metabolic changes included hyperoxia 31 up/18 down, LPS 7 up/4 down, exposure interaction 8. Hyperoxia increased Intestinimonas abundance, whereas LPS decreased Clostridiales, Dorea, and Intestinimonas; exposure interaction affected Blautia. Differential co-expression analysis on multi-omics data identified exposure-altered modules. Hyperoxia metabolomics response was integrated with a published matching transcriptome, identifying four induced genes (ALDOA, GAA, NEU1, RENBP), which positively correlated with BPD severity in a published human newborn cohort., Conclusions: We report hyperoxia and LPS lung microbiome and metabolome signatures in a clinically relevant BPD model. We identified four genes correlating with BPD status in preterm infants that are promising targets for therapy and prevention., Impact: Using multi-omics, we identified and correlated key biomarkers of hyperoxia and LPS on murine lung micro-landscape and examined their potential clinical implication, which shows strong clinical relevance for future research. Using a double-hit model of clinical relevance to bronchopulmonary dysplasia, we are the first to report integrated metabolomic/microbiome landscape changes and identify novel disease biomarker candidates., (© 2022. The Author(s), under exclusive licence to the International Pediatric Research Foundation, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
19. IDH1 p.R132H ctDNA and D-2-hydroxyglutarate as CSF biomarkers in patients with IDH-mutant gliomas.
- Author
-
Fujita Y, Nunez-Rubiano L, Dono A, Bellman A, Shah M, Rodriguez JC, Putluri V, Kamal AHM, Putluri N, Riascos RF, Zhu JJ, Esquenazi Y, and Ballester LY
- Subjects
- Biomarkers, Brain Neoplasms cerebrospinal fluid, Brain Neoplasms diagnosis, Glutarates, Humans, Mutation, Prospective Studies, Circulating Tumor DNA cerebrospinal fluid, Glioma diagnosis, Isocitrate Dehydrogenase cerebrospinal fluid, Isocitrate Dehydrogenase genetics
- Abstract
Introduction: We aimed to evaluate IDH1 p.R132H mutation and 2-hydroxyglutarate (2HG) in cerebrospinal fluid (CSF) as biomarkers for patients with IDH-mutant gliomas., Methods: CSF was collected from patients with infiltrating glioma, and 2HG levels were measured by liquid chromatography-mass spectrometry. IDH1 p.R132H mutant allele frequency (MAF) in CSF-ctDNA was measured by digital droplet PCR (ddPCR). Tumor volume was measured from standard-of-care magnetic resonance images., Results: The study included 48 patients, 6 with IDH-mutant and 42 with IDH-wildtype gliomas, and 57 samples, 9 from the patients with IDH-mutant and 48 from the patients with IDH-wildtype gliomas. ctDNA was detected in 7 of the 9 samples from patients with IDH-mutant glioma, and IDH1 p.R132H mutation was detected in 5 of the 7 samples. The MAF ranged from 0.3 to 39.95%. Total 2HG level, D-2HG level, and D/L-2HG ratio in CSF were significantly higher in patients with IDH-mutant gliomas than in patients with IDH-wildtype gliomas. D-2HG level and D/L-2HG ratio correlated with total tumor volume in patients with IDH-mutant gliomas but not in patients with IDH-wildtype gliomas., Conclusion: Our results suggest that detection of IDH1 p.R132H mutation by ddPCR and increased D-2HG level in CSF may help identify IDH-mutant gliomas. Our results also suggest that D-2HG level and D/L-2HG ratio correlate with tumor volume in patients with IDH-mutant gliomas. Further prospective studies with larger cohorts are needed to validate these findings., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
20. Glutathione peroxidase 2 is a metabolic driver of the tumor immune microenvironment and immune checkpoint inhibitor response.
- Author
-
Ahmed KM, Veeramachaneni R, Deng D, Putluri N, Putluri V, Cardenas MF, Wheeler DA, Decker WK, Frederick AI, Kazi S, Sikora AG, Sandulache VC, and Frederick MJ
- Subjects
- Animals, Dinoprostone, Glutathione Peroxidase genetics, Mice, Tumor Microenvironment, Glutathione Peroxidase metabolism, Immune Checkpoint Inhibitors, NF-E2-Related Factor 2 metabolism
- Abstract
Background: The existence of immunologically 'cold tumors' frequently found across a wide spectrum of tumor types represents a significant challenge for cancer immunotherapy. Cold tumors have poor baseline pan-leukocyte infiltration, including a low prevalence of cytotoxic lymphocytes, and not surprisingly respond unfavorably to immune checkpoint (IC) inhibitors. We hypothesized that cold tumors harbor a mechanism of immune escape upstream and independent of ICs that may be driven by tumor biology rather than differences in mutational neoantigen burden., Methods: Using a bioinformatic approach to analyze TCGA (The Cancer Genome Atlas) RNA sequencing data we identified genes upregulated in cold versus hot tumors across four different smoking-related cancers, including squamous carcinomas from the oral cavity (OCSCC) and lung (LUSC), and adenocarcinomas of the bladder (BLCA) and lung (LUAD). Biological significance of the gene most robustly associated with a cold tumor phenotype across all four tumor types, glutathione peroxidase 2 (GPX2), was further evaluated using a combination of in silico analyses and functional genomic experiments performed both in vitro and in in vivo with preclinical models of oral cancer., Results: Elevated RNA expression of five metabolic enzymes including GPX2, aldo-keto reductase family 1 members AKR1C1, AKR1C3, and cytochrome monoxygenases (CP4F11 and CYP4F3) co-occurred in cold tumors across all four smoking-related cancers. These genes have all been linked to negative regulation of arachidonic acid metabolism-a well-established inflammatory pathway-and are also known downstream targets of the redox sensitive Nrf2 transcription factor pathway. In OCSCC, LUSC, and LUAD, GPX2 expression was highly correlated with Nrf2 activation signatures, also elevated in cold tumors. In BLCA, however, GPX2 correlated more strongly than Nrf2 signatures with decreased infiltration of multiple leukocyte subtypes. GPX2 inversely correlated with expression of multiple pro- inflammatory cytokines/chemokines and NF-kB activation in cell lines and knockdown of GPX2 led to increased secretion of prostaglandin E2 (PGE2) and interleukin-6. Conversely, GPX2 overexpression led to reduced PGE2 production in a murine OCSCC model (MOC1). GPX2 overexpressing MOC1 tumors had a more suppressive tumor immune microenvironment and responded less favorably to anti-cytotoxic T-lymphocytes-associated protein 4 IC therapy in mice., Conclusion: GPX2 overexpression represents a novel potentially targetable effector of immune escape in cold tumors., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
21. Uridine Diphosphate Glucuronosyl Transferase 2B28 (UGT2B28) Promotes Tumor Progression and Is Elevated in African American Prostate Cancer Patients.
- Author
-
Ravindran A, Krieger KL, Kaushik AK, Hovington H, Mehdi S, Piyarathna DWB, Putluri V, Basil P, Rasaily U, Gu F, Dang T, Choi JM, Sonavane R, Jung SY, Wang L, Mehra R, Weigel NL, Putluri N, Rowley DR, Palapattu GS, Guillemette C, Lacombe L, Lévesque É, and Sreekumar A
- Subjects
- Black or African American genetics, Humans, Male, Neoplastic Processes, Uridine Diphosphate, Androgens, Glucuronosyltransferase metabolism, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology
- Abstract
Prostate cancer (PCa) is the second most diagnosed cancer in the United States and is associated with metabolic reprogramming and significant disparities in clinical outcomes among African American (AA) men. While the cause is likely multi-factorial, the precise reasons for this are unknown. Here, we identified a higher expression of the metabolic enzyme UGT2B28 in localized PCa and metastatic disease compared to benign adjacent tissue, in AA PCa compared to benign adjacent tissue, and in AA PCa compared to European American (EA) PCa. UGT2B28 was found to be regulated by both full-length androgen receptor (AR) and its splice variant, AR-v7. Genetic knockdown of UGT2B28 across multiple PCa cell lines (LNCaP, LAPC-4, and VCaP), both in androgen-replete and androgen-depleted states resulted in impaired 3D organoid formation and a significant delay in tumor take and growth rate of xenograft tumors, all of which were rescued by re-expression of UGT2B28. Taken together, our findings demonstrate a key role for the UGT2B28 gene in promoting prostate tumor growth.
- Published
- 2022
- Full Text
- View/download PDF
22. Lipidomic Profiling Identifies a Novel Lipid Signature Associated with Ethnicity-Specific Disparity of Bladder Cancer.
