1
|
Pujana-Vaquerizo M, Bozal-Basterra L, Carracedo A. Metabolic adaptations in prostate cancer. Br J Cancer 2024; 131:1250-1262. [PMID: 38969865 PMCID: PMC11473656 DOI: 10.1038/s41416-024-02762-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024] Open
Abstract
Prostate cancer is one of the most commonly diagnosed cancers in men and is a major cause of cancer-related deaths worldwide. Among the molecular processes that contribute to this disease, the weight of metabolism has been placed under the limelight in recent years. Tumours exhibit metabolic adaptations to comply with their biosynthetic needs. However, metabolites also play an important role in supporting cell survival in challenging environments or remodelling the tumour microenvironment, thus being recognized as a hallmark in cancer. Prostate cancer is uniquely driven by androgen receptor signalling, and this knowledge has also influenced the paths of cancer metabolism research. This review provides a comprehensive perspective on the metabolic adaptations that support prostate cancer progression beyond androgen signalling, with a particular focus on tumour cell intrinsic and extrinsic pathways.
Collapse
Affiliation(s)
- Mikel Pujana-Vaquerizo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Laura Bozal-Basterra
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029, Madrid, Spain.
- Traslational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, Baracaldo, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
2
|
Galey L, Olanrewaju A, Nabi H, Paquette JS, Pouliot F, Audet-Walsh É. PSA, an outdated biomarker for prostate cancer: In search of a more specific biomarker, citrate takes the spotlight. J Steroid Biochem Mol Biol 2024; 243:106588. [PMID: 39025336 DOI: 10.1016/j.jsbmb.2024.106588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
The prevailing biomarker employed for prostate cancer (PCa) screening and diagnosis is the prostate-specific antigen (PSA). Despite excellent sensitivity, PSA lacks specificity, leading to false positives, unnecessary biopsies and overdiagnosis. Consequently, PSA is increasingly less used by clinicians, thus underscoring the imperative for the identification of new biomarkers. An emerging biomarker in this context is citrate, a molecule secreted by the normal prostate, which has been shown to be inversely correlated with PCa. Here, we discuss about PSA and its usage for PCa diagnosis, its lack of specificity, and the various conditions that can affect its levels. We then provide our vision about what we think would be a valuable addition to our PCa diagnosis toolkit, citrate. We describe the unique citrate metabolic program in the prostate and how this profile is reprogrammed during carcinogenesis. Finally, we summarize the evidence that supports the usage of citrate as a biomarker for PCa diagnosis, as it can be measured in various patient samples and be analyzed by several methods. The unique relationship between citrate and PCa, combined with the stability of citrate levels in other prostate-related conditions and the simplicity of its detection, further accentuates its potential as a biomarker.
Collapse
Affiliation(s)
- Lucas Galey
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada
| | - Ayokunle Olanrewaju
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Hermann Nabi
- Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada
| | - Jean-Sébastien Paquette
- Laboratoire de recherche et d'innovation en médecine de première ligne (ARIMED), Groupe de médecine de famille universitaire de Saint-Charles-Borromée, CISSS Lanaudière, Saint-Charles-Borromée, QC, Canada; VITAM Research Centre for Sustainable Health, Québec, QC, Canada; Department of Family Medicine and Emergency Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Frédéric Pouliot
- Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada; Department of Family Medicine and Emergency Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Department of surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada.
| |
Collapse
|
3
|
Evin D, Evinová A, Baranovičová E, Šarlinová M, Jurečeková J, Kaplán P, Poláček H, Halašová E, Dušenka R, Briš L, Brožová MK, Sivoňová MK. Integrative Metabolomic Analysis of Serum and Selected Serum Exosomal microRNA in Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2024; 25:2630. [PMID: 38473877 DOI: 10.3390/ijms25052630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains a lethal disease due to the absence of effective therapies. A more comprehensive understanding of molecular events, encompassing the dysregulation of microRNAs (miRs) and metabolic reprogramming, holds the potential to unveil precise mechanisms underlying mCRPC. This study aims to assess the expression of selected serum exosomal miRs (miR-15a, miR-16, miR-19a-3p, miR-21, and miR-141a-3p) alongside serum metabolomic profiling and their correlation in patients with mCRPC and benign prostate hyperplasia (BPH). Blood serum samples from mCRPC patients (n = 51) and BPH patients (n = 48) underwent metabolome analysis through 1H-NMR spectroscopy. The expression levels of serum exosomal miRs in mCRPC and BPH patients were evaluated using a quantitative real-time polymerase chain reaction (qRT-PCR). The 1H-NMR metabolomics analysis revealed significant alterations in lactate, acetate, citrate, 3-hydroxybutyrate, and branched-chain amino acids (BCAAs, including valine, leucine, and isoleucine) in mCRPC patients compared to BPH patients. MiR-15a, miR-16, miR-19a-3p, and miR-21 exhibited a downregulation of more than twofold in the mCRPC group. Significant correlations were predominantly observed between lactate, citrate, acetate, and miR-15a, miR-16, miR-19a-3p, and miR-21. The importance of integrating metabolome analysis of serum with selected serum exosomal miRs in mCRPC patients has been confirmed, suggesting their potential utility for distinguishing of mCRPC from BPH.
Collapse
Affiliation(s)
- Daniel Evin
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
- Clinic of Nuclear Medicine, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Andrea Evinová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Eva Baranovičová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Miroslava Šarlinová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Jana Jurečeková
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Peter Kaplán
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Hubert Poláček
- Clinic of Nuclear Medicine, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Erika Halašová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Róbert Dušenka
- Clinic of Urology, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lukáš Briš
- Clinic of Urology, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Martina Knoško Brožová
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Monika Kmeťová Sivoňová
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
4
|
Gonthier K, Weidmann C, Berthiaume L, Jobin C, Lacouture A, Lafront C, Harvey M, Neveu B, Loehr J, Bergeron A, Fradet Y, Lacombe L, Riopel J, Latulippe É, Atallah C, Shum M, Lambert J, Pouliot F, Pelletier M, Audet‐Walsh É. Isocitrate dehydrogenase 1 sustains a hybrid cytoplasmic-mitochondrial tricarboxylic acid cycle that can be targeted for therapeutic purposes in prostate cancer. Mol Oncol 2023; 17:2109-2125. [PMID: 37086156 PMCID: PMC10552900 DOI: 10.1002/1878-0261.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/07/2023] [Accepted: 04/21/2023] [Indexed: 04/23/2023] Open
Abstract
The androgen receptor (AR) is an established orchestrator of cell metabolism in prostate cancer (PCa), notably by inducing an oxidative mitochondrial program. Intriguingly, AR regulates cytoplasmic isocitrate dehydrogenase 1 (IDH1), but not its mitochondrial counterparts IDH2 and IDH3. Here, we aimed to understand the functional role of IDH1 in PCa. Mouse models, in vitro human PCa cell lines, and human patient-derived organoids (PDOs) were used to study the expression and activity of IDH enzymes in the normal prostate and PCa. Genetic and pharmacological inhibition of IDH1 was then combined with extracellular flux analyses and gas chromatography-mass spectrometry for metabolomic analyses and cancer cell proliferation in vitro and in vivo. In PCa cells, more than 90% of the total IDH activity is mediated through IDH1 rather than its mitochondrial counterparts. This profile seems to originate from the specialized prostate metabolic program, as observed using mouse prostate and PDOs. Pharmacological and genetic inhibition of IDH1 impaired mitochondrial respiration, suggesting that this cytoplasmic enzyme contributes to the mitochondrial tricarboxylic acid cycle (TCA) in PCa. Mass spectrometry-based metabolomics confirmed this hypothesis, showing that inhibition of IDH1 impairs carbon flux into the TCA cycle. Consequently, inhibition of IDH1 decreased PCa cell proliferation in vitro and in vivo. These results demonstrate that PCa cells have a hybrid cytoplasmic-mitochondrial TCA cycle that depends on IDH1. This metabolic enzyme represents a metabolic vulnerability of PCa cells and a potential new therapeutic target.
