151
|
Hönigova K, Navratil J, Peltanova B, Polanska HH, Raudenska M, Masarik M. Metabolic tricks of cancer cells. Biochim Biophys Acta Rev Cancer 2022; 1877:188705. [PMID: 35276232 DOI: 10.1016/j.bbcan.2022.188705] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/11/2022] [Accepted: 02/26/2022] [Indexed: 12/15/2022]
Abstract
One of the characteristics of cancer cells important for tumorigenesis is their metabolic plasticity. Indeed, in various stress conditions, cancer cells can reshape their metabolic pathways to support the increased energy request due to continuous growth and rapid proliferation. Moreover, selective pressures in the tumor microenvironment, such as hypoxia, acidosis, and competition for resources, force cancer cells to adapt by complete reorganization of their metabolism. In this review, we highlight the characteristics of cancer metabolism and discuss its clinical significance, since overcoming metabolic plasticity of cancer cells is a key objective of modern cancer therapeutics and a better understanding of metabolic reprogramming may lead to the identification of possible targets for cancer therapy.
Collapse
Affiliation(s)
- Katerina Hönigova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Jiri Navratil
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Barbora Peltanova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Hana Holcova Polanska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Martina Raudenska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, CZ-252 50 Vestec, Czech Republic.
| |
Collapse
|
152
|
Simões RV, Henriques RN, Cardoso BM, Fernandes FF, Carvalho T, Shemesh N. Glucose fluxes in glycolytic and oxidative pathways detected in vivo by deuterium magnetic resonance spectroscopy reflect proliferation in mouse glioblastoma. Neuroimage Clin 2022; 33:102932. [PMID: 35026626 PMCID: PMC8760481 DOI: 10.1016/j.nicl.2021.102932] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/23/2022]
Abstract
We performed dynamic glucose enhanced (DGE) 2H-MRS in mouse GBM tumors. Marchenko-Pastur PCA denoising of 2H-MRS spectra improved kinetic quantification. Metabolic kinetics revealed differential glucose pathway fluxes in non-necrotic tumors. Modulation of glucose metabolism reflected tumor heterogeneity (proliferation).
Objectives Glioblastoma multiforme (GBM), the most aggressive glial brain tumors, can metabolize glucose through glycolysis and mitochondrial oxidation pathways. While specific dependencies on those pathways are increasingly associated with treatment response, detecting such GBM subtypes in vivo remains elusive. Here, we develop a dynamic glucose-enhanced deuterium spectroscopy (DGE 2H-MRS) approach for differentially assessing glucose turnover rates through glycolysis and mitochondrial oxidation in mouse GBM and explore their association with histologic features of the tumor and its microenvironment. Materials and methods GL261 and CT2A glioma allografts were induced in immunocompetent mice and scanned in vivo at 9.4 Tesla, harnessing DGE 2H-MRS with volume selection and Marchenko-Pastur PCA (MP-PCA) denoising to achieve high temporal resolution. Each tumor was also classified by histopathologic analysis and assessed for cell proliferation (Ki67 immunostaining), while the respective cell lines underwent in situ extracellular flux analysis to assess mitochondrial function. Results MP-PCA denoising of in vivo DGE 2H-MRS data significantly improved the time-course detection (~2-fold increased Signal-to-Noise Ratio) and fitting precision (−19 ± 1 % Cramér-Rao Lower Bounds) of 2H-labelled glucose, and glucose-derived glutamate-glutamine (Glx) and lactate pools in GL261 and CT2A orthotopic tumors. Kinetic modeling further indicated inter-tumor heterogeneity of glucose consumption rate for glycolysis and oxidation during a defined epoch of active proliferation in both cohorts (19 ± 1 days post-induction), with consistent volumes (38.3 ± 3.4 mm3) and perfusion properties prior to marked necrosis. Histopathologic analysis of these tumors revealed clear differences in tumor heterogeneity between the two GBM models, aligned with metabolic differences of the respective cell lines monitored in situ. Importantly, glucose oxidation (i.e. Glx synthesis and elimination rates: 0.40 ± 0.08 and 0.12 ± 0.03 mM min−1, respectively) strongly correlated with cell proliferation across the pooled cohorts (R = 0.82, p = 0.001; and R = 0.80, p = 0.002, respectively), regardless of tumor morphologic features or in situ metabolic characteristics of each GBM model. Conclusions Our fast DGE 2H-MRS enables the quantification of glucose consumption rates through glycolysis and mitochondrial oxidation in mouse GBM, which is relevant for assessing their modulation in vivo according to tumor microenvironment features such as cell proliferation. This novel application augurs well for non-invasive metabolic characterization of glioma or other cancers with mitochondrial oxidation dependencies.