- Author
-
Kami Reddy KR, Piyarathna DWB, Kamal AHM, Putluri V, Ravi SS, Bollag RJ, Terris MK, Lotan Y, and Putluri N
- Abstract
Bladder Cancer (BLCA) is the ninth most frequently diagnosed cancer globally and the sixth most common cancer in the US. African Americans (AA) exhibit half the BLCA incidence compared to European Americans (EA), but they have a 70% higher risk of cancer-related death; unfortunately, this disparity in BLCA mortality remains poorly understood. In this study, we have used an ethnicity-balanced cohort for unbiased lipidomics profiling to study the changes in the lipid fingerprint for AA and EA BLCA tissues collected from similar geographical regions to determine a signature of ethnic-specific alterations. We identified 86 lipids significantly altered between self-reported AA and EA BLCA patients from Augusta University (AU) cohort. The majority of altered lipids belong to phosphatidylcholines (PCs), phosphatidylethanolamines (PEs), ly sophosphatidylcholines (lysoPCs), phosphatidylserines (PSs), and diglycerides (DGs). Interestingly, levels of four lysoPCs (lyso PCs 20:3, lyso PCs 22:1, lyso PCs 22:2, and lyso PCs 26:1) were elevated while, in contrast, the majority of the PCs were reduced in AA BLCA. Significant alterations in long-chain monounsaturated (MonoUN) and polyunsaturated (PolyUN) lipids were also observed between AA and EA BLCA tumor tissues. These first-in-field results implicate ethnic-specific lipid alterations in BLCA.
- Published
- 2022
- Full Text
- View/download PDF
23. Metabolic adaptation of ovarian tumors in patients treated with an IDO1 inhibitor constrains antitumor immune responses.
- Author
-
Odunsi K, Qian F, Lugade AA, Yu H, Geller MA, Fling SP, Kaiser JC, Lacroix AM, D'Amico L, Ramchurren N, Morishima C, Disis ML, Dennis L, Danaher P, Warren S, Nguyen VA, Ravi S, Tsuji T, Rosario S, Zha W, Hutson A, Liu S, Lele S, Zsiros E, McGray AJR, Chiello J, Koya R, Chodon T, Morrison CD, Putluri V, Putluri N, Mager DE, Gunawan R, Cheever MA, Battaglia S, and Matsuzaki J
- Subjects
- Animals, Female, Humans, Lymphocyte Activation, Mice, NAD, Tryptophan metabolism, Tumor Microenvironment, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Ovarian Neoplasms drug therapy
- Abstract
To uncover underlying mechanisms associated with failure of indoleamine 2,3-dioxygenase 1 (IDO1) blockade in clinical trials, we conducted a pilot, window-of-opportunity clinical study in 17 patients with newly diagnosed advanced high-grade serous ovarian cancer before their standard tumor debulking surgery. Patients were treated with the IDO1 inhibitor epacadostat, and immunologic, transcriptomic, and metabolomic characterization of the tumor microenvironment was undertaken in baseline and posttreatment tumor biopsies. IDO1 inhibition resulted in efficient blockade of the kynurenine pathway of tryptophan degradation and was accompanied by a metabolic adaptation that shunted tryptophan catabolism toward the serotonin pathway. This resulted in elevated nicotinamide adenine dinucleotide (NAD
+ ), which reduced T cell proliferation and function. Because NAD+ metabolites could be ligands for purinergic receptors, we investigated the impact of blocking purinergic receptors in the presence or absence of NAD+ on T cell proliferation and function in our mouse model. We demonstrated that A2a and A2b purinergic receptor antagonists, SCH58261 or PSB1115, respectively, rescued NAD+ -mediated suppression of T cell proliferation and function. Combining IDO1 inhibition and A2a/A2b receptor blockade improved survival and boosted the antitumor immune signature in mice with IDO1 overexpressing ovarian cancer. These findings elucidate the downstream adaptive metabolic consequences of IDO1 blockade in ovarian cancers that may undermine antitumor T cell responses in the tumor microenvironment.- Published
- 2022
- Full Text
- View/download PDF
24. Lipid Alterations in African American Men with Prostate Cancer.
- Author
-
Ravindran A, Piyarathna DWB, Gohlke J, Putluri V, Soni T, Lloyd S, Castro P, Pennathur S, Jones JA, Ittmann M, Putluri N, Michailidis G, Rajendiran TM, and Sreekumar A
- Abstract
African-American (AA) men are more than twice as likely to die of prostate cancer (PCa) than European American (EA) men. Previous in silico analysis revealed enrichment of altered lipid metabolic pathways in pan-cancer AA tumors. Here, we performed global unbiased lipidomics profiling on 48 matched localized PCa and benign adjacent tissues (30 AA, 24 ancestry-verified, and 18 EA, 8 ancestry verified) and quantified 429 lipids belonging to 14 lipid classes. Significant alterations in long chain polyunsaturated lipids were observed between PCa and benign adjacent tissues, low and high Gleason tumors, as well as associated with early biochemical recurrence, both in the entire cohort, and within AA patients. Alterations in cholesteryl esters, and phosphatidyl inositol classes of lipids delineated AA and EA PCa, while the levels of lipids belonging to triglycerides, phosphatidyl glycerol, phosphatidyl choline, phosphatidic acid, and cholesteryl esters distinguished AA and EA PCa patients with biochemical recurrence. These first-in-field results implicate lipid alterations as biological factors for prostate cancer disparities.
- Published
- 2021
- Full Text
- View/download PDF
25. Role of Human NADPH Quinone Oxidoreductase (NQO1) in Oxygen-Mediated Cellular Injury and Oxidative DNA Damage in Human Pulmonary Cells.
- Author
-
Burke R, Chu C, Zhou GD, Putluri V, Putluri N, Stading RE, Couroucli X, Lingappan K, and Moorthy B
- Subjects
- Humans, Lung pathology, Lung metabolism, Lung physiopathology, NAD(P)H Dehydrogenase (Quinone) metabolism, Oxidative Stress
- Abstract
Supplemental oxygen administration is frequently used in premature infants and adults with pulmonary insufficiency. NADPH quinone oxidoreductase (NQO1) protects cells from oxidative injury by decreasing reactive oxygen species (ROS). In this investigation, we tested the hypothesis that overexpression of NQO1 in BEAS-2B cells will mitigate cell injury and oxidative DNA damage caused by hyperoxia and that A-1221C single nucleotide polymorphism (SNP) in the NQO1 promoter would display altered susceptibility to hyperoxia-mediated toxicity. Using stable transfected BEAS-2B cells, we demonstrated that hyperoxia decreased cell viability in control cells (Ctr), but this effect was differentially mitigated in cells overexpressing NQO1 under the regulation of the CMV viral promoter, the wild-type NQO1 promoter ( NQO1-NQO1 ), or the NQO1 promoter carrying the SNP. Interestingly, hyperoxia decreased the formation of bulky oxidative DNA adducts or 8-hydroxy-2'-deoxyguanosine (8-OHdG) in Ctr cells. qPCR studies showed that mRNA levels of CYP1A1 and NQO1 were inversely related to DNA adduct formation, suggesting the protective role of these enzymes against oxidative DNA injury. In SiRNA experiments entailing the NQO1-NQO1 promoter, hyperoxia caused decreased cell viability, and this effect was potentiated in cells treated with CYP1A1 siRNA. We also found that hyperoxia caused a marked induction of DNA repair genes DDB2 and XPC in Ctr cells, supporting the idea that hyperoxia in part caused attenuation of bulky oxidative DNA lesions by enhancing nucleotide excision repair (NER) pathways. In summary, our data support a protective role for human NQO1 against oxygen-mediated toxicity and oxidative DNA lesions in human pulmonary cells, and protection against toxicity was partially lost in SNP cells. Moreover, we also demonstrate a novel protective role for CYP1A1 in the attenuation of oxidative cells and DNA injury. Future studies on the mechanisms of attenuation of oxidative injury by NQO1 should help in developing novel approaches for the prevention/treatment of ARDS in humans., Competing Interests: The authors declare that there is no conflict of interest regarding the publication of this paper., (Copyright © 2021 Rebecca Burke et al.)