Collapse
Affiliation(s)
- Kevin Gonthier
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Department of Molecular Medicine, Faculty of MedicineUniversité LavalQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Cindy Weidmann
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Line Berthiaume
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Cynthia Jobin
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Department of Molecular Medicine, Faculty of MedicineUniversité LavalQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Aurélie Lacouture
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Department of Molecular Medicine, Faculty of MedicineUniversité LavalQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Camille Lafront
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Department of Molecular Medicine, Faculty of MedicineUniversité LavalQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Mario Harvey
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Bertrand Neveu
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Oncology AxisCentre de recherche du CHU de Québec – Université LavalCanada
| | - Jérémy Loehr
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Alain Bergeron
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Oncology AxisCentre de recherche du CHU de Québec – Université LavalCanada
- Department of Surgery, Faculty of MedicineUniversité LavalQuébecCanada
| | - Yves Fradet
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Oncology AxisCentre de recherche du CHU de Québec – Université LavalCanada
- Department of Surgery, Faculty of MedicineUniversité LavalQuébecCanada
| | - Louis Lacombe
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Oncology AxisCentre de recherche du CHU de Québec – Université LavalCanada
- Department of Surgery, Faculty of MedicineUniversité LavalQuébecCanada
| | - Julie Riopel
- Anatomopathology Service, Department of Laboratory MedicineCHU de Québec – Université LavalCanada
| | - Éva Latulippe
- Department of PathologyCHU de Québec – Université LavalCanada
| | - Chantal Atallah
- Department of PathologyCHU de Québec – Université LavalCanada
| | - Michael Shum
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Department of Molecular Medicine, Faculty of MedicineUniversité LavalQuébecCanada
| | - Jean‐Philippe Lambert
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Department of Molecular Medicine, Faculty of MedicineUniversité LavalQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Big Data Research CenterUniversité LavalQuébecQCCanada
| | - Frédéric Pouliot
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Oncology AxisCentre de recherche du CHU de Québec – Université LavalCanada
- Department of Surgery, Faculty of MedicineUniversité LavalQuébecCanada
| | - Martin Pelletier
- Infectious and Immune Disease AxisCHU de Québec‐Université Laval Research CenterCanada
- ARThrite Research CenterUniversité LavalQuébecQCCanada
- Department of Microbiology‐Infectious Diseases and Immunology, Faculty of MedicineUniversité LavalQuébecQCCanada
| | - Étienne Audet‐Walsh
- Endocrinology – Nephrology Research AxisCHU de Québec‐Université Laval Research CenterCanada
- Department of Molecular Medicine, Faculty of MedicineUniversité LavalQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| |
Collapse
|
5
|
Zeković M, Bumbaširević U, Živković M, Pejčić T. Alteration of Lipid Metabolism in Prostate Cancer: Multifaceted Oncologic Implications. Int J Mol Sci 2023; 24:ijms24021391. [PMID: 36674910 PMCID: PMC9863986 DOI: 10.3390/ijms24021391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Cancer is increasingly recognized as an extraordinarily heterogeneous disease featuring an intricate mutational landscape and vast intra- and intertumor variability on both genetic and phenotypic levels. Prostate cancer (PCa) is the second most prevalent malignant disease among men worldwide. A single metabolic program cannot epitomize the perplexing reprogramming of tumor metabolism needed to sustain the stemness of neoplastic cells and their prominent energy-consuming functional properties, such as intensive proliferation, uncontrolled growth, migration, and invasion. In cancerous tissue, lipids provide the structural integrity of biological membranes, supply energy, influence the regulation of redox homeostasis, contribute to plasticity, angiogenesis and microenvironment reshaping, mediate the modulation of the inflammatory response, and operate as signaling messengers, i.e., lipid mediators affecting myriad processes relevant for the development of the neoplasia. Comprehensive elucidation of the lipid metabolism alterations in PCa, the underlying regulatory mechanisms, and their implications in tumorigenesis and the progression of the disease are gaining growing research interest in the contemporary urologic oncology. Delineation of the unique metabolic signature of the PCa featuring major aberrant pathways including de novo lipogenesis, lipid uptake, storage and compositional reprogramming may provide novel, exciting, and promising avenues for improving diagnosis, risk stratification, and clinical management of such a complex and heterogeneous pathology.
Collapse
Affiliation(s)
- Milica Zeković
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Uros Bumbaširević
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marko Živković
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Tomislav Pejčić
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
6
|
Dai X, Thompson EW, Ostrikov K(K. Receptor-Mediated Redox Imbalance: An Emerging Clinical Avenue against Aggressive Cancers. Biomolecules 2022; 12:biom12121880. [PMID: 36551308 PMCID: PMC9775490 DOI: 10.3390/biom12121880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer cells are more vulnerable to abnormal redox fluctuations due to their imbalanced antioxidant system, where cell surface receptors sense stress and trigger intracellular signal relay. As canonical targets of many targeted therapies, cell receptors sensitize the cells to specific drugs. On the other hand, cell target mutations are commonly associated with drug resistance. Thus, exploring effective therapeutics targeting diverse cell receptors may open new clinical avenues against aggressive cancers. This paper uses focused case studies to reveal the intrinsic relationship between the cell receptors of different categories and the primary cancer hallmarks that are associated with the responses to external or internal redox perturbations. Cold atmospheric plasma (CAP) is examined as a promising redox modulation medium and highly selective anti-cancer therapeutic modality featuring dynamically varying receptor targets and minimized drug resistance against aggressive cancers.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Correspondence:
| | - Erik W. Thompson
- School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Kostya (Ken) Ostrikov
- School of Chemistry, Physics and Center for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| |
Collapse
|
7
|
Pei Y, Ning R, Hu W, Li P, Zhang Z, Deng Y, Hong Z, Sun Y, Guo X, Zhang Q. Carbon Ion Radiotherapy Induce Metabolic Inhibition After Functional Imaging-Guided Simultaneous Integrated Boost for Prostate Cancer. Front Oncol 2022; 12:845583. [PMID: 35936669 PMCID: PMC9354483 DOI: 10.3389/fonc.2022.845583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeAs local recurrence remains a challenge and the advantages of the simultaneous integrated boost (SIB) technique have been validated in photon radiotherapy, we applied the SIB technique to CIRT. The aim was to investigate the metabolomic changes of the CIRT with concurrent androgen deprivation therapy (ADT) in localized prostate cancer (PCa) and the unique metabolic effect of the SIB technique.Material and MethodsThis study enrolled 24 pathologically confirmed PCa patients. All patients went through CIRT with concurrent ADT. The gross target volume (GTV) boost was defined as positive lesions on both 68Ga-PSMA PET/CT and mpMRI images. Urine samples collected before and after CIRT were analyzed by the Q-TOF UPLC-MS/MS system. R platform and MetDNA were used for peak detection and identification. Statistical analysis and metabolic pathway analysis were performed on Metaboanalyst.ResultsThe metabolite profiles were significantly altered after CIRT. The most significantly altered metabolic pathway is PSMA participated alanine, aspartate and glutamate metabolism. Metabolites in this pathway showed a trend to be better suppressed in the SIB group. A total of 11 identified metabolites were significantly discriminative between two groups and all of them were better down-regulated in the SIB group. Meanwhile, among these metabolites, three metabolites in DNA damage and repair related purine metabolism were down-regulated to a greater extent in the SIB group.ConclusionMetabolic dysfunction was one of the typical characteristics of PCa. CIRT with ADT showed a powerful inhibition of PCa metabolism, especially in PSMA participated metabolic pathway. The SIB CIRT showed even better performance on down-regulation of most metabolism than uniform-dose-distribution CIRT. Meanwhile, the SIB CIRT also showed its unique superiority to inhibit purine metabolism. PSMA PET/CT guided SIB CIRT showed its potentials to further benefit PCa patients.
Collapse
Affiliation(s)
- Yulei Pei
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
| | - Renli Ning
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Wei Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
| | - Ping Li
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
| | - Yong Deng
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Zhengshan Hong
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Yun Sun
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- *Correspondence: Qing Zhang, ; Xiaomao Guo, ; Yun Sun,
| | - Xiaomao Guo
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- *Correspondence: Qing Zhang, ; Xiaomao Guo, ; Yun Sun,
| | - Qing Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy lon Radiation Therapy, Shanghai, China
- *Correspondence: Qing Zhang, ; Xiaomao Guo, ; Yun Sun,
| |
Collapse
|
8
|
Resurreccion EP, Fong KW. The Integration of Metabolomics with Other Omics: Insights into Understanding Prostate Cancer. Metabolites 2022; 12:metabo12060488. [PMID: 35736421 PMCID: PMC9230859 DOI: 10.3390/metabo12060488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Our understanding of prostate cancer (PCa) has shifted from solely caused by a few genetic aberrations to a combination of complex biochemical dysregulations with the prostate metabolome at its core. The role of metabolomics in analyzing the pathophysiology of PCa is indispensable. However, to fully elucidate real-time complex dysregulation in prostate cells, an integrated approach based on metabolomics and other omics is warranted. Individually, genomics, transcriptomics, and proteomics are robust, but they are not enough to achieve a holistic view of PCa tumorigenesis. This review is the first of its kind to focus solely on the integration of metabolomics with multi-omic platforms in PCa research, including a detailed emphasis on the metabolomic profile of PCa. The authors intend to provide researchers in the field with a comprehensive knowledge base in PCa metabolomics and offer perspectives on overcoming limitations of the tool to guide future point-of-care applications.