Collapse
Affiliation(s)
- Rui V Simões
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| | - Rafael N Henriques
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Beatriz M Cardoso
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Tânia Carvalho
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
153
|
Kong F, Fang C, Zhang Y, Duan L, Du D, Xu G, Li X, Li H, Yin Y, Xu H, Zhang K. Abundance and Metabolism Disruptions of Intratumoral Microbiota by Chemical and Physical Actions Unfreeze Tumor Treatment Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105523. [PMID: 35037431 PMCID: PMC8895135 DOI: 10.1002/advs.202105523] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/17/2021] [Indexed: 12/19/2022]
Abstract
Intratumoral or intestinal microbiota correlates with tumorigenesis and progression, and microbiota regulation for reinforcing various anti-tumor approaches is of significant importance, which, however, suffers from no precise regulation method and unclear underlying mechanism. Herein, a microbiome metabolism-engineered phototherapy strategy is established, wherein Nb2 C/Au nanocomposite and the corresponding phototherapy are harnessed to realize "chemical" and "physical" bacterial regulations. Flora analysis and mass spectrometry (MS) and metabonomics combined tests demonstrate that the synergistic microbiota regulations can alter the abundance, diversity of intratumoral microbiome, and disrupt metabolic pathways of microbiome and tumor microenvironment, wherein the differential singling pathways and biosynthetic necessities or metabolites that can affect tumor progression are identified. As well, anti-TNFα is introduced to unite with bacterial regulation to synergistically mitigate bacterial-induced inflammation, which, along with the metabolism disruptions of intratumoral microbiota and tumor microenvironment, unfreezes tumor resistance and harvests significantly-intensified phototherapy-based anti-tumor outcomes against 4T1 and CT26 tumors. The clear underlying principles of microbiome-regulated tumorigenesis and the established microbiome metabolism regulation method provide distinctive insights into tumor therapy, and can be also extended to other gut microbiome-associated lesions interference.
Collapse
Affiliation(s)
- Fanlei Kong
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
- Department of Medical UltrasoundAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineNo. 261 Huansha RoadHangzhou310006P. R. China
| | - Chao Fang
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
- Department of Medical Ultrasound and Department of RadiologyGuangxi Medical University Cancer Hospitaland Guangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNo. 71 Hedi RoadNanning530021P. R. China
| | - Yan Zhang
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Lixia Duan
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
- Department of Medical Ultrasound and Department of RadiologyGuangxi Medical University Cancer Hospitaland Guangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNo. 71 Hedi RoadNanning530021P. R. China
| | - Dou Du
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Guang Xu
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Xiaolong Li
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Hongyan Li
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Yifei Yin
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Huixiong Xu
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Kun Zhang
- Department of Medical Ultrasound and Central LaboratoryUltrasound Research and Education InstituteShanghai Tenth People's HospitalTongji University School of MedicineNo. 301 Yan‐chang‐zhong RoadShanghai200072P. R. China
- Department of Medical Ultrasound and Department of RadiologyGuangxi Medical University Cancer Hospitaland Guangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNo. 71 Hedi RoadNanning530021P. R. China
| |
Collapse
|
154
|
Fidelito G, Watt MJ, Taylor RA. Personalized Medicine for Prostate Cancer: Is Targeting Metabolism a Reality? Front Oncol 2022; 11:778761. [PMID: 35127483 PMCID: PMC8813754 DOI: 10.3389/fonc.2021.778761] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer invokes major shifts in gene transcription and metabolic signaling to mediate alterations in nutrient acquisition and metabolic substrate selection when compared to normal tissues. Exploiting such metabolic reprogramming is proposed to enable the development of targeted therapies for prostate cancer, yet there are several challenges to overcome before this becomes a reality. Herein, we outline the role of several nutrients known to contribute to prostate tumorigenesis, including fatty acids, glucose, lactate and glutamine, and discuss the major factors contributing to variability in prostate cancer metabolism, including cellular heterogeneity, genetic drivers and mutations, as well as complexity in the tumor microenvironment. The review draws from original studies employing immortalized prostate cancer cells, as well as more complex experimental models, including animals and humans, that more accurately reflect the complexity of the in vivo tumor microenvironment. In synthesizing this information, we consider the feasibility and potential limitations of implementing metabolic therapies for prostate cancer management.