- Published
- 2021
- Full Text
- View/download PDF
26. Thioredoxin reductase is a major regulator of metabolism in leukemia cells.
- Author
-
Karunanithi S, Liu R, Hou Y, Gonzalez G, Oldford N, Roe AJ, Idipilly N, Gupta K, Amara CS, Putluri S, Lee GK, Valentin-Goyco J, Stetson L, Moreton SA, Putluri V, Kavuri SM, Saunthararajah Y, de Lima M, Tochtrop GP, Putluri N, and Wald DN
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Cell Proliferation, Glycolysis, Pentose Phosphate Pathway, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute genetics, Thioredoxin-Disulfide Reductase metabolism, Thioredoxin-Disulfide Reductase genetics
- Abstract
Despite the fact that AML is the most common acute leukemia in adults, patient outcomes are poor necessitating the development of novel therapies. We identified that inhibition of Thioredoxin Reductase (TrxR) is a promising strategy for AML and report a highly potent and specific inhibitor of TrxR, S-250. Both pharmacologic and genetic inhibition of TrxR impairs the growth of human AML in mouse models. We found that TrxR inhibition leads to a rapid and marked impairment of metabolism in leukemic cells subsequently leading to cell death. TrxR was found to be a major and direct regulator of metabolism in AML cells through impacts on both glycolysis and the TCA cycle. Studies revealed that TrxR directly regulates GAPDH leading to a disruption of glycolysis and an increase in flux through the pentose phosphate pathway (PPP). The combined inhibition of TrxR and the PPP led to enhanced leukemia growth inhibition. Overall, TrxR abrogation, particularly with S-250, was identified as a promising strategy to disrupt AML metabolism., (© 2021. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)
- Published
- 2021
- Full Text
- View/download PDF
27. Cannabinoid Receptor Activation on Haematopoietic Cells and Enterocytes Protects against Colitis.
- Author
-
Becker W, Alrafas HR, Busbee PB, Walla MD, Wilson K, Miranda K, Cai G, Putluri V, Putluri N, Nagarkatti M, and Nagarkatti PS
- Subjects
- Animals, Cannabinoid Receptor Agonists pharmacology, Colonoscopy methods, Drug Monitoring, Gastrointestinal Microbiome drug effects, Gastrointestinal Microbiome immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Protective Agents pharmacology, Cannabidiol pharmacology, Colitis chemically induced, Colitis metabolism, Colitis pathology, Colitis therapy, Dronabinol pharmacology, Enterocytes drug effects, Enterocytes metabolism, Enterocytes pathology, Immunity, Cellular drug effects, Immunity, Cellular physiology, Receptor, Cannabinoid, CB2 genetics, Receptor, Cannabinoid, CB2 metabolism
- Abstract
Background and Aims: Cannabinoid receptor [CB] activation can attenuate inflammatory bowel disease [IBD] in experimental models and human cohorts. However, the roles of the microbiome, metabolome, and the respective contributions of haematopoietic and non-haematopoietic cells in the anti-colitic effects of cannabinoids have yet to be determined., Methods: Female C57BL/6 mice were treated with either cannabidiol [CBD], Δ 9-tetrahydrocannabinol [THC], a combination of CBD and THC, or vehicle, in several models of chemically induced colitis. Clinical parameters of colitis were assessed by colonoscopy, histology, flow cytometry, and detection of serum biomarkers; single-cell RNA sequencing and qRT-PCR were used to evaluate the effects of cannabinoids on enterocytes. Immune cell transfer from CB2 knockout mice was used to evaluate the contribution of haematopoietic and non-haematopoietic cells to colitis protection., Results: We found that THC prevented colitis and that CBD, at the dose tested, provided little benefit to the amelioration of colitis, nor when added synergistically with THC. THC increased colonic barrier integrity by stimulating mucus and tight junction and antimicrobial peptide production, and these effects were specific to the large intestine. THC increased colonic Gram-negative bacteria, but the anti-colitic effects of THC were independent of the microbiome. THC acted both on immune cells via CB2 and on enterocytes, to attenuate colitis., Conclusions: Our findings demonstrate how cannabinoid receptor activation on both immune cells and colonocytes is critical to prevent colonic inflammation. These studies also suggest how cannabinoid receptor activation can be used as a preventive and therapeutic modality against colitis., (© The Author(s) 2020. Published by Oxford University Press on behalf of European Crohn’s and Colitis Organisation. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
28. IDO1 Expression in Ovarian Cancer Induces PD-1 in T Cells via Aryl Hydrocarbon Receptor Activation.
- Author
-
Amobi-McCloud A, Muthuswamy R, Battaglia S, Yu H, Liu T, Wang J, Putluri V, Singh PK, Qian F, Huang RY, Putluri N, Tsuji T, Lugade AA, Liu S, and Odunsi K
- Subjects
- Animals, Binding Sites, Biomarkers, Disease Models, Animal, Disease Susceptibility, Female, Gene Expression Regulation, Neoplastic, Humans, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Mice, Ovarian Neoplasms pathology, Programmed Cell Death 1 Receptor chemistry, Programmed Cell Death 1 Receptor metabolism, Protein Binding, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Indoleamine-Pyrrole 2,3,-Dioxygenase genetics, Ovarian Neoplasms etiology, Ovarian Neoplasms metabolism, Programmed Cell Death 1 Receptor genetics, Receptors, Aryl Hydrocarbon metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
The immunoregulatory enzyme, indoleamine 2,3-dioxygenase (IDO1) and the PD-1/PD-L1 axis are potent mechanisms that impede effective anti-tumor immunity in ovarian cancer. However, whether the IDO pathway regulates PD-1 expression in T cells is currently unknown. Here we show that tumoral IDO1 expression led to profound changes in tryptophan, nicotinate/nicotinamide, and purine metabolic pathways in the ovarian tumor microenvironment, and to an increased frequency of PD-1
+ CD8+ tumor infiltrating T cells. We determined that activation of the aryl hydrocarbon receptor (AHR) by kynurenine induced PD-1 expression, and this effect was significantly abrogated by the AHR antagonist CH223191. Mechanistically, kynurenine alters chromatin accessibility in regulatory regions of T cell inhibitory receptors, allowing AHR to bind to consensus XRE motifs in the promoter region of PD-1. These results enable the design of strategies to target the IDO1 and AHR pathways for enhancing anti-tumor immunity in ovarian cancer., Competing Interests: KO is a co-founder of Tactiva Therapeutics and receives research support from AstraZeneca and Tesaro. The remaining 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 © 2021 Amobi-McCloud, Muthuswamy, Battaglia, Yu, Liu, Wang, Putluri, Singh, Qian, Huang, Putluri, Tsuji, Lugade, Liu and Odunsi.)- Published
- 2021
- Full Text
- View/download PDF
29. Isoform-specific Activities of Androgen Receptor and its Splice Variants in Prostate Cancer Cells.
- Author
-
Nagandla H, Robertson MJ, Putluri V, Putluri N, Coarfa C, and Weigel NL
- Subjects
- Alternative Splicing genetics, Cell Line, Tumor, Gene Expression Regulation, Neoplastic genetics, HEK293 Cells, Humans, Lipid Metabolism genetics, Male, Mutant Proteins physiology, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Protein Isoforms physiology, Receptors, Androgen chemistry, Receptors, Androgen genetics, Prostatic Neoplasms, Castration-Resistant genetics, Receptors, Androgen physiology
- Abstract
Androgen receptor (AR) signaling continues to drive castration-resistant prostate cancer (CRPC) in spite of androgen deprivation therapy (ADT). Constitutively active shorter variants of AR, lacking the ligand binding domain, are frequently expressed in CRPC and have emerged as a potential mechanism for prostate cancer to escape ADT. ARv7 and ARv567es are 2 of the most commonly detected variants of AR in clinical samples of advanced, metastatic prostate cancer. It is not clear if variants of AR merely act as weaker substitutes for AR or can mediate unique isoform-specific activities different from AR. In this study, we employed LNCaP prostate cancer cell lines with inducible expression of ARv7 or ARv567es to delineate similarities and differences in transcriptomics, metabolomics, and lipidomics resulting from the activation of AR, ARv7, or ARv567es. While the majority of target genes were similarly regulated by the action of all 3 isoforms, we found a clear difference in transcriptomic activities of AR versus the variants, and a few differences between ARv7 and ARv567es. Some of the target gene regulation by AR isoforms was similar in the VCaP background as well. Differences in downstream activities of AR isoforms were also evident from comparison of the metabolome and lipidome in an LNCaP model. Overall our study implies that shorter variants of AR are capable of mediating unique downstream activities different from AR and some of these are isoform specific., (© The Author(s) 2020. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