Collapse
Affiliation(s)
- Eleazer P. Resurreccion
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA;
| | - Ka-wing Fong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA;
- Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA
- Correspondence: ; Tel.: +1-859-562-3455
| |
Collapse
|
9
|
Frégeau-Proulx L, Lacouture A, Berthiaume L, Weidmann C, Harvey M, Gonthier K, Pelletier JF, Neveu B, Jobin C, Bastien D, Bergeron A, Fradet Y, Lacombe L, Laverdière I, Atallah C, Pouliot F, Audet-Walsh É. Multiple metabolic pathways fuel the truncated tricarboxylic acid cycle of the prostate to sustain constant citrate production and secretion. Mol Metab 2022; 62:101516. [PMID: 35598879 PMCID: PMC9168698 DOI: 10.1016/j.molmet.2022.101516] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 11/15/2022] Open
Abstract
Objective The prostate is metabolically unique: it produces high levels of citrate for secretion via a truncated tricarboxylic acid (TCA) cycle to maintain male fertility. In prostate cancer (PCa), this phenotype is reprogrammed, making it an interesting therapeutic target. However, how the truncated prostate TCA cycle works is still not completely understood. Methods We optimized targeted metabolomics in mouse and human organoid models in ex vivo primary culture. We then used stable isotope tracer analyses to identify the pathways that fuel citrate synthesis. Results First, mouse and human organoids were shown to recapitulate the unique citrate-secretory program of the prostate, thus representing a novel model that reproduces this unusual metabolic profile. Using stable isotope tracer analysis, several key nutrients were shown to allow the completion of the prostate TCA cycle, revealing a much more complex metabolic profile than originally anticipated. Indeed, along with the known pathway of aspartate replenishing oxaloacetate, glutamine was shown to fuel citrate synthesis through both glutaminolysis and reductive carboxylation in a GLS1-dependent manner. In human organoids, aspartate entered the TCA cycle at the malate entry point, upstream of oxaloacetate. Our results demonstrate that the citrate-secretory phenotype of prostate organoids is supported by the known aspartate–oxaloacetate–citrate pathway, but also by at least three additional pathways: glutaminolysis, reductive carboxylation, and aspartate–malate conversion. Conclusions Our results add a significant new dimension to the prostate citrate-secretory phenotype, with at least four distinct pathways being involved in citrate synthesis. Better understanding this distinctive citrate metabolic program will have applications in both male fertility as well as in the development of novel targeted anti-metabolic therapies for PCa. Targeted metabolomics and stable isotope tracer analysis were optimized in mouse and human prostate organoids. Organoids recapitulate the unique citrate-secretory phenotype of the prostate. Glutamine fuels citrate synthesis for secretion by glutaminolysis and reductive carboxylation. Aspartate enters the TCA cycle at different entry points in mouse and human prostate organoids for citrate production. We revealed a much more complex TCA cycle in the prostate than originally anticipated.
Collapse
Affiliation(s)
- Lilianne Frégeau-Proulx
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Aurélie Lacouture
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Line Berthiaume
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Cindy Weidmann
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Mario Harvey
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Kevin Gonthier
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Jean-François Pelletier
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada
| | - Bertrand Neveu
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada
| | - Cynthia Jobin
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Dominic Bastien
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada
| | - Alain Bergeron
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Yves Fradet
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Louis Lacombe
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Isabelle Laverdière
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Faculty of Pharmacy, Université Laval, Québec, QC, Canada; Department of Pharmacy, CHU de Québec - Université Laval, Québec, QC, Canada
| | - Chantal Atallah
- Department of Pathology, CHU de Québec - Université Laval, Québec, QC, Canada
| | - Frédéric Pouliot
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada; Oncology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Axis, CHU de Québec - Université Laval Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada.
| |
Collapse
|
10
|
Carbon sources and pathways for citrate secreted by human prostate cancer cells determined by NMR tracing and metabolic modeling. Proc Natl Acad Sci U S A 2022; 119:e2024357119. [PMID: 35353621 PMCID: PMC9168453 DOI: 10.1073/pnas.2024357119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The human prostate accumulates high luminal citrate levels to serve sperm viability. There is only indirect qualitative evidence about metabolic pathways and carbon sources maintaining these levels. Human citrate-secreting prostate cancer cells were supplied with 13C-labeled substrates, and NMR spectra of extracellular fluid were recorded. We report absolute citrate production rates of prostate cells and direct evidence that glucose is the main carbon source for secreted citrate. Pyruvate carboxylase provides sufficient anaplerotic carbons to support citrate secretion. Glutamine carbons exchange with carbons for secreted citrate but are likely not involved in its net synthesis. Moreover, we developed metabolic models employing the 13C distribution in extracellular citrate as input to assess intracellular pathways followed by carbons toward citrate. Prostate epithelial cells have the unique capacity to secrete large amounts of citrate, but the carbon sources and metabolic pathways that maintain this production are not well known. We mapped potential pathways for citrate carbons in the human prostate cancer metastasis cell lines LNCaP and VCaP, for which we first established that they secrete citrate (For LNCaP 5.6 ± 0.9 nmol/h per 106 cells). Using 13C-labeled substrates, we traced the incorporation of 13C into citrate by NMR of extracellular fluid. Our results provide direct evidence that glucose is a main carbon source for secreted citrate. We also demonstrate that carbons from supplied glutamine flow via oxidative Krebs cycle and reductive carboxylation routes to positions in secreted citrate but likely do not contribute to its net synthesis. The potential anaplerotic carbon sources aspartate and asparagine did not contribute to citrate carbons. We developed a quantitative metabolic model employing the 13C distribution in extracellular citrate after 13C glucose and pyruvate application to assess intracellular pathways of carbons for secreted citrate. From this model, it was estimated that in LNCaP about 21% of pyruvate entering the Krebs cycle is converted via pyruvate carboxylase as an anaplerotic route at a rate more than sufficient to compensate carbon loss of this cycle by citrate secretion. This model provides an estimation of the fraction of molecules, including citrate, leaving the Krebs cycle at every turn. The measured ratios of 13C atoms at different positions in extracellular citrate may serve as biomarkers for (malignant) epithelial cell metabolism.
Collapse
|
11
|
He Z, Gao Y, Li T, Yu C, Ou L, Luo C. HepaCAM‑PIK3CA axis regulates the reprogramming of glutamine metabolism to inhibit prostate cancer cell proliferation. Int J Oncol 2022; 60:37. [PMID: 35191516 PMCID: PMC8878713 DOI: 10.3892/ijo.2022.5327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Energy metabolism reprogramming is becoming an increasingly important hallmark of cancer. Specifically, cancers tend to undergo metabolic reprogramming to upregulate a cell-dependent glutamine (Gln) metabolism. Notably, hepatocellular cell adhesion molecule (HepaCAM) has been previously reported to serve a key role as a tumour suppressor. However, the possible regulatory role of HepaCAM in Gln metabolism in prostate cancer (PCa) remains poorly understood. In the present study, bioinformatics analysis predicted a significant negative correlation among the expression of HepaCAM, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA), glutaminase (GLS) and solute carrier family 1 member 5 (SLC1A5), components of Gln metabolism, in clinical and genomic datasets. Immunohistochemistry results verified a negative correlation between HepaCAM and PIK3CA expression in PCa tissues. Subsequently, liquid chromatography-tandem mass spectrometry (LC-MS/MS) and gas chromatography-mass spectrometry (GC-MS) assays were performed, and the results revealed markedly reduced levels of Gln and metabolic flux in the blood samples of patients with PCa and in PCa cells. Mechanistically, overexpression of HepaCAM inhibited Gln metabolism and proliferation by regulating PIK3CA in PCa cells. In addition, Gln metabolism was discovered to be stress-resistant in PCa cells, since the expression levels of GLS and SLC1A5 remained high for a period of time after Gln starvation. However, overexpression of HepaCAM reversed this resistance to some extent. Additionally, alpelisib, a specific inhibitor of PIK3CA, effectively potentiated the inhibitory effects of HepaCAM overexpression on Gln metabolism and cell proliferation through mass spectrometry and CCK-8 experiments. In addition, the inhibitory effect of PIK3CA on the growth of tumor tissue in nude mice was also confirmed by immunohistochemistry in vivo. To conclude, the results from the present study revealed an abnormal Gln metabolic profile in the blood samples of patients with PCa, suggesting that it can be applied as a clinical diagnostic tool for PCa. Additionally, a key role of the HepaCAM/PIK3CA axis in regulating Gln metabolism, cell proliferation and tumour growth was identified. The combination of alpelisib treatment with the upregulation of HepaCAM expression may serve as a novel method for treating patients with PCa.