Collapse
Affiliation(s)
- Gio Fidelito
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Matthew J. Watt
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Renea A. Taylor, ; Matthew J. Watt,
| | - Renea A. Taylor
- Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, VIC, Australia
- Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Renea A. Taylor, ; Matthew J. Watt,
| |
Collapse
|
155
|
Raftopulos NL, Washaya TC, Niederprüm A, Egert A, Hakeem-Sanni MF, Varney B, Aishah A, Georgieva ML, Olsson E, Dos Santos DZ, Nassar ZD, Cochran BJ, Nagarajan SR, Kakani MS, Hastings JF, Croucher DR, Rye KA, Butler LM, Grewal T, Hoy AJ. Prostate cancer cell proliferation is influenced by LDL-cholesterol availability and cholesteryl ester turnover. Cancer Metab 2022; 10:1. [PMID: 35033184 PMCID: PMC8760736 DOI: 10.1186/s40170-021-00278-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/24/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Prostate cancer growth is driven by androgen receptor signaling, and advanced disease is initially treatable by depleting circulating androgens. However, prostate cancer cells inevitably adapt, resulting in disease relapse with incurable castrate-resistant prostate cancer. Androgen deprivation therapy has many side effects, including hypercholesterolemia, and more aggressive and castrate-resistant prostate cancers typically feature cellular accumulation of cholesterol stored in the form of cholesteryl esters. As cholesterol is a key substrate for de novo steroidogenesis in prostate cells, this study hypothesized that castrate-resistant/advanced prostate cancer cell growth is influenced by the availability of extracellular, low-density lipoprotein (LDL)-derived, cholesterol, which is coupled to intracellular cholesteryl ester homeostasis. METHODS C4-2B and PC3 prostate cancer cells were cultured in media supplemented with fetal calf serum (FCS), charcoal-stripped FCS (CS-FCS), lipoprotein-deficient FCS (LPDS), or charcoal-stripped LPDS (CS-LPDS) and analyzed by a variety of biochemical techniques. Cell viability and proliferation were measured by MTT assay and Incucyte, respectively. RESULTS Reducing lipoprotein availability led to a reduction in cholesteryl ester levels and cell growth in C4-2B and PC3 cells, with concomitant reductions in PI3K/mTOR and p38MAPK signaling. This reduced growth in LPDS-containing media was fully recovered by supplementation of exogenous low-density lipoprotein (LDL), but LDL only partially rescued growth of cells cultured with CS-LPDS. This growth pattern was not associated with changes in androgen receptor signaling but rather increased p38MAPK and MEK1/ERK/MSK1 activation. The ability of LDL supplementation to rescue cell growth required cholesterol esterification as well as cholesteryl ester hydrolysis activity. Further, growth of cells cultured in low androgen levels (CS-FCS) was suppressed when cholesteryl ester hydrolysis was inhibited. CONCLUSIONS Overall, these studies demonstrate that androgen-independent prostate cancer cell growth can be influenced by extracellular lipid levels and LDL-cholesterol availability and that uptake of extracellular cholesterol, through endocytosis of LDL-derived cholesterol and subsequent delivery and storage in the lipid droplet as cholesteryl esters, is required to support prostate cancer cell growth. This provides new insights into the relationship between extracellular cholesterol, intracellular cholesterol metabolism, and prostate cancer cell growth and the potential mechanisms linking hypercholesterolemia and more aggressive prostate cancer.