30. STAT1 Dissociates Adipose Tissue Inflammation From Insulin Sensitivity in Obesity.
- Author
-
Cox AR, Chernis N, Bader DA, Saha PK, Masschelin PM, Felix JB, Sharp R, Lian Z, Putluri V, Rajapakshe K, Kim KH, Villareal DT, Armamento-Villareal R, Wu H, Coarfa C, Putluri N, and Hartig SM
- Subjects
- Adipocytes metabolism, Animals, Energy Metabolism genetics, Energy Metabolism physiology, Female, Glucose metabolism, Homeostasis physiology, Humans, Interferon-gamma genetics, Interferon-gamma metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Obesity metabolism, RNA Interference, Receptors, Interferon genetics, Receptors, Interferon metabolism, STAT1 Transcription Factor genetics, Interferon gamma Receptor, Adipose Tissue metabolism, Gene Expression Regulation physiology, Inflammation metabolism, Insulin Resistance physiology, STAT1 Transcription Factor metabolism
- Abstract
Obesity fosters low-grade inflammation in white adipose tissue (WAT) that may contribute to the insulin resistance that characterizes type 2 diabetes. However, the causal relationship of these events remains unclear. The established dominance of STAT1 function in the immune response suggests an obligate link between inflammation and the comorbidities of obesity. To this end, we sought to determine how STAT1 activity in white adipocytes affects insulin sensitivity. STAT1 expression in WAT inversely correlated with fasting plasma glucose in both obese mice and humans. Metabolomic and gene expression profiling established STAT1 deletion in adipocytes ( STAT1
a-KO ) enhanced mitochondrial function and accelerated tricarboxylic acid cycle flux coupled with reduced fat cell size in subcutaneous WAT depots. STAT1a-KO reduced WAT inflammation, but insulin resistance persisted in obese mice. Rather, elimination of type I cytokine interferon-γ activity enhanced insulin sensitivity in diet-induced obesity. Our findings reveal a permissive mechanism that bridges WAT inflammation to whole-body insulin sensitivity., (© 2020 by the American Diabetes Association.)- Published
- 2020
- Full Text
- View/download PDF
31. Epigenetic loss of AOX1 expression via EZH2 leads to metabolic deregulations and promotes bladder cancer progression.
- Author
-
Vantaku V, Putluri V, Bader DA, Maity S, Ma J, Arnold JM, Rajapakshe K, Donepudi SR, von Rundstedt FC, Devarakonda V, Dubrulle J, Karanam B, McGuire SE, Stossi F, Jain AK, Coarfa C, Cao Q, Sikora AG, Villanueva H, Kavuri SM, Lotan Y, Sreekumar A, and Putluri N
- Subjects
- Aldehyde Oxidase metabolism, Animals, Cell Line, Tumor, Disease Progression, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Kynurenine metabolism, Male, Metabolomics, Mice, NADP metabolism, Neoplasm Invasiveness, Neoplasm Staging, Nucleotides biosynthesis, Pentose Phosphate Pathway genetics, RNA-Seq, Tissue Array Analysis, Tryptophan metabolism, Urinary Bladder Neoplasms pathology, Xenograft Model Antitumor Assays, Aldehyde Oxidase genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Urinary Bladder pathology, Urinary Bladder Neoplasms genetics
- Abstract
Advanced Bladder Cancer (BLCA) remains a clinical challenge that lacks effective therapeutic measures. Here, we show that distinct, stage-wise metabolic alterations in BLCA are associated with the loss of function of aldehyde oxidase (AOX1). AOX1 associated metabolites have a high predictive value for advanced BLCA and our findings demonstrate that AOX1 is epigenetically silenced during BLCA progression by the methyltransferase activity of EZH2. Knockdown (KD) of AOX1 in normal bladder epithelial cells re-wires the tryptophan-kynurenine pathway resulting in elevated NADP levels which may increase metabolic flux through the pentose phosphate (PPP) pathway, enabling increased nucleotide synthesis, and promoting cell invasion. Inhibition of NADP synthesis rescues the metabolic effects of AOX1 KD. Ectopic AOX1 expression decreases NADP production, PPP flux and nucleotide synthesis, while decreasing invasion in cell line models and suppressing growth in tumor xenografts. Further gain and loss of AOX1 confirm the EZH2-dependent activation, metabolic deregulation, and tumor growth in BLCA. Our findings highlight the therapeutic potential of AOX1 and provide a basis for the development of prognostic markers for advanced BLCA.
- Published
- 2020
- Full Text
- View/download PDF
32. miR-30a targets gene networks that promote browning of human and mouse adipocytes.
- Author
-
Saha PK, Hamilton MP, Rajapakshe K, Putluri V, Felix JB, Masschelin P, Cox AR, Bajaj M, Putluri N, Coarfa C, and Hartig SM
- Subjects
- Adipocytes, White metabolism, Animals, Blood Glucose metabolism, Cells, Cultured, Fatty Acid-Binding Proteins metabolism, Humans, Hypoglycemic Agents pharmacology, Insulin Resistance genetics, Metabolomics, Mice, MicroRNAs genetics, Oligopeptides metabolism, Organelle Biogenesis, PPAR gamma agonists, Rosiglitazone pharmacology, Adipocytes, Brown physiology, Gene Regulatory Networks genetics, MicroRNAs physiology
- Abstract
MicroRNA-30a (miR-30a) impacts adipocyte function, and its expression in white adipose tissue (WAT) correlates with insulin sensitivity in obesity. Bioinformatic analysis demonstrates that miR-30a expression contributes to 2% of all miRNA expression in human tissues. However, molecular mechanisms of miR-30a function in fat cells remain unclear. Here, we expanded our understanding of how miR-30a expression contributes to antidiabetic peroxisome proliferator-activated receptor-γ (PPARγ) agonist activity and metabolic functions in adipocytes. We found that WAT isolated from diabetic patients shows reduced miR-30a levels and diminished expression of the canonical PPARγ target genes ADIPOQ and FABP4 relative to lean counterparts. In human adipocytes, miR-30a required PPARγ for maximal expression, and the PPARγ agonist rosiglitazone robustly induced miR-30a but not other miR-30 family members. Transcriptional activity studies in human adipocytes also revealed that ectopic expression of miR-30a enhanced the activity of rosiglitazone coupled with higher expression of fatty acid and glucose metabolism markers. Diabetic mice that overexpress ectopic miR-30a in subcutaneous WAT display durable reductions in serum glucose and insulin levels for more than 30 days. In agreement with our in vitro findings, RNA-seq coupled with Gene Set Enrichment Analysis (GSEA) suggested that miR-30a enabled activation of the beige fat program in vivo, as evidenced by enhanced mitochondrial biogenesis and induction of UCP1 expression. Metabolomic and gene expression profiling established that the long-term effects of ectopic miR-30a expression enable accelerated glucose metabolism coupled with subcutaneous WAT hyperplasia. Together, we establish a putative role of miR-30a in mediating PPARγ activity and advancing metabolic programs of white to beige fat conversion.
- Published
- 2020
- Full Text
- View/download PDF
33. Correction: Epigenetic loss of AOX1 expression via EZH2 leads to metabolic deregulations and promotes bladder cancer progression.
- Author
-
Vantaku V, Putluri V, Bader DA, Maity S, Ma J, Arnold JM, Rajapakshe K, Donepudi SR, von Rundstedt FC, Devarakonda V, Dubrulle J, Karanam B, McGuire SE, Stossi F, Jain AK, Coarfa C, Cao Q, Sikora AG, Villanueva H, Kavuri SM, Lotan Y, Sreekumar A, and Putluri N
- Abstract
After publication of this Article, the Authors noticed errors in some of the Figures. In Figures 2e, 2f-g, 4a, 4j, 5a and 6b, unmatched β-actin was inadvertently used as loading control for the immunoblots. These have been corrected using repeat data from a similar set of samples and the revised Figures containing matched β-actin and their respective quantification data are included below. In Figure 7a, the same image was inadvertently used to represent tumors 3 and 5 in the control group. This error has been corrected using original images of tumors 3 and 5 in the control group. Additional corrections have been made in the Article and Figure legends to enhance the clarity of the description. NAD was replaced by NADP. NAD/NADP was replaced by NADP/NADPH. The description of the antibody source and dilution for the antigens PFKFB4 (Abcam, 1:1000), G6PD, and HK1 (Cell Signaling, 1:1,000) have been included in the Methods section for Western Blot. The legend for Figure 4e and 4j has been updated. The HTML and PDF versions of this Article have been corrected. The scientific conclusions of this paper have not been affected.