Collapse
Affiliation(s)
- Zhenting He
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yingying Gao
- Department of Laboratory Diagnosis, People's Hospital of Chongqing Banan District, Chongqing 401320, P.R. China
| | - Ting Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chaowen Yu
- Center for Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Liping Ou
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chunli Luo
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
12
|
Labanca E, Yang J, Shepherd PDA, Wan X, Starbuck MW, Guerra LD, Anselmino N, Bizzotto JA, Dong J, Chinnaiyan AM, Ravoori MK, Kundra V, Broom BM, Corn PG, Troncoso P, Gueron G, Logothethis CJ, Navone NM. Fibroblast Growth Factor Receptor 1 Drives the Metastatic Progression of Prostate Cancer. Eur Urol Oncol 2021; 5:164-175. [PMID: 34774481 DOI: 10.1016/j.euo.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND No curative therapy is currently available for metastatic prostate cancer (PCa). The diverse mechanisms of progression include fibroblast growth factor (FGF) axis activation. OBJECTIVE To investigate the molecular and clinical implications of fibroblast growth factor receptor 1 (FGFR1) and its isoforms (α/β) in the pathogenesis of PCa bone metastases. DESIGN, SETTING, AND PARTICIPANTS In silico, in vitro, and in vivo preclinical approaches were used. RNA-sequencing and immunohistochemical (IHC) studies in human samples were conducted. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS In mice, bone metastases (chi-square/Fisher's test) and survival (Mantel-Cox) were assessed. In human samples, FGFR1 and ladinin 1 (LAD1) analysis associated with PCa progression were evaluated (IHC studies, Fisher's test). RESULTS AND LIMITATIONS FGFR1 isoform expression varied among PCa subtypes. Intracardiac injection of mice with FGFR1-expressing PC3 cells reduced mouse survival (α, p < 0.0001; β, p = 0.032) and increased the incidence of bone metastases (α, p < 0.0001; β, p = 0.02). Accordingly, IHC studies of human castration-resistant PCa (CRPC) bone metastases revealed significant enrichment of FGFR1 expression compared with treatment-naïve, nonmetastatic primary tumors (p = 0.0007). Expression of anchoring filament protein LAD1 increased in FGFR1-expressing PC3 cells and was enriched in human CRPC bone metastases (p = 0.005). CONCLUSIONS FGFR1 expression induces bone metastases experimentally and is significantly enriched in human CRPC bone metastases, supporting its prometastatic effect in PCa. LAD1 expression, found in the prometastatic PCa cells expressing FGFR1, was also enriched in CRPC bone metastases. Our studies support and provide a roadmap for the development of FGFR blockade for advanced PCa. PATIENT SUMMARY We studied the role of fibroblast growth factor receptor 1 (FGFR1) in prostate cancer (PCa) progression. We found that PCa cells with high FGFR1 expression increase metastases and that FGFR1 expression is increased in human PCa bone metastases, and identified genes that could participate in the metastases induced by FGFR1. These studies will help pinpoint PCa patients who use fibroblast growth factor to progress and will benefit by the inhibition of this pathway.
Collapse
Affiliation(s)
- Estefania Labanca
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jun Yang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter D A Shepherd
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xinhai Wan
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael W Starbuck
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Leah D Guerra
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicolas Anselmino
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan A Bizzotto
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jiabin Dong
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Murali K Ravoori
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vikas Kundra
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bradley M Broom
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul G Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Geraldine Gueron
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Christopher J Logothethis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nora M Navone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Singh R, Mills IG. The Interplay Between Prostate Cancer Genomics, Metabolism, and the Epigenome: Perspectives and Future Prospects. Front Oncol 2021; 11:704353. [PMID: 34660272 PMCID: PMC8511631 DOI: 10.3389/fonc.2021.704353] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is a high-incidence cancer, often detected late in life. The prostate gland is an accessory gland that secretes citrate; an impaired citrate secretion reflects imbalances in the activity of enzymes in the TCA Cycle in mitochondria. Profiling studies on prostate tumours have identified significant metabolite, proteomic, and transcriptional modulations with an increased mitochondrial metabolic activity associated with localised prostate cancer. Here, we focus on the androgen receptor, c-Myc, phosphatase and tensin Homolog deleted on chromosome 10 (PTEN), and p53 as amongst the best-characterised genomic drivers of prostate cancer implicated in metabolic dysregulation and prostate cancer progression. We outline their impact on metabolic function before discussing how this may affect metabolite pools and in turn chromatin structure and the epigenome. We reflect on some recent literature indicating that mitochondrial mutations and OGlcNAcylation may also contribute to this crosstalk. Finally, we discuss the technological challenges of assessing crosstalk given the significant differences in the spatial sensitivity and throughput of genomic and metabolomic profiling approaches.
Collapse
Affiliation(s)
- Reema Singh
- Nuffield Department of Surgical Sciences John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Ian G. Mills
- Nuffield Department of Surgical Sciences John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Patrick G Johnston Centre for Cancer Research, Queen’s University of Belfast, Belfast, United Kingdom
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
14
|
Mamouni K, Kallifatidis G, Lokeshwar BL. Targeting Mitochondrial Metabolism in Prostate Cancer with Triterpenoids. Int J Mol Sci 2021; 22:2466. [PMID: 33671107 PMCID: PMC7957768 DOI: 10.3390/ijms22052466] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Metabolic reprogramming is a hallmark of malignancy. It implements profound metabolic changes to sustain cancer cell survival and proliferation. Although the Warburg effect is a common feature of metabolic reprogramming, recent studies have revealed that tumor cells also depend on mitochondrial metabolism. Due to the essential role of mitochondria in metabolism and cell survival, targeting mitochondria in cancer cells is an attractive therapeutic strategy. However, the metabolic flexibility of cancer cells may enable the upregulation of compensatory pathways, such as glycolysis, to support cancer cell survival when mitochondrial metabolism is inhibited. Thus, compounds capable of targeting both mitochondrial metabolism and glycolysis may help overcome such resistance mechanisms. Normal prostate epithelial cells have a distinct metabolism as they use glucose to sustain physiological citrate secretion. During the transformation process, prostate cancer cells consume citrate to mainly power oxidative phosphorylation and fuel lipogenesis. A growing number of studies have assessed the impact of triterpenoids on prostate cancer metabolism, underlining their ability to hit different metabolic targets. In this review, we critically assess the metabolic transformations occurring in prostate cancer cells. We will then address the opportunities and challenges in using triterpenoids as modulators of prostate cancer cell metabolism.