Collapse
Affiliation(s)
- Nikki L Raftopulos
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Tinashe C Washaya
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Andreas Niederprüm
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Ruprecht Karl University of Heidelberg, Baden-Wuerttemberg, Heidelberg, Germany
| | - Antonia Egert
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Mariam F Hakeem-Sanni
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Bianca Varney
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Atqiya Aishah
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Mariya L Georgieva
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ellinor Olsson
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Diandra Z Dos Santos
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Zeyad D Nassar
- Adelaide Medical School and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Blake J Cochran
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Shilpa R Nagarajan
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Meghna S Kakani
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jordan F Hastings
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - David R Croucher
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Hospital Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Kerry-Anne Rye
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
156
|
Smalling RV, Bechard ME, Duryea J, Kingsley PJ, Roberts ER, Marnett LJ, Bilbao D, Stauffer SR, McDonald OG. Aminopyridine analogs selectively target metastatic pancreatic cancer. Oncogene 2022; 41:1518-1525. [PMID: 35031771 DOI: 10.1038/s41388-022-02183-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/17/2021] [Accepted: 01/07/2022] [Indexed: 12/20/2022]
Abstract
Metastatic outgrowth is supported by metabolic adaptations that may differ from the primary tumor of origin. However, it is unknown if such adaptations are therapeutically actionable. Here we report a novel aminopyridine compound that targets a unique Phosphogluconate Dehydrogenase (PGD)-dependent metabolic adaptation in distant metastases from pancreatic cancer patients. Compared to structurally similar analogs, 6-aminopicolamine (6AP) potently and selectively reversed PGD-dependent metastatic properties, including intrinsic tumorigenic capacity, excess glucose consumption, and global histone hyperacetylation. 6AP acted as a water-soluble prodrug that was converted into intracellular bioactive metabolites that inhibited PGD in vitro, and 6AP monotherapy demonstrated anti-metastatic efficacy with minimal toxicity in vivo. Collectively, these studies identify 6AP and possibly other 6-aminopyridines as well-tolerated prodrugs with selectivity for metastatic pancreatic cancers. If unique metabolic adaptations are a common feature of metastatic or otherwise aggressive human malignancies, then such dependencies could provide a largely untapped pool of druggable targets for patients with advanced cancers.
Collapse
Affiliation(s)
- Rana V Smalling
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew E Bechard
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeff Duryea
- Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Philip J Kingsley
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Evan R Roberts
- Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lawrence J Marnett
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Daniel Bilbao
- Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shaun R Stauffer
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Therapeutics Discovery, Cleveland Clinic, Cleveland, OH, USA
| | - Oliver G McDonald
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA. .,Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
157
|
Gonçalves AC, Richiardone E, Jorge J, Polónia B, Xavier CPR, Salaroglio IC, Riganti C, Vasconcelos MH, Corbet C, Sarmento-Ribeiro AB. Impact of cancer metabolism on therapy resistance - Clinical implications. Drug Resist Updat 2021; 59:100797. [PMID: 34955385 DOI: 10.1016/j.drup.2021.100797] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite an increasing arsenal of anticancer therapies, many patients continue to have poor outcomes due to the therapeutic failures and tumor relapses. Indeed, the clinical efficacy of anticancer therapies is markedly limited by intrinsic and/or acquired resistance mechanisms that can occur in any tumor type and with any treatment. Thus, there is an urgent clinical need to implement fundamental changes in the tumor treatment paradigm by the development of new experimental strategies that can help to predict the occurrence of clinical drug resistance and to identify alternative therapeutic options. Apart from mutation-driven resistance mechanisms, tumor microenvironment (TME) conditions generate an intratumoral phenotypic heterogeneity that supports disease progression and dismal outcomes. Tumor cell metabolism is a prototypical example of dynamic, heterogeneous, and adaptive phenotypic trait, resulting from the combination of intrinsic [(epi)genetic changes, tissue of origin and differentiation dependency] and extrinsic (oxygen and nutrient availability, metabolic interactions within the TME) factors, enabling cancer cells to survive, metastasize and develop resistance to anticancer therapies. In this review, we summarize the current knowledge regarding metabolism-based mechanisms conferring adaptive resistance to chemo-, radio-and immunotherapies as well as targeted therapies. Furthermore, we report the role of TME-mediated intratumoral metabolic heterogeneity in therapy resistance and how adaptations in amino acid, glucose, and lipid metabolism support the growth of therapy-resistant cancers and/or cellular subpopulations. We also report the intricate interplay between tumor signaling and metabolic pathways in cancer cells and discuss how manipulating key metabolic enzymes and/or providing dietary changes may help to eradicate relapse-sustaining cancer cells. Finally, in the current era of personalized medicine, we describe the strategies that may be applied to implement metabolic profiling for tumor imaging, biomarker identification, selection of tailored treatments and monitoring therapy response during the clinical management of cancer patients.