- Published
- 2020
- Full Text
- View/download PDF
34. Gnotobiotic Rats Reveal That Gut Microbiota Regulates Colonic mRNA of Ace2 , the Receptor for SARS-CoV-2 Infectivity.
- Author
-
Yang T, Chakraborty S, Saha P, Mell B, Cheng X, Yeo JY, Mei X, Zhou G, Mandal J, Golonka R, Yeoh BS, Putluri V, Piyarathna DWB, Putluri N, McCarthy CG, Wenceslau CF, Sreekumar A, Gewirtz AT, Vijay-Kumar M, and Joe B
- Subjects
- Angiotensin-Converting Enzyme 2, Animals, Betacoronavirus isolation & purification, Betacoronavirus physiology, COVID-19, Correlation of Data, Gene Expression Regulation, Germ-Free Life, RNA, Messenger analysis, Rats, SARS-CoV-2, Colitis metabolism, Colitis virology, Colon metabolism, Colon pathology, Colon virology, Coronavirus Infections metabolism, Coronavirus Infections pathology, Coronavirus Infections physiopathology, Gastrointestinal Microbiome physiology, Pandemics, Peptidyl-Dipeptidase A metabolism, Pneumonia, Viral metabolism, Pneumonia, Viral pathology, Pneumonia, Viral physiopathology
- Published
- 2020
- Full Text
- View/download PDF
35. Acquisition of Cisplatin Resistance Shifts Head and Neck Squamous Cell Carcinoma Metabolism toward Neutralization of Oxidative Stress.
- Author
-
Yu W, Chen Y, Putluri N, Coarfa C, Robertson MJ, Putluri V, Stossi F, Dubrulle J, Mancini MA, Pang JC, Nguyen T, Baluya D, Myers JN, Lai SY, and Sandulache VC
- Abstract
Background : Cisplatin (CDDP) is commonly utilized in the treatment of advanced solid tumors including head and neck squamous cell carcinoma (HNSCC). Cisplatin response remains highly variable among individual tumors and development of cisplatin resistance is common. We hypothesized that development of cisplatin resistance is partially driven by metabolic reprogramming. Methods : Using a pre-clinical HNSCC model and an integrated approach to steady state metabolomics, metabolic flux and gene expression data we characterized the interaction between cisplatin resistance and metabolic reprogramming. Results : Cisplatin toxicity in HNSCC was driven by generation of intra-cellular oxidative stress. This was validated by demonstrating that acquisition of cisplatin resistance generates cross-resistance to ferroptosis agonists despite the fact that cisplatin itself does not trigger ferroptosis. Acquisition of cisplatin resistance dysregulated the expression of genes involved in amino acid, fatty acid metabolism and central carbon catabolic pathways, enhanced glucose catabolism and serine synthesis. Acute cisplatin exposure increased intra-tumoral levels of S-methyl-5-thiadenosine (MTA) precursors and metabotoxins indicative of generalized oxidative stress. Conclusions: Acquisition of cisplatin resistance is linked to metabolic recovery from oxidative stress. Although this portends poor effectiveness for directed metabolic targeting, it supports the potential for biomarker development of cisplatin effectiveness using an integrated approach.
- Published
- 2020
- Full Text
- View/download PDF
36. Epigenome environment interactions accelerate epigenomic aging and unlock metabolically restricted epigenetic reprogramming in adulthood.
- Author
-
Treviño LS, Dong J, Kaushal A, Katz TA, Jangid RK, Robertson MJ, Grimm SL, Ambati CSR, Putluri V, Cox AR, Kim KH, May TD, Gallo MR, Moore DD, Hartig SM, Foulds CE, Putluri N, Coarfa C, and Walker CL
- Subjects
- Animals, DNA Methylation drug effects, DNA Methylation genetics, Early Growth Response Protein 1 genetics, Endocrine Disruptors toxicity, Epigenesis, Genetic drug effects, Epigenesis, Genetic genetics, Epigenomics methods, Female, Gene-Environment Interaction, Genome-Wide Association Study, Male, Rats, Epigenome genetics
- Abstract
Our early-life environment has a profound influence on developing organs that impacts metabolic function and determines disease susceptibility across the life-course. Using a rat model for exposure to an endocrine disrupting chemical (EDC), we show that early-life chemical exposure causes metabolic dysfunction in adulthood and reprograms histone marks in the developing liver to accelerate acquisition of an adult epigenomic signature. This epigenomic reprogramming persists long after the initial exposure, but many reprogrammed genes remain transcriptionally silent with their impact on metabolism not revealed until a later life exposure to a Western-style diet. Diet-dependent metabolic disruption was largely driven by reprogramming of the Early Growth Response 1 (EGR1) transcriptome and production of metabolites in pathways linked to cholesterol, lipid and one-carbon metabolism. These findings demonstrate the importance of epigenome:environment interactions, which early in life accelerate epigenomic aging, and later in adulthood unlock metabolically restricted epigenetic reprogramming to drive metabolic dysfunction.
- Published
- 2020
- Full Text
- View/download PDF
37. Correction: Expression of ganglioside GD2, reprogram the lipid metabolism and EMT phenotype in bladder cancer.
- Author
-
Vantaku V, Donepudi SR, Ambati CR, Jin F, Putluri V, Nguyen K, Rajapakshe K, Coarfa C, Battula VL, Lotan Y, and Putluri N
- Abstract
[This corrects the article DOI: 10.18632/oncotarget.21038.]., (Copyright: © 2019 Vantaku et al.)
- Published
- 2019
- Full Text
- View/download PDF
38. DNA methylation patterns in bladder tumors of African American patients point to distinct alterations in xenobiotic metabolism.
- Author
-
Vantaku V, Amara CS, Piyarathna DWB, Donepudi SR, Ambati CR, Putluri V, Tang W, Rajapakshe K, Estecio MR, Terris MK, Castro PD, Ittmann MM, Williams SB, Lerner SP, Sreekumar A, Bollag R, Coarfa C, Kornberg MD, Lotan Y, Ambs S, and Putluri N
- Subjects
- Black or African American genetics, Chromatography, Liquid, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Glucuronic Acid analysis, Glucuronic Acid metabolism, Humans, Metabolomics, Promoter Regions, Genetic, S-Adenosylhomocysteine analysis, S-Adenosylhomocysteine metabolism, S-Adenosylmethionine analysis, S-Adenosylmethionine metabolism, Tandem Mass Spectrometry, Urinary Bladder Neoplasms metabolism, White People genetics, CpG Islands, DNA Methylation, Inactivation, Metabolic genetics, Urinary Bladder Neoplasms genetics
- Abstract
Racial/ethnic disparities have a significant impact on bladder cancer outcomes with African American patients demonstrating inferior survival over European-American patients. We hypothesized that epigenetic difference in methylation of tumor DNA is an underlying cause of this survival health disparity. We analyzed bladder tumors from African American and European-American patients using reduced representation bisulfite sequencing (RRBS) to annotate differentially methylated DNA regions. Liquid chromatography-mass spectrometry (LC-MS/MS) based metabolomics and flux studies were performed to examine metabolic pathways that showed significant association to the discovered DNA methylation patterns. RRBS analysis showed frequent hypermethylated CpG islands in African American patients. Further analysis showed that these hypermethylated CpG islands in patients are commonly located in the promoter regions of xenobiotic enzymes that are involved in bladder cancer progression. On follow-up, LC-MS/MS revealed accumulation of glucuronic acid, S-adenosylhomocysteine, and a decrease in S-adenosylmethionine, corroborating findings from the RRBS and mRNA expression analysis indicating increased glucuronidation and methylation capacities in African American patients. Flux analysis experiments with 13C-labeled glucose in cultured African American bladder cancer cells confirmed these findings. Collectively, our studies revealed robust differences in methylation-related metabolism and expression of enzymes regulating xenobiotic metabolism in African American patients indicate that race/ethnic differences in tumor biology may exist in bladder cancer., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
39. Liver- and Microbiome-derived Bile Acids Accumulate in Human Breast Tumors and Inhibit Growth and Improve Patient Survival.