Collapse
Affiliation(s)
- Kenza Mamouni
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA;
- Research Service, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Georgios Kallifatidis
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA;
- Research Service, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Biological Sciences, Augusta University, Augusta, GA 30912, USA
| | - Bal L. Lokeshwar
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA;
- Research Service, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
15
|
Ingram LM, Finnerty MC, Mansoura M, Chou CW, Cummings BS. Identification of lipidomic profiles associated with drug-resistant prostate cancer cells. Lipids Health Dis 2021; 20:15. [PMID: 33596934 PMCID: PMC7890620 DOI: 10.1186/s12944-021-01437-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The association of circulating lipids with clinical outcomes of drug-resistant castration-resistant prostate cancer (DR-CRPC) is not fully understood. While it is known that increases in select lipids correlate to decreased survival, neither the mechanisms mediating these alterations nor the correlation of resistance to drug treatments is well characterized. METHODS This gap-in-knowledge was addressed using in vitro models of non-cancerous, hormone-sensitive, CRPC and drug-resistant cell lines combined with quantitative LC-ESI-Orbitrap-MS (LC-ESI-MS/MS) lipidomic analysis and subsequent analysis such as Metaboanalyst and Lipid Pathway Enrichment Analysis (LIPEA). RESULTS Several lipid regulatory pathways were identified that are associated with Docetaxel resistance in prostate cancer (PCa). These included those controlling glycerophospholipid metabolism, sphingolipid signaling and ferroptosis. In total, 7460 features were identified as being dysregulated between the cell lines studied, and 21 lipid species were significantly altered in drug-resistant cell lines as compared to nonresistant cell lines. Docetaxel resistance cells (PC3-Rx and DU145-DR) had higher levels of phosphatidylcholine (PC), oxidized lipid species, phosphatidylethanolamine (PE), and sphingomyelin (SM) as compared to parent control cells (PC-3 and DU-145). Alterations were also identified in the levels of phosphatidic acid (PA) and diacylglyceride (DAG), whose levels are regulated by Lipin (LPIN), a phosphatidic acid phosphatase that converts PA to DAG. Data derived from cBioPortal demonstrated a population of PCa patients expressing mutations aligning with amplification of LPIN1, LPIN2 and LPIN3 genes. Lipin amplification in these genes correlated to decreased survival in these patients. Lipin-1 mRNA expression also showed a similar trend in PCa patient data. Lipin-1, but not Lipin-2 or - 3, was detected in several prostate cancer cells, and was increased in 22RV1 and PC-3 cell lines. The increased expression of Lipin-1 in these cells correlated with the level of PA. CONCLUSION These data identify lipids whose levels may correlate to Docetaxel sensitivity and progression of PCa. The data also suggest a correlation between the expression of Lipin-1 in cells and patients with regards to prostate cancer cell aggressiveness and patient survivability. Ultimately, these data may be useful for identifying markers of lethal and/or metastatic prostate cancer.
Collapse
Affiliation(s)
- Lishann M Ingram
- Pharmaceutical and Biomedical Sciences, 450 College of Pharmacy South, University of Georgia, Athens, GA, 30602, USA
| | - Morgan C Finnerty
- Pharmaceutical and Biomedical Sciences, 450 College of Pharmacy South, University of Georgia, Athens, GA, 30602, USA
| | - Maryam Mansoura
- Pharmaceutical and Biomedical Sciences, 450 College of Pharmacy South, University of Georgia, Athens, GA, 30602, USA
| | - Chau-Wen Chou
- Proteomics and Mass Spectrometry Facility (PAMS), Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Brian S Cummings
- Pharmaceutical and Biomedical Sciences, 450 College of Pharmacy South, University of Georgia, Athens, GA, 30602, USA.
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA.
| |
Collapse
|
16
|
Rai A, Noor S, Ahmad SI, Alajmi MF, Hussain A, Abbas H, Hasan GM. Recent Advances and Implication of Bioengineered Nanomaterials in Cancer Theranostics. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:91. [PMID: 33494239 PMCID: PMC7909769 DOI: 10.3390/medicina57020091] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023]
Abstract
Cancer is one of the most common causes of death and affects millions of lives every year. In addition to non-infectious carcinogens, infectious agents contribute significantly to increased incidence of several cancers. Several therapeutic techniques have been used for the treatment of such cancers. Recently, nanotechnology has emerged to advance the diagnosis, imaging, and therapeutics of various cancer types. Nanomaterials have multiple advantages over other materials due to their small size and high surface area, which allow retention and controlled drug release to improve the anti-cancer property. Most cancer therapies have been known to damage healthy cells due to poor specificity, which can be avoided by using nanosized particles. Nanomaterials can be combined with various types of biomaterials to make it less toxic and improve its biocompatibility. Based on these properties, several nanomaterials have been developed which possess excellent anti-cancer efficacy potential and improved diagnosis. This review presents the latest update on novel nanomaterials used to improve the diagnostic and therapeutic of pathogen-associated and non-pathogenic cancers. We further highlighted mechanistic insights into their mode of action, improved features, and limitations.
Collapse
Affiliation(s)
- Ayushi Rai
- Department of Nanoscience, Central University of Gujarat, Sector 29, Gandhinagar 382030, India;
| | - Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India;
| | - Syed Ishraque Ahmad
- Department of Chemistry, Zakir Husain Delhi College, University of Delhi, New Delhi 110002, India;
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.)
| | - Hashim Abbas
- Department of Medicine, Nottingham University Hospitals, NHS Trust, Nottingham NG7 2UH, UK;
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
17
|
Sawant Dessai A, Dominguez MP, Chen UI, Hasper J, Prechtl C, Yu C, Katsuta E, Dai T, Zhu B, Jung SY, Putluri N, Takabe K, Zhang XHF, O'Malley BW, Dasgupta S. Transcriptional Repression of SIRT3 Potentiates Mitochondrial Aconitase Activation to Drive Aggressive Prostate Cancer to the Bone. Cancer Res 2021; 81:50-63. [PMID: 33115805 PMCID: PMC7878313 DOI: 10.1158/0008-5472.can-20-1708] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/08/2020] [Accepted: 10/21/2020] [Indexed: 11/16/2022]
Abstract
Metabolic dysregulation is a known hallmark of cancer progression, yet the oncogenic signals that promote metabolic adaptations to drive metastatic cancer remain unclear. Here, we show that transcriptional repression of mitochondrial deacetylase sirtuin 3 (SIRT3) by androgen receptor (AR) and its coregulator steroid receptor coactivator-2 (SRC-2) enhances mitochondrial aconitase (ACO2) activity to favor aggressive prostate cancer. ACO2 promoted mitochondrial citrate synthesis to facilitate de novo lipogenesis, and genetic ablation of ACO2 reduced total lipid content and severely repressed in vivo prostate cancer progression. A single acetylation mark lysine258 on ACO2 functioned as a regulatory motif, and the acetylation-deficient Lys258Arg mutant was enzymatically inactive and failed to rescue growth of ACO2-deficient cells. Acetylation of ACO2 was reversibly regulated by SIRT3, which was predominantly repressed in many tumors including prostate cancer. Mechanistically, SRC-2-bound AR formed a repressive complex by recruiting histone deacetylase 2 to the SIRT3 promoter, and depletion of SRC-2 enhanced SIRT3 expression and simultaneously reduced acetylated ACO2. In human prostate tumors, ACO2 activity was significantly elevated, and increased expression of SRC-2 with concomitant reduction of SIRT3 was found to be a genetic hallmark enriched in prostate cancer metastatic lesions. In a mouse model of spontaneous bone metastasis, suppression of SRC-2 reactivated SIRT3 expression and was sufficient to abolish prostate cancer colonization in the bone microenvironment, implying this nuclear-mitochondrial regulatory axis is a determining factor for metastatic competence. SIGNIFICANCE: This study highlights the importance of mitochondrial aconitase activity in the development of advanced metastatic prostate cancer and suggests that blocking SRC-2 to enhance SIRT3 expression may be therapeutically valuable. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/1/50/F1.large.jpg.