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| | - Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Bárbara Polónia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | | | - Chiara Riganti
- Department of Oncology, School of Medicine, University of Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium.
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| |
Collapse
|
158
|
Hoy AJ, Nagarajan SR, Butler LM. Tumour fatty acid metabolism in the context of therapy resistance and obesity. Nat Rev Cancer 2021; 21:753-766. [PMID: 34417571 DOI: 10.1038/s41568-021-00388-4] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
Fatty acid metabolism is known to support tumorigenesis and disease progression as well as treatment resistance through enhanced lipid synthesis, storage and catabolism. More recently, the role of membrane fatty acid composition, for example, ratios of saturated, monounsaturated and polyunsaturated fatty acids, in promoting cell survival while limiting lipotoxicity and ferroptosis has been increasingly appreciated. Alongside these insights, it has become clear that tumour cells exhibit plasticity with respect to fatty acid metabolism, responding to extratumoural and systemic metabolic signals, such as obesity and cancer therapeutics, to promote the development of aggressive, treatment-resistant disease. Here, we describe cellular fatty acid metabolic changes that are connected to therapy resistance and contextualize obesity-associated changes in host fatty acid metabolism that likely influence the local tumour microenvironment to further modify cancer cell behaviour while simultaneously creating potential new vulnerabilities.
Collapse
Affiliation(s)
- Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| | - Shilpa R Nagarajan
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
159
|
Metabolic Plasticity in Melanoma Progression and Response to Oncogene Targeted Therapies. Cancers (Basel) 2021; 13:cancers13225810. [PMID: 34830962 PMCID: PMC8616485 DOI: 10.3390/cancers13225810] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Targeted anti-cancer therapies have revolutionised melanoma patient care; however, cures remain uncommon due to acquired drug resistance that results in disease relapse. Recent insights from the clinic and experimental settings have identified a key role for metabolic plasticity, defined as the flexibility to utilise different nutrients and process them in different ways, in both disease progression and response to targeted therapies. Here, we discuss how this plasticity creates a moving target with important implications for identifying new combination therapies. Abstract Resistance to therapy continues to be a barrier to curative treatments in melanoma. Recent insights from the clinic and experimental settings have highlighted a range of non-genetic adaptive mechanisms that contribute to therapy resistance and disease relapse, including transcriptional, post-transcriptional and metabolic reprogramming. A growing body of evidence highlights the inherent plasticity of melanoma metabolism, evidenced by reversible metabolome alterations and flexibility in fuel usage that occur during metastasis and response to anti-cancer therapies. Here, we discuss how the inherent metabolic plasticity of melanoma cells facilitates both disease progression and acquisition of anti-cancer therapy resistance. In particular, we discuss in detail the different metabolic changes that occur during the three major phases of the targeted therapy response—the early response, drug tolerance and acquired resistance. We also discuss how non-genetic programs, including transcription and translation, control this process. The prevalence and diverse array of these non-genetic resistance mechanisms poses a new challenge to the field that requires innovative strategies to monitor and counteract these adaptive processes in the quest to prevent therapy resistance.