- Author
-
Tang W, Putluri V, Ambati CR, Dorsey TH, Putluri N, and Ambs S
- Subjects
- Breast Neoplasms mortality, Breast Neoplasms pathology, Cell Proliferation physiology, Female, Humans, Metabolome, Microbiota, Prognosis, Survival Rate, Tumor Cells, Cultured, Bile Acids and Salts metabolism, Breast Neoplasms metabolism, Liver metabolism
- Abstract
Purpose: Metabolomics is a discovery tool for novel associations of metabolites with disease. Here, we interrogated the metabolome of human breast tumors to describe metabolites whose accumulation affects tumor biology., Experimental Design: We applied large-scale metabolomics followed by absolute quantification and machine learning-based feature selection using LASSO to identify metabolites that show a robust association with tumor biology and disease outcome. Key observations were validated with the analysis of an independent dataset and cell culture experiments., Results: LASSO-based feature selection revealed an association of tumor glycochenodeoxycholate levels with improved breast cancer survival, which was confirmed using a Cox proportional hazards model. Absolute quantification of four bile acids, including glycochenodeoxycholate and microbiome-derived deoxycholate, corroborated the accumulation of bile acids in breast tumors. Levels of glycochenodeoxycholate and other bile acids showed an inverse association with the proliferation score in tumors and the expression of cell-cycle and G
2 -M checkpoint genes, which was corroborated with cell culture experiments. Moreover, tumor levels of these bile acids markedly correlated with metabolites in the steroid metabolism pathway and increased expression of key genes in this pathway, suggesting that bile acids may interfere with hormonal pathways in the breast. Finally, a proteome analysis identified the complement and coagulation cascade as being upregulated in glycochenodeoxycholate-high tumors., Conclusions: We describe the unexpected accumulation of liver- and microbiome-derived bile acids in breast tumors. Tumors with increased bile acids show decreased proliferation, thus fall into a good prognosis category, and exhibit significant changes in steroid metabolism., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
40. Erratum to "Distinct Lipidomic Landscapes Associated with Clinical Stages of Urothelial Cancer of the Bladder" [Eur Urol Focus 2018;4:907-915].
- Author
-
Piyarathna DWB, Rajendiran TM, Putluri V, Vantaku V, Soni T, von Rundstedt FC, Donepudi SR, Jin F, Maity S, Ambati CR, Dong J, Gödde D, Roth S, Störkel S, Degener S, Michailidis G, Lerner SP, Pennathur S, Lotan Y, Coarfa C, Sreekumar A, and Putluri N
- Published
- 2019
- Full Text
- View/download PDF
41. Association between elevated placental polycyclic aromatic hydrocarbons (PAHs) and PAH-DNA adducts from Superfund sites in Harris County, and increased risk of preterm birth (PTB).
- Author
-
Suter MA, Aagaard KM, Coarfa C, Robertson M, Zhou G, Jackson BP, Thompson D, Putluri V, Putluri N, Hagan J, Wang L, Jiang W, Lingappan K, and Moorthy B
- Subjects
- Female, Gene Expression Regulation, Humans, Infant, Newborn, Metabolome, Pregnancy, Risk Factors, Texas, DNA Adducts analysis, Environmental Pollution, Placenta metabolism, Polycyclic Aromatic Hydrocarbons analysis, Premature Birth chemically induced
- Abstract
The preterm birth (PTB) rate in Harris County, Texas, exceeds the U.S. rate (11.4% vs.9.6%), and there are 15 active Superfund sites in Harris County. Polycyclic aromatic hydrocarbons (PAHs) are contaminants of concern (COC) at Superfund sites across the nation. In this investigation, we tested the hypothesis that higher levels of exposure to PAHs and PAH-DNA adducts in placenta of women living near Superfund sites contribute to the increased rate of PTBs. Levels of benzo[a]pyene (BP), benzo[b]fluorene (BbF) and dibenz[a,h]anthracene (DBA), were higher in placentae from preterm deliveries compared with term deliveries in women living near Superfund sites, whereas this was not the case for women living in non-Superfund site areas. Among the PAHs, DBA levels were significantly higher than BP or BbF, and DBA levels were inversely correlated with gestational age at delivery and birth weight. Bulky PAH-DNA adducts are more prevalent in placental tissue from individuals residing near Superfund sites. Expression of Ah receptor (AHR) and NF-E2-related factor 2 (NRF2) was decreased in preterm deliveries in subjects residing near Superfund sites. Unbiased metabolomics revealed alterations in pathways involved in pentose phosphate, inositol phosphate and starch and sucrose metabolism in preterm subjects in Superfund site areas. In summary, this is the first report showing an association between PAH levels, DNA adducts, and modulation of endogenous metabolic pathways with PTBs in subjects residing near Superfund sites, and further studies could lead to novel strategies in the understanding of the mechanisms by which PAHs contribute to PTBs in women., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
42. Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression.
- Author
-
Hu Q, Ye Y, Chan LC, Li Y, Liang K, Lin A, Egranov SD, Zhang Y, Xia W, Gong J, Pan Y, Chatterjee SS, Yao J, Evans KW, Nguyen TK, Park PK, Liu J, Coarfa C, Donepudi SR, Putluri V, Putluri N, Sreekumar A, Ambati CR, Hawke DH, Marks JR, Gunaratne PH, Caudle AS, Sahin AA, Hortobagyi GN, Meric-Bernstam F, Chen L, Yu D, Hung MC, Curran MA, Han L, Lin C, and Yang L
- Subjects
- Adenoma genetics, Adenoma metabolism, Animals, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Disease Models, Animal, Disease Progression, Humans, Mice, Neoplasms metabolism, Neoplasms pathology, Phosphorylation, Receptors, G-Protein-Coupled antagonists & inhibitors, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Tumor Suppressor Protein p53 metabolism, Ubiquitination, Xenograft Model Antitumor Assays, Antigen Presentation immunology, Gene Expression Regulation, Neoplastic, Neoplasms genetics, Neoplasms immunology, Oncogenes, RNA, Long Noncoding genetics, Tumor Escape genetics, Tumor Escape immunology
- Abstract
How tumor cells genetically lose antigenicity and evade immune checkpoints remains largely elusive. We report that tissue-specific expression of the human long noncoding RNA LINK-A in mouse mammary glands initiates metastatic mammary gland tumors, which phenotypically resemble human triple-negative breast cancer (TNBC). LINK-A expression facilitated crosstalk between phosphatidylinositol-(3,4,5)-trisphosphate and inhibitory G-protein-coupled receptor (GPCR) pathways, attenuating protein kinase A-mediated phosphorylation of the E3 ubiquitin ligase TRIM71. Consequently, LINK-A expression enhanced K48-polyubiquitination-mediated degradation of the antigen peptide-loading complex (PLC) and intrinsic tumor suppressors Rb and p53. Treatment with LINK-A locked nucleic acids or GPCR antagonists stabilized the PLC components, Rb and p53, and sensitized mammary gland tumors to immune checkpoint blockers. Patients with programmed ccll death protein-1(PD-1) blockade-resistant TNBC exhibited elevated LINK-A levels and downregulated PLC components. Hence we demonstrate lncRNA-dependent downregulation of antigenicity and intrinsic tumor suppression, which provides the basis for developing combinational immunotherapy treatment regimens and early TNBC prevention.
- Published
- 2019
- Full Text
- View/download PDF
43. Multi-omics Integration Analysis Robustly Predicts High-Grade Patient Survival and Identifies CPT1B Effect on Fatty Acid Metabolism in Bladder Cancer.
- Author
-
Vantaku V, Dong J, Ambati CR, Perera D, Donepudi SR, Amara CS, Putluri V, Ravi SS, Robertson MJ, Piyarathna DWB, Villanueva M, von Rundstedt FC, Karanam B, Ballester LY, Terris MK, Bollag RJ, Lerner SP, Apolo AB, Villanueva H, Lee M, Sikora AG, Lotan Y, Sreekumar A, Coarfa C, and Putluri N
- Subjects
- Biomarkers, Tumor metabolism, Humans, Lipidomics methods, Liver Neoplasms genetics, Liver Neoplasms metabolism, Liver Neoplasms mortality, Liver Neoplasms secondary, Metabolome, Neoplasm Grading, Predictive Value of Tests, Survival Rate, Transcriptome, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms mortality, Urinary Bladder Neoplasms pathology, Carnitine O-Palmitoyltransferase metabolism, Fatty Acids metabolism, Urinary Bladder Neoplasms metabolism
- Abstract
Purpose: The perturbation of metabolic pathways in high-grade bladder cancer has not been investigated. We aimed to identify a metabolic signature in high-grade bladder cancer by integrating unbiased metabolomics, lipidomics, and transcriptomics to predict patient survival and to discover novel therapeutic targets., Experimental Design: We performed high-resolution liquid chromatography mass spectrometry (LC-MS) and bioinformatic analysis to determine the global metabolome and lipidome in high-grade bladder cancer. We further investigated the effects of impaired metabolic pathways using in vitro and in vivo models., Results: We identified 519 differential metabolites and 19 lipids that were differentially expressed between low-grade and high-grade bladder cancer using the NIST MS metabolomics compendium and lipidblast MS/MS libraries, respectively. Pathway analysis revealed a unique set of biochemical pathways that are highly deregulated in high-grade bladder cancer. Integromics analysis identified a molecular gene signature associated with poor patient survival in bladder cancer. Low expression of CPT1B in high-grade tumors was associated with low FAO and low acyl carnitine levels in high-grade bladder cancer, which were confirmed using tissue microarrays. Ectopic expression of the CPT1B in high-grade bladder cancer cells led to reduced EMT in in vitro , and reduced cell proliferation, EMT, and metastasis in vivo ., Conclusions: Our study demonstrates a novel approach for the integration of metabolomics, lipidomics, and transcriptomics data, and identifies a common gene signature associated with poor survival in patients with bladder cancer. Our data also suggest that impairment of FAO due to downregulation of CPT1B plays an important role in the progression toward high-grade bladder cancer and provide potential targets for therapeutic intervention., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