Collapse
Affiliation(s)
- Abhisha Sawant Dessai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Uan-I Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - John Hasper
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Christian Prechtl
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Cuijuan Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Eriko Katsuta
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Tao Dai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Bokai Zhu
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Biochemistry, Baylor College of Medicine, Houston, Texas
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| |
Collapse
|
18
|
Cardoso HJ, Carvalho TMA, Fonseca LRS, Figueira MI, Vaz CV, Socorro S. Revisiting prostate cancer metabolism: From metabolites to disease and therapy. Med Res Rev 2020; 41:1499-1538. [PMID: 33274768 DOI: 10.1002/med.21766] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/24/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PCa), one of the most commonly diagnosed cancers worldwide, still presents important unmet clinical needs concerning treatment. In the last years, the metabolic reprogramming and the specificities of tumor cells emerged as an exciting field for cancer therapy. The unique features of PCa cells metabolism, and the activation of specific metabolic pathways, propelled the use of metabolic inhibitors for treatment. The present work revises the knowledge of PCa metabolism and the metabolic alterations that underlie the development and progression of the disease. A focus is given to the role of bioenergetic sources, namely, glucose, lipids, and glutamine sustaining PCa cell survival and growth. Moreover, it is described as the action of oncogenes/tumor suppressors and sex steroid hormones in the metabolic reprogramming of PCa. Finally, the status of PCa treatment based on the inhibition of metabolic pathways is presented. Globally, this review updates the landscape of PCa metabolism, highlighting the critical metabolic alterations that could have a clinical and therapeutic interest.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Tiago M A Carvalho
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Lara R S Fonseca
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
19
|
He J, Mao Y, Huang W, Li M, Zhang H, Qing Y, Lu S, Xiao H, Li K. Methylcrotonoyl-CoA Carboxylase 2 Promotes Proliferation, Migration and Invasion and Inhibits Apoptosis of Prostate Cancer Cells Through Regulating GLUD1-P38 MAPK Signaling Pathway. Onco Targets Ther 2020; 13:7317-7327. [PMID: 32801758 PMCID: PMC7395692 DOI: 10.2147/ott.s249906] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 07/08/2020] [Indexed: 12/25/2022] Open
Abstract
Purpose Prostate cancer (PCa) is the most common cancer in American men, and the mechanisms of development and progression are still not completely clear. Methylcrotonoyl-CoA carboxylase 2 (MCCC2) was previously identified overexpressed in PCa with lymph node metastasis, but its specific role and mechanisms need further investigation. This study aimed to investigate the role of MCCC2 in PCa cells and its underlying mechanisms. Materials and Methods Quantitative RT-PCR and Western blotting were used to detect MCCC2 mRNA and protein expression in normal prostate epithelium and cancerous cells. Upon manipulation of MCCC2 expression, cell proliferation was measured by CCK-8 assays and migration and invasion were determined by transwell assays. Changes of apoptosis, cell cycle and mitochondrial membrane potential were evaluated by flow cytometry. MCCC2-mediated signaling pathways were screened by bioinformatics and verified by RT-PCR and Western blotting. Finally, immunohistochemistry was performed to detect the expression of MCCC2 and glutamate dehydrogenase 1 (GLUD1) in PCa tissues to analyze their correlation. Results We demonstrated that MCCC2 promoted cell proliferation, migration and invasion but inhibited apoptosis in PCa cells. In addition, MCCC2 in 22Rv1 cells induced mitochondrial damage. In PCa tissues, MCCC2 overexpression associated with lymph node metastasis (P=0.001) and high Gleason scores (P<0.001). MCCC2 positively correlated with GLUD1 expression in PCa tissues (r=0.435, P<0.001). Ectopic overexpression of MCCC2 up-regulated GLUD1 and p38 MAPK expression, whereas inhibition of MCCC2 decreased GLUD1 and p38 MAPK expression. Conclusion MCCC2 exerts oncogenic function in PCa through regulating GLUD1-p38 MAPK signaling pathway, and it may be a potential treatment target.
Collapse
Affiliation(s)
- Jianwen He
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Yunhua Mao
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Wentao Huang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Mingzhao Li
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Huimin Zhang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Yunhao Qing
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Shuo Lu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Hengjun Xiao
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Ke Li
- Department of Urology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| |
Collapse
|
20
|
Chen X, Wei L, Yang L, Guo W, Guo Q, Zhou Y. Glycolysis inhibition and apoptosis induction in human prostate cancer cells by FV-429-mediated regulation of AR-AKT-HK2 signaling network. Food Chem Toxicol 2020; 143:111517. [PMID: 32619556 DOI: 10.1016/j.fct.2020.111517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 01/22/2023]
Abstract
Prostate cancer (PCa) depends on androgen receptor (AR) signaling to regulate cell metabolism, including glycolysis, and thereby promotes tumor growth. Glycolysis is overactive in PCa and associated with poor prognosis, but the therapeutic efficacy of glycolysis inhibitors has thus far been limited by their inability to induce cell death. FV-429, a flavonoid derivative of Wogonin, is a glycolysis inhibitor that has shown anti-cancer promise. In this study, we used FV-429 as an anti-PCa agent and investigated its mechanisms of action. In vitro, both the glycolytic ability and the viability of PCa cells were inhibited by FV-429. We found that FV-429 could induce mitochondrial dysfunction and apoptosis, with AKT-HK2 signaling pathway playing a key role. In addition, FV-429 had a pro-apoptotic effect on human prostate cancer cells that relied on the inhibition of AR expression and activity. In vivo, FV-429 exerted significant tumor-repressing activity with high safety in the xenograft model using LNCaP cells. In summary, we demonstrated that FV-429 induced glycolysis inhibition and apoptosis in human prostate cancer cells by downregulating the AR-AKT-HK2 signaling network, making FV-429 a promising candidate as one therapeutic agent for advanced PCa.
Collapse
Affiliation(s)
- Xian Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Liliang Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Wenjing Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| | - Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
21
|
Li T, Li Y, Liu T, Hu B, Li J, Liu C, Liu T, Li F. Mitochondrial PAK6 inhibits prostate cancer cell apoptosis via the PAK6-SIRT4-ANT2 complex. Theranostics 2020; 10:2571-2586. [PMID: 32194820 PMCID: PMC7052886 DOI: 10.7150/thno.42874] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/11/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale: P21-activated kinase 6 (PAK6) is a member of the class II PAKs family, which is a conserved family of serine/threonine kinases. Although the effects of PAK6 on many malignancies, especially in prostate cancer, have been studied for a long time, the role of PAK6 in mitochondria remains unknown. Methods: The expression of PAK6, SIRT4 and ANT2 in prostate cancer and adjacent non-tumor tissues was detected by immunohistochemistry. Immunofuorescence and immunoelectron microscopy were used to determine the subcellular localization of PAK6. Immunoprecipitation, immunofuorescence and ubiquitination assays were performed to determine how PAK6 regulates SIRT4, how SIRT4 regulates ANT2, and how PAK6 regulates ANT2. Flow cytometry detection and xenograft models were used to evaluate the impact of ANT2 mutant expression on the prostate cancer cell cycle and apoptosis regulation. Results: The present study revealed that the PAK6-SIRT4-ANT2 complex is involved in mitochondrial apoptosis in prostate cancer cells. It was found that PAK6 is mainly located in the mitochondrial inner membrane, in which PAK6 promotes SIRT4 ubiquitin-mediated proteolysis. Furthermore, SIRT4 deprives the ANT2 acetylation at K105 to promote its ubiquitination degradation. Hence, PAK6 adjusts the acetylation level of ANT2 through the PAK6-SIRT4-ANT2 pathway, in order to regulate the stability of ANT2. Meanwhile, PAK6 directly phosphorylates ANT2 atT107 to inhibit the apoptosis of prostate cancer cells. Therefore, the phosphorylation and deacetylation modifications of ANT2 are mutually regulated, leading to tumor growth in vivo. Consistently, these clinical prostate cancer tissue evaluations reveal that PAK6 is positively correlated with ANT2 expression, but negatively correlated with SIRT4. Conclusion: These present findings suggest the pivotal role of the PAK6-SIRT4-ANT2 complex in the apoptosis of prostate cancer. This complex could be a potential biomarker for the treatment and prognosis of prostate cancer.
Collapse
Affiliation(s)
- Tingting Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Tong Liu
- Medical Research Center, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Bingtao Hu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Jiabin Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Chen Liu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| | - Tao Liu
- Department of Urology, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, Shenyang 110122, Liaoning, China
| |
Collapse
|
22
|
Gonthier K, Poluri RTK, Audet-Walsh É. Functional genomic studies reveal the androgen receptor as a master regulator of cellular energy metabolism in prostate cancer. J Steroid Biochem Mol Biol 2019; 191:105367. [PMID: 31051242 DOI: 10.1016/j.jsbmb.2019.04.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/19/2022]
Abstract
Sex-steroid hormones have been investigated for decades for their oncogenic properties in hormone-dependent cancers. The increasing body of knowledge on the biological actions of androgens in prostate cancer has led to the development of several targeted therapies that still represent the standard of care for cancer patients to this day. In the prostate, androgens promote cellular differentiation and proper tissue development. These hormones also promote the aberrant proliferation and survival of prostate cancer cells. Over the past few years, sequencing technologies for functional genomic analyses have rapidly expanded, revealing novel functions of sex-steroid hormone receptors other than their classic roles. In this article, we will focus on transcriptomic- and genomic-based evidence that demonstrates the importance of the androgen receptor signaling in the regulation of prostate cancer cell metabolism. This is significant because the reprogramming of cell metabolism is a hallmark of cancer. In fact, it is clear now that the androgen receptor contributes to the reprogramming of specific cellular metabolic pathways that promote tumor growth and disease progression, including aerobic glycolysis, mitochondrial respiration, fatty acid ß-oxidation, and de novo lipid synthesis. Overall, beyond regulating development, differentiation, and proliferation, the androgen receptor is also a master regulator of cellular energy metabolism.