Collapse
|
160
|
Choudhury FK. Mitochondrial Redox Metabolism: The Epicenter of Metabolism during Cancer Progression. Antioxidants (Basel) 2021; 10:antiox10111838. [PMID: 34829708 PMCID: PMC8615124 DOI: 10.3390/antiox10111838] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial redox metabolism is the central component in the cellular metabolic landscape, where anabolic and catabolic pathways are reprogrammed to maintain optimum redox homeostasis. During different stages of cancer, the mitochondrial redox status plays an active role in navigating cancer cells’ progression and regulating metabolic adaptation according to the constraints of each stage. Mitochondrial reactive oxygen species (ROS) accumulation induces malignant transformation. Once vigorous cell proliferation renders the core of the solid tumor hypoxic, the mitochondrial electron transport chain mediates ROS signaling for bringing about cellular adaptation to hypoxia. Highly aggressive cells are selected in this process, which are capable of progressing through the enhanced oxidative stress encountered during different stages of metastasis for distant colonization. Mitochondrial oxidative metabolism is suppressed to lower ROS generation, and the overall cellular metabolism is reprogrammed to maintain the optimum NADPH level in the mitochondria required for redox homeostasis. After reaching the distant organ, the intrinsic metabolic limitations of that organ dictate the success of colonization and flexibility of the mitochondrial metabolism of cancer cells plays a pivotal role in their adaptation to the new environment.
Collapse
Affiliation(s)
- Feroza K Choudhury
- Drug Metabolism and Pharmacokinetics Department, Genentech Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
161
|
The Long Non-Coding RNA SAMMSON Is a Regulator of Chemosensitivity and Metabolic Orientation in MCF-7 Doxorubicin-Resistant Breast Cancer Cells. BIOLOGY 2021; 10:biology10111156. [PMID: 34827149 PMCID: PMC8615054 DOI: 10.3390/biology10111156] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 12/25/2022]
Abstract
Simple Summary Breast cancer is the most common cancer in women, representing about one third of cancers in developed countries. Despite recent advances in diagnostic methods and increasingly early detection, breast cancer recurrence occurs in more than 20% of patients. Chemoresistance represents an important cause of this recurrence, but the mechanisms involved in this phenomenon, are still largely unknown. One feature of chemoresistant cancer cells is the reorientation of the energetic metabolism to sustain cell proliferation. Recently, long non-coding RNAs (lncRNAs) have emerged as important regulators of cellular metabolic orientation. In the present work, we gave special attention to the long non-coding RNA SAMMSON and addressed the role of this lncRNA in metabolic orientation and chemoresistance of doxorubicin-resistant breast cancer cells. The results shed light on the possible modulation of the SAMMSON expression as an innovative therapeutic approach to target chemoresistant cancer cells specifically. Abstract Despite improvements in therapeutic strategies for treating breast cancers, tumor relapse and chemoresistance remain major issues in patient outcomes. Indeed, cancer cells display a metabolic plasticity allowing a quick adaptation to the tumoral microenvironment and to cellular stresses induced by chemotherapy. Recently, long non-coding RNA molecules (lncRNAs) have emerged as important regulators of cellular metabolic orientation. In the present study, we addressed the role of the long non-coding RNA molecule (lncRNA) SAMMSON on the metabolic reprogramming and chemoresistance of MCF-7 breast cancer cells resistant to doxorubicin (MCF-7dox). Our results showed an overexpression of SAMMSON in MCF-7dox compared to doxorubicin-sensitive cells (MCF-7). Silencing of SAMMSON expression by siRNA in MCF-7dox cells resulted in a metabolic rewiring with improvement of oxidative metabolism, decreased mitochondrial ROS production, increased mitochondrial replication, transcription and translation and an attenuation of chemoresistance. These results highlight the role of SAMMSON in the metabolic adaptations leading to the development of chemoresistance in breast cancer cells. Thus, targeting SAMMSON expression levels represents a promising therapeutic route to circumvent doxorubicin resistance in breast cancers.