44. Methionine-Homocysteine Pathway in African-American Prostate Cancer.
- Author
-
Gohlke JH, Lloyd SM, Basu S, Putluri V, Vareed SK, Rasaily U, Piyarathna DWB, Fuentes H, Rajendiran TM, Dorsey TH, Ambati CR, Sonavane R, Karanam B, Bhowmik SK, Kittles R, Ambs S, Mims MP, Ittmann M, Jones JA, Palapattu G, Putluri N, Michailidis G, and Sreekumar A
- Abstract
African American (AA) men have a 60% higher incidence and two times greater risk of dying of prostate cancer (PCa) than European American men, yet there is limited insight into the molecular mechanisms driving this difference. To our knowledge, metabolic alterations, a cancer-associated hallmark, have not been reported in AA PCa, despite their importance in tumor biology. Therefore, we measured 190 metabolites across ancestry-verified AA PCa/benign adjacent tissue pairs (n = 33 each) and identified alterations in the methionine-homocysteine pathway utilizing two-sided statistical tests for all comparisons. Consistent with this finding, methionine and homocysteine were elevated in plasma from AA PCa patients using case-control (AA PCa vs AA control, methionine: P = .0007 and homocysteine: P < .0001), biopsy cohorts (AA biopsy positive vs AA biopsy negative, methionine: P = .0002 and homocysteine: P < .0001), and race assignments based on either self-report (AA PCa vs European American PCa, methionine: P = .001, homocysteine: P < .0001) or West African ancestry (upper tertile vs middle tertile, homocysteine: P < .0001; upper tertile vs low tertile, homocysteine: P = .002). These findings demonstrate reprogrammed metabolism in AA PCa patients and provide a potential biological basis for PCa disparities.
- Published
- 2019
- Full Text
- View/download PDF
45. Serum Metabolic Profiling Identified a Distinct Metabolic Signature in Bladder Cancer Smokers: A Key Metabolic Enzyme Associated with Patient Survival.
- Author
-
Amara CS, Ambati CR, Vantaku V, Badrajee Piyarathna DW, Donepudi SR, Ravi SS, Arnold JM, Putluri V, Chatta G, Guru KA, Badr H, Terris MK, Bollag RJ, Sreekumar A, Apolo AB, and Putluri N
- Subjects
- Female, Humans, Male, Metabolomics, Middle Aged, Survival Analysis, Urinary Bladder Neoplasms mortality, Cigarette Smoking adverse effects, Urinary Bladder Neoplasms etiology
- Abstract
Background: The current system to predict the outcome of smokers with bladder cancer is insufficient due to complex genomic and transcriptomic heterogeneities. This study aims to identify serum metabolite-associated genes related to survival in this population., Methods: We performed LC/MS-based targeted metabolomic analysis for >300 metabolites in serum obtained from two independent cohorts of bladder cancer never smokers, smokers, healthy smokers, and healthy never smokers. A subset of differential metabolites was validated using Biocrates absoluteIDQ p180 Kit. Genes associated with differential metabolites were integrated with a publicly available cohort of The Cancer Genome Atlas (TCGA) to obtain an intersecting signature specific for bladder cancer smokers., Results: Forty metabolites (FDR < 0.25) were identified to be differential between bladder cancer never smokers and smokers. Increased abundance of amino acids (tyrosine, phenylalanine, proline, serine, valine, isoleucine, glycine, and asparagine) and taurine were observed in bladder cancer smokers. Integration of differential metabolomic gene signature and transcriptomics data from TCGA cohort revealed an intersection of 17 genes that showed significant correlation with patient survival in bladder cancer smokers. Importantly, catechol-O-methyltransferase, iodotyrosine deiodinase, and tubulin tyrosine ligase showed a significant association with patient survival in publicly available bladder cancer smoker datasets and did not have any clinical association in never smokers., Conclusions: Serum metabolic profiling of bladder cancer smokers revealed dysregulated amino acid metabolism. It provides a distinct gene signature that shows a prognostic value in predicting bladder cancer smoker survival., Impact: Serum metabolic signature-derived genes act as a predictive tool for studying the bladder cancer progression in smokers., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
46. A Prospective Targeted Serum Metabolomics Study of Pancreatic Cancer in Postmenopausal Women.
- Author
-
Jiao L, Maity S, Coarfa C, Rajapakshe K, Chen L, Jin F, Putluri V, Tinker LF, Mo Q, Chen F, Sen S, Sangi-Hyghpeykar H, El-Serag HB, and Putluri N
- Subjects
- Aged, Case-Control Studies, Female, Humans, Middle Aged, Postmenopause, Prospective Studies, Biomarkers, Tumor blood, Metabolome, Metabolomics methods, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology
- Abstract
To examine the association between metabolic deregulation and pancreatic cancer, we conducted a two-stage case-control targeted metabolomics study using prediagnostic sera collected one year before diagnosis in the Women's Health Initiative study. We used the LC/MS to quantitate 470 metabolites in 30 matched case/control pairs. From 180 detectable metabolites, we selected 14 metabolites to be validated in additional 18 matched case/control pairs. We used the paired t test to compare the concentrations of each metabolite between cases and controls and used the log fold change (FC) to indicate the magnitude of difference. FDR adjusted q-value < 0.25 was indicated statistically significant. Logistic regression model and ROC curve analysis were used to evaluate the clinical utility of the metabolites. Among 30 case/control pairs, 1-methyl-l-tryptophan (L-1MT) was significantly lower in the cases than in the controls (log
2 FC = -0.35; q-value = 0.03). The area under the ROC curve was 0.83 in the discrimination analysis based on the levels of L-1MT, acadesine, and aspartic acid. None of the metabolites was validated in additional independent 18 case/control pairs. No significant association was found between the examined metabolites and undiagnosed pancreatic cancer., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
47. Large-scale profiling of serum metabolites in African American and European American patients with bladder cancer reveals metabolic pathways associated with patient survival.
- Author
-
Vantaku V, Donepudi SR, Piyarathna DWB, Amara CS, Ambati CR, Tang W, Putluri V, Chandrashekar DS, Varambally S, Terris MK, Davies K, Ambs S, Bollag R, Apolo AB, Sreekumar A, and Putluri N
- Subjects
- Amino Acids blood, Case-Control Studies, Chromatography, Liquid, Female, Humans, Lipids blood, Male, Mass Spectrometry, Metabolic Networks and Pathways, Survival Analysis, Urinary Bladder Neoplasms ethnology, Urinary Bladder Neoplasms mortality, Black or African American statistics & numerical data, Metabolomics methods, Urinary Bladder Neoplasms blood, White People statistics & numerical data
- Abstract
Background: African Americans (AAs) experience a disproportionally high rate of bladder cancer (BLCA) deaths even though their incidence rates are lower than those of other patient groups. Using a metabolomics approach, this study investigated how AA BLCA may differ molecularly from European Americans (EAs) BLCA, and it examined serum samples from patients with BLCA with the aim of identifying druggable metabolic pathways in AA patients., Methods: Targeted metabolomics was applied to measure more than 300 metabolites in serum samples from 2 independent cohorts of EA and AA patients with BLCA and healthy EA and AA controls via liquid chromatography-mass spectrometry, and this was followed by the identification of altered metabolic pathways with a focus on AA BLCA. A subset of the differential metabolites was validated via absolute quantification with the Biocrates AbsoluteIDQ p180 kit. The clinical significance of the findings was further examined in The Cancer Genomic Atlas BLCA data set., Results: Fifty-three metabolites, mainly related to amino acid, lipid, and nucleotide metabolism, were identified that showed significant differences in abundance between AA and EA BLCA. For example, the levels of taurine, glutamine, glutamate, aspartate, and serine were elevated in serum samples from AA patients versus EA patients. By mapping these metabolites to genes, this study identified significant relations with regulators of metabolism such as malic enzyme 3, prolyl 3-hydroxylase 2, and lysine demethylase 2A that predicted patient survival exclusively in AA patients with BLCA., Conclusions: This metabolic profile of serum samples might be used to assess risk progression in AA BLCA. These first-in-field findings describe metabolic alterations in AA BLCA and emphasize a potential biological basis for BLCA health disparities., (© 2019 American Cancer Society.)