Collapse
Affiliation(s)
- Kevin Gonthier
- Department of Molecular Medicine, Axe Endocrinologie - Néphrologie du Centre de recherche du CHU de Québec, Canada; Centre de recherche sur le cancer - Université Laval, Canada
| | - Raghavendra Tejo Karthik Poluri
- Department of Molecular Medicine, Axe Endocrinologie - Néphrologie du Centre de recherche du CHU de Québec, Canada; Centre de recherche sur le cancer - Université Laval, Canada
| | - Étienne Audet-Walsh
- Department of Molecular Medicine, Axe Endocrinologie - Néphrologie du Centre de recherche du CHU de Québec, Canada; Centre de recherche sur le cancer - Université Laval, Canada.
| |
Collapse
|
23
|
Cui J, Wang L, Ren X, Zhang Y, Zhang H. LRPPRC: A Multifunctional Protein Involved in Energy Metabolism and Human Disease. Front Physiol 2019; 10:595. [PMID: 31178748 PMCID: PMC6543908 DOI: 10.3389/fphys.2019.00595] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 04/26/2019] [Indexed: 12/26/2022] Open
Abstract
The pentatricopeptide repeat (PPR) family plays a major role in RNA stability, regulation, processing, splicing, translation, and editing. Leucine-rich PPR-motif-containing protein (LRPPRC), a member of the PPR family, is a known gene mutation that causes Leigh syndrome French-Canadian. Recently, growing evidence has pointed out that LRPPRC dysregulation is related to various diseases ranging from tumors to viral infections. This review presents available published data on the LRPPRC protein function and its role in tumors and other diseases. As a multi-functional protein, LRPPRC regulates a myriad of biological processes, including energy metabolism and maturation and the export of nuclear mRNA. Overexpression of LRPPRC has been observed in various human tumors and is associated with poor prognosis. Downregulation of LRPPRC inhibits growth and invasion, induces apoptosis, and overcomes drug resistance in tumor cells. In addition, LRPPRC plays a potential role in Parkinson's disease, neurofibromatosis 1, viral infections, and venous thromboembolism. Further investigating these new functions of LRPPRC should provide novel opportunities for a better understanding of its pathological role in diseases from tumors to viral infections and as a potential biomarker and molecular target for disease treatment.
Collapse
Affiliation(s)
- Jie Cui
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Li Wang
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Xiaoyue Ren
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Yamin Zhang
- Department of Oncology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China.,College of General Practitioners, Xi'an Medical University, Xi'an, China
| | - Hongyi Zhang
- College of General Practitioners, Xi'an Medical University, Xi'an, China.,Department of Urology, The First Affiliated Hospital, Xi'an Medical University, Xi'an, China
| |
Collapse
|
24
|
Transporter and protease mediated delivery of platinum complexes for precision oncology. J Biol Inorg Chem 2019; 24:457-466. [DOI: 10.1007/s00775-019-01660-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/09/2019] [Indexed: 01/03/2023]
|
25
|
Gómez-Cebrián N, Rojas-Benedicto A, Albors-Vaquer A, López-Guerrero JA, Pineda-Lucena A, Puchades-Carrasco L. Metabolomics Contributions to the Discovery of Prostate Cancer Biomarkers. Metabolites 2019; 9:metabo9030048. [PMID: 30857149 PMCID: PMC6468766 DOI: 10.3390/metabo9030048] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer (PCa) is one of the most frequently diagnosed cancers and a leading cause of death among men worldwide. Despite extensive efforts in biomarker discovery during the last years, currently used clinical biomarkers are still lacking enough specificity and sensitivity for PCa early detection, patient prognosis, and monitoring. Therefore, more precise biomarkers are required to improve the clinical management of PCa patients. In this context, metabolomics has shown to be a promising and powerful tool to identify novel PCa biomarkers in biofluids. Thus, changes in polyamines, tricarboxylic acid (TCA) cycle, amino acids, and fatty acids metabolism have been reported in different studies analyzing PCa patients' biofluids. The review provides an up-to-date summary of the main metabolic alterations that have been described in biofluid-based studies of PCa patients, as well as a discussion regarding their potential to improve clinical PCa diagnosis and prognosis. Furthermore, a summary of the most significant findings reported in these studies and the connections and interactions between the different metabolic changes described has also been included, aiming to better describe the specific metabolic signature associated to PCa.
Collapse
Affiliation(s)
- Nuria Gómez-Cebrián
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain.
- Joint Research Unit in Clinical Metabolomics, Centro de Investigación Príncipe Felipe/Instituto de Investigación Sanitaria La Fe, Valencia 46012, Spain.
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, Valencia 46009, Spain.
| | - Ayelén Rojas-Benedicto
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain.
- Joint Research Unit in Clinical Metabolomics, Centro de Investigación Príncipe Felipe/Instituto de Investigación Sanitaria La Fe, Valencia 46012, Spain.
| | - Arturo Albors-Vaquer
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain.
- Joint Research Unit in Clinical Metabolomics, Centro de Investigación Príncipe Felipe/Instituto de Investigación Sanitaria La Fe, Valencia 46012, Spain.
| | | | - Antonio Pineda-Lucena
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, Valencia 46026, Spain.
- Joint Research Unit in Clinical Metabolomics, Centro de Investigación Príncipe Felipe/Instituto de Investigación Sanitaria La Fe, Valencia 46012, Spain.
| | - Leonor Puchades-Carrasco
- Joint Research Unit in Clinical Metabolomics, Centro de Investigación Príncipe Felipe/Instituto de Investigación Sanitaria La Fe, Valencia 46012, Spain.
| |
Collapse
|
26
|
Bok R, Lee J, Sriram R, Keshari K, Sukumar S, Daneshmandi S, Korenchan DE, Flavell RR, Vigneron DB, Kurhanewicz J, Seth P. The Role of Lactate Metabolism in Prostate Cancer Progression and Metastases Revealed by Dual-Agent Hyperpolarized 13C MRSI. Cancers (Basel) 2019; 11:cancers11020257. [PMID: 30813322 PMCID: PMC6406929 DOI: 10.3390/cancers11020257] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/08/2019] [Accepted: 02/20/2019] [Indexed: 01/11/2023] Open
Abstract
This study applied a dual-agent, 13C-pyruvate and 13C-urea, hyperpolarized 13C magnetic resonance spectroscopic imaging (MRSI) and multi-parametric (mp) 1H magnetic resonance imaging (MRI) approach in the transgenic adenocarcinoma of mouse prostate (TRAMP) model to investigate changes in tumor perfusion and lactate metabolism during prostate cancer development, progression and metastases, and after lactate dehydrogenase-A (LDHA) knock-out. An increased Warburg effect, as measured by an elevated hyperpolarized (HP) Lactate/Pyruvate (Lac/Pyr) ratio, and associated Ldha expression and LDH activity were significantly higher in high- versus low-grade TRAMP tumors and normal prostates. The hypoxic tumor microenvironment in high-grade tumors, as measured by significantly decreased HP 13C-urea perfusion and increased PIM staining, played a key role in increasing lactate production through increased Hif1α and then Ldha expression. Increased lactate induced Mct4 expression and an acidic tumor microenvironment that provided a potential mechanism for the observed high rate of lymph node (86%) and liver (33%) metastases. The Ldha knockdown in the triple-transgenic mouse model of prostate cancer resulted in a significant reduction in HP Lac/Pyr, which preceded a reduction in tumor volume or apparent water diffusion coefficient (ADC). The Ldha gene knockdown significantly reduced primary tumor growth and reduced lymph node and visceral metastases. These data suggested a metabolic transformation from low- to high-grade prostate cancer including an increased Warburg effect, decreased perfusion, and increased metastatic potential. Moreover, these data suggested that LDH activity and lactate are required for tumor progression. The lactate metabolism changes during prostate cancer provided the motivation for applying hyperpolarized 13C MRSI to detect aggressive disease at diagnosis and predict early therapeutic response.
Collapse
Affiliation(s)
- Robert Bok
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - Jessie Lee
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - Kayvan Keshari
- Department of Radiology, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY 10065, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA.
| | - Subramaniam Sukumar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - Saeed Daneshmandi
- Department of Medicine, Division of Interdisciplinary Medicine, Beth Israel Deaconess Medical Center, Beth Israel Cancer Center, Harvard Medical School, Boston, MA 02215, USA.
| | - David E Korenchan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - Daniel B Vigneron
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA.
| | - Pankaj Seth
- Department of Medicine, Division of Interdisciplinary Medicine, Beth Israel Deaconess Medical Center, Beth Israel Cancer Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
27
|
Vishwanath V, Mayer D, Fu D, Wnorowski A, Siddiqui MM. Hyperpolarized 13C magnetic resonance imaging, using metabolic imaging to improve the detection and management of prostate, bladder, and kidney urologic malignancies. Transl Androl Urol 2018; 7:855-863. [PMID: 30456188 PMCID: PMC6212626 DOI: 10.21037/tau.2018.08.27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Approximately 25% of the 2 million new cancer diagnoses in the United States in 2018 were comprised of malignancies of the urogenital system. Of these cancers, 75% occurred in the kidney/renal pelvis, prostate, and urinary bladder. Early diagnosis is beneficial to long-term survival. Currently, urologists rely heavily on computed tomography (CT), magnetic resonance imaging (MRI), ultrasound (US), and positron emission tomography (PET) to both diagnose and offer prognoses, but these techniques are limited in their resolution and are more effective when cancers have reached macroscopic size in later stages. Recent developments in cancer metabolomics have revealed that cancerous cells preferentially upregulate specific metabolic pathways as a means of conserving their resources and maximizing their growth potential. This has opened a new avenue for early diagnosis with much higher resolution, reliability, and accuracy through 13C hyperpolarized MRI. Preferential cancer pathways can be elucidated through this technique using 13C-labeled molecules utilized for energy generation and tumor growth. As these pathways are identified, targeted therapies are being designed to inhibit these pathways to allow for treatment that is cytotoxic to malignant cells but preserves native cells. In this paper, we review the current understanding of urologic cancer metabolomics, specifically in the kidney, prostate, and bladder. We will review the basic physics of MRI and demonstrate how hyperpolarized 13C MRI offers an innovative solution to early diagnosis as well as creates novel avenues for more targeted therapy.
Collapse
Affiliation(s)
- Vijay Vishwanath
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dirk Mayer
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dexue Fu
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amelia Wnorowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mohummad Minhaj Siddiqui
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.,The Veterans Health Administration Research and Development Service, Baltimore, MD, USA
| |
Collapse
|
28
|
Eidelman E, Tripathi H, Fu DX, Siddiqui MM. Linking cellular metabolism and metabolomics to risk-stratification of prostate cancer clinical aggressiveness and potential therapeutic pathways. Transl Androl Urol 2018; 7:S490-S497. [PMID: 30363493 PMCID: PMC6178321 DOI: 10.21037/tau.2018.04.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer treatment is based on the stratification of disease as low-, intermediate- or high-risk. This stratification has been largely based on anatomic pathology of the disease, as well as through the use of prostate specific antigen (PSA). However, despite this stratification, there remains heterogeneity within the current classification schema. Utilizing a metabolic approach may help to further establish novel biomolecular markers of disease aggressiveness. These markers may eventually be useful in not only the diagnosis of disease but in creating tumor specific targeted therapy for improved clinical outcomes.
Collapse
Affiliation(s)
- Eric Eidelman
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hemantkumar Tripathi
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - De-Xue Fu
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M Minhaj Siddiqui
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Gonzalez-Menendez P, Hevia D, Mayo JC, Sainz RM. The dark side of glucose transporters in prostate cancer: Are they a new feature to characterize carcinomas? Int J Cancer 2017; 142:2414-2424. [DOI: 10.1002/ijc.31165] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/01/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Pedro Gonzalez-Menendez
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| | - David Hevia
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| | - Juan C. Mayo
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| | - Rosa M. Sainz
- Department of Morphology and Cell Biology; Redox Biology Unit, University Institute of Oncology of Asturias (IUOPA). University of Oviedo. Facultad de Medicina.; Oviedo Spain
| |
Collapse
|
30
|
Eidelman E, Twum-Ampofo J, Ansari J, Siddiqui MM. The Metabolic Phenotype of Prostate Cancer. Front Oncol 2017; 7:131. [PMID: 28674679 PMCID: PMC5474672 DOI: 10.3389/fonc.2017.00131] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer is the most common non-cutaneous cancer in men in the United States. Cancer metabolism has emerged as a contemporary topic of great interest for improved mechanistic understanding of tumorigenesis. Prostate cancer is a disease model of great interest from a metabolic perspective. Prostatic tissue exhibits unique metabolic activity under baseline conditions. Benign prostate cells accumulate zinc, and this excess zinc inhibits citrate oxidation and metabolism within the citric acid cycle, effectively resulting in citrate production. Malignant cells, however, actively oxidize citrate and resume more typical citric acid cycle function. Of further interest, prostate cancer does not exhibit the Warburg effect, an increase in glucose uptake, seen in many other cancers. These cellular metabolic differences and others are of clinical interest as they present a variety of potential therapeutic targets. Furthermore, understanding of the metabolic profile differences between benign prostate versus low- and high-grade prostate cancers also represents an avenue to better understand cancer progression and potentially develop new diagnostic testing. In this paper, we review the current state of knowledge on the metabolic phenotypes of prostate cancer.
Collapse
Affiliation(s)
- Eric Eidelman
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore MD, United States
| | - Jeffrey Twum-Ampofo
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore MD, United States
| | - Jamal Ansari
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore MD, United States
| | - Mohummad Minhaj Siddiqui
- Division of Urology, Department of Surgery, University of Maryland School of Medicine, Baltimore MD, United States.,The Veterans Health Administration Research and Development Service, Baltimore, MD, United States.,Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore MD, United States
| |
Collapse
|
31
|
Naponelli V, Ramazzina I, Lenzi C, Bettuzzi S, Rizzi F. Green Tea Catechins for Prostate Cancer Prevention: Present Achievements and Future Challenges. Antioxidants (Basel) 2017; 6:antiox6020026. [PMID: 28379200 PMCID: PMC5488006 DOI: 10.3390/antiox6020026] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/01/2017] [Accepted: 04/03/2017] [Indexed: 12/13/2022] Open
Abstract
Green tea catechins (GTCs) are a family of chemically related compounds usually classified as antioxidant molecules. Epidemiological evidences, supported by interventional studies, highlighted a more than promising role for GTCs in human prostate cancer (PCa) chemoprevention. In the last decades, many efforts have been made to gain new insights into the mechanism of action of GTCs. Now it is clear that GTCs' anticancer action can no longer be simplistically limited to their direct antioxidant/pro-oxidant properties. Recent contributions to the advancement of knowledge in this field have shown that GTCs specifically interact with cellular targets, including cell surface receptors, lipid rafts, and endoplasmic reticulum, modulate gene expression through direct effect on transcription factors or indirect epigenetic mechanisms, and interfere with intracellular proteostasis at various levels. Many of the effects observed in vitro are dose and cell context dependent and take place at concentrations that cannot be achieved in vivo. Poor intestinal absorption together with an extensive systemic and enteric metabolism influence GTCs' bioavailability through still poorly understood mechanisms. Recent efforts to develop delivery systems that increase GTCs' overall bioavailability, by means of biopolymeric nanoparticles, represent the main way to translate preclinical results in a real clinical scenario for PCa chemoprevention.
Collapse
Affiliation(s)
- Valeria Naponelli
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma 43126, Italy.
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy.
- National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, Rome 00136, Italy.
- Fondazione Umberto Veronesi, Piazza Velasca 5, Milan 20122, Italy.
| | - Ileana Ramazzina
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma 43126, Italy.
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy.
- National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, Rome 00136, Italy.
| | - Chiara Lenzi
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma 43126, Italy.
| | - Saverio Bettuzzi
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma 43126, Italy.
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy.
- National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, Rome 00136, Italy.
| | - Federica Rizzi
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, Parma 43126, Italy.
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy.
- National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, Rome 00136, Italy.
| |
Collapse
|