- Published
- 2019
- Full Text
- View/download PDF
48. Measurement of methylated metabolites using Liquid Chromatography-Mass Spectrometry and its biological application.
- Author
-
Ambati CR, Vantaku V, Donepudi SR, Amara CS, Ravi SS, Mandalapu A, Perla M, Putluri V, Sreekumar A, and Putluri N
- Abstract
Methylation aberrations play an important role in many metabolic disorders including cancer. Methylated metabolites are direct indicators of metabolic aberrations, and currently, there is no Liquid chromatography - Mass spectrometry (LC-MS) based method available to cover all classes of methylated metabolites at low detection limits. In this study, we have developed a method for the detection of methylated metabolites, and it's biological application. In this approach, we used a HILIC based HPLC with MS to measure methylated organic acids, amino acids, and nucleotides. These metabolites were separated from each other by their hydrophobic interactions and analyzed by targeted metabolomics of single reaction monitoring by positive and negative mode of electrospray ionization. These metabolites were quantified, and the interday reproducibility was <10% relative standard deviation. Furthermore, by applying this method, we identified high levels of methylated metabolites in bladder cancer cell lines compared to benign cells. In vitro treatment of cancer cells with methylation inhibitor, 5- aza-2'-deoxycytidine showed a decrease in these methylated metabolites. This data indicates that HPLC analysis using this HILIC based method could be a powerful tool for measuring methylated metabolites in biological specimens. This method is rapid, sensitive, selective, and precise to measure methylated metabolites., Competing Interests: Conflict of interest statement: Authors do not have any conflict of interest
- Published
- 2019
- Full Text
- View/download PDF
49. Mitochondrial pyruvate import is a metabolic vulnerability in androgen receptor-driven prostate cancer.
- Author
-
Bader DA, Hartig SM, Putluri V, Foley C, Hamilton MP, Smith EA, Saha PK, Panigrahi A, Walker C, Zong L, Martini-Stoica H, Chen R, Rajapakshe K, Coarfa C, Sreekumar A, Mitsiades N, Bankson JA, Ittmann MM, O'Malley BW, Putluri N, and McGuire SE
- Subjects
- Animals, Biological Transport, Cell Line, Tumor, Disease Models, Animal, Gene Expression Regulation, Neoplastic, Glutamine metabolism, Humans, Male, Metabolic Networks and Pathways, Mice, Mice, Transgenic, Mitochondrial Membrane Transport Proteins genetics, Mitochondrial Membrane Transport Proteins metabolism, Monocarboxylic Acid Transporters metabolism, Prostatic Neoplasms mortality, Prostatic Neoplasms pathology, Prostatic Neoplasms, Castration-Resistant etiology, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Protein Binding, Signal Transduction, Mitochondria metabolism, Prostatic Neoplasms etiology, Prostatic Neoplasms metabolism, Pyruvic Acid metabolism, Receptors, Androgen metabolism
- Abstract
Specific metabolic underpinnings of androgen receptor (AR)-driven growth in prostate adenocarcinoma (PCa) are largely undefined, hindering the development of strategies to leverage the metabolic dependencies of this disease when hormonal manipulations fail. Here we show that the mitochondrial pyruvate carrier (MPC), a critical metabolic conduit linking cytosolic and mitochondrial metabolism, is transcriptionally regulated by AR. Experimental MPC inhibition restricts proliferation and metabolic outputs of the citric acid cycle (TCA) including lipogenesis and oxidative phosphorylation in AR-driven PCa models. Mechanistically, metabolic disruption resulting from MPC inhibition activates the eIF2α/ATF4 integrated stress response (ISR). ISR signaling prevents cell cycle progression while coordinating salvage efforts, chiefly enhanced glutamine assimilation into the TCA, to regain metabolic homeostasis. We confirm that MPC function is operant in PCa tumors in-vivo using isotopomeric metabolic flux analysis. In turn, we apply a clinically viable small molecule targeting the MPC, MSDC0160, to pre-clinical PCa models and find that MPC inhibition suppresses tumor growth in hormone-responsive and castrate-resistant conditions. Collectively, our findings characterize the MPC as a tractable therapeutic target in AR-driven prostate tumors., Competing Interests: Competing Interests The authors declare no competing interests.
- Published
- 2019
- Full Text
- View/download PDF
50. Distinct Lipidomic Landscapes Associated with Clinical Stages of Urothelial Cancer of the Bladder.
- Author
-
Piyarathna DWB, Rajendiran TM, Putluri V, Vantaku V, Soni T, von Rundstedt FC, Donepudi SR, Jin F, Maity S, Ambati CR, Dong J, Gödde D, Roth S, Störkel S, Degener S, Michailidis G, Lerner SP, Pennathur S, Lotan Y, Coarfa C, Sreekumar A, and Putluri N
- Subjects
- Carcinoma, Transitional Cell genetics, Carcinoma, Transitional Cell pathology, Case-Control Studies, Chromatography, Liquid, Computational Biology, Diglycerides metabolism, Female, Humans, Lipid Metabolism genetics, Lymph Nodes pathology, Lysophospholipids metabolism, Male, Mass Spectrometry, Neoplasm Invasiveness, Neoplasm Staging, Nitrogenous Group Transferases genetics, Nitrogenous Group Transferases metabolism, Phosphatidylethanolamines metabolism, Phosphatidylserines metabolism, Phosphorylcholine metabolism, Principal Component Analysis, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms pathology, Carcinoma, Transitional Cell metabolism, Fatty Acids metabolism, Phospholipids metabolism, Triglycerides metabolism, Urinary Bladder Neoplasms metabolism
- Abstract
Background: The first global lipidomic profiles associated with urothelial cancer of the bladder (UCB) and its clinical stages associated with progression were identified., Objective: To identify lipidomic signatures associated with survival and different clinical stages of UCB., Design, Setting, and Participants: Pathologically confirmed 165 bladder-derived tissues (126 UCB, 39 benign adjacent or normal bladder tissues). UCB tissues included Ta (n=16), T1 (n=30), T2 (n=43), T3 (n=27), and T4 (n=9); lymphovascular invasion (LVI) positive (n=52) and negative (n=69); and lymph node status N0 (n=28), N1 (n=11), N2 (n=9), N3 (n=3), and Nx (n=75)., Results and Limitations: UCB tissues have higher levels of phospholipids and fatty acids, and reduced levels of triglycerides compared with benign tissues. A total of 59 genes associated with altered lipids in UCB strongly correlate with patient survival in an UCB public dataset. Within UCB, there was a progressive decrease in the levels of phosphatidylserine (PS), phosphatidylethanolamines (PEs), and phosphocholines, whereas an increase in the levels of diacylglycerols (DGs) with tumor stage. Transcript and protein expression of phosphatidylserine synthase 1, which converts DGs to PSs, decreased progressively with tumor stage. Levels of DGs and lyso-PEs were significantly elevated in tumors with LVI and lymph node involvement, respectively. Lack of carcinoma in situ and treatment information is the limitation of our study., Conclusions: To date, this is the first study describing the global lipidomic profiles associated with UCB and identifies lipids associated with tumor stages, LVI, and lymph node status. Our data suggest that triglycerides serve as the primary energy source in UCB, while phospholipid alterations could affect membrane structure and/or signaling associated with tumor progression., Patient Summary: Lipidomic alterations identified in this study set the stage for characterization of pathways associated with these altered lipids that, in turn, could inform the development of first-of-its-kind lipid-based noninvasive biomarkers and novel therapeutic targets for aggressive urothelial cancer of the bladder., (Copyright © 2017 European Association of Urology. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.