Collapse
|
162
|
Iorio M, Umesh Ganesh N, De Luise M, Porcelli AM, Gasparre G, Kurelac I. The Neglected Liaison: Targeting Cancer Cell Metabolic Reprogramming Modifies the Composition of Non-Malignant Populations of the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13215447. [PMID: 34771610 PMCID: PMC8582418 DOI: 10.3390/cancers13215447] [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: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Metabolic reprogramming is a well-known hallmark of cancer, whereby the development of drugs that target cancer cell metabolism is gaining momentum. However, when establishing preclinical studies and clinical trials, it is often neglected that a tumor mass is a complex system in which cancer cells coexist and interact with several types of microenvironment populations, including endothelial cells, fibroblasts and immune cells. We are just starting to understand how such populations are affected by the metabolic changes occurring in a transformed cell and little is known about the impact of metabolism-targeting drugs on the non-malignant tumor components. Here we provide a general overview of the links between cancer cell metabolism and tumor microenvironment (TME), particularly focusing on the emerging literature reporting TME-specific effects of metabolic therapies.
Collapse
Affiliation(s)
- Maria Iorio
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Nikkitha Umesh Ganesh
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Monica De Luise
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Anna Maria Porcelli
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
- Interdepartmental Center of Industrial Research (CIRI) Life Science and Health Technologies, University of Bologna, 40064 Ozzano dell’Emilia, Italy
| | - Giuseppe Gasparre
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Ivana Kurelac
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (M.I.); (N.U.G.); (M.D.L.); (G.G.)
- Center for Applied Biomedical Research, University of Bologna, 40138 Bologna, Italy;
- Centro Studi e Ricerca sulle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
- Correspondence: ; Tel.: +39-051-2088-418
| |
Collapse
|
163
|
Sulforaphane Causes Cell Cycle Arrest and Apoptosis in Human Glioblastoma U87MG and U373MG Cell Lines under Hypoxic Conditions. Int J Mol Sci 2021; 22:ijms222011201. [PMID: 34681862 PMCID: PMC8541491 DOI: 10.3390/ijms222011201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor. The median survival rate from diagnosis ranges from 15 to 17 months because the tumor is resistant to most therapeutic strategies. GBM exhibits microvascular hyperplasia and pronounced necrosis triggered by hypoxia. Sulforaphane (SFN), an isothiocyanate derived from cruciferous vegetables, has already demonstrated the ability to inhibit cell proliferation, by provoking cell cycle arrest, and leading to apoptosis in many cell lines. In this study, we investigated the antineoplastic effects of SFN [20-80 μM for 48 h] in GBM cells under normoxic and hypoxic conditions. Cell viability assays, flow cytometry, and Western blot results revealed that SFN could induce apoptosis of GBM cells in a dose-dependent manner, under both conditions. In particular, SFN significantly induced caspase 3/7 activation and DNA fragmentation. Moreover, our results demonstrated that SFN suppressed GBM cells proliferation by arresting the cell cycle at the S-phase, also under hypoxic condition, and that these effects may be due in part to its ability to induce oxidative stress by reducing glutathione levels and to increase the phosphorylation of extracellular signal-regulated kinases (ERKs). Overall, we hypothesized that SFN treatment might serve as a potential therapeutic strategy, alone or in combination, against GBM.
Collapse
|
164
|
Cacciola NA, Sgadari M, Sepe F, Petillo O, Margarucci S, Martano M, Maiolino P, Restucci B. Metabolic Flexibility in Canine Mammary Tumors: Implications of the Carnitine System. Animals (Basel) 2021; 11:ani11102969. [PMID: 34679988 PMCID: PMC8532965 DOI: 10.3390/ani11102969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Deregulation of fatty acid catabolism provides an alternative energy source to glycolysis for cancer cell survival and proliferation. The regulator enzymes of the carnitine system (CS), responsible for the transport of fatty acids across mitochondrial membranes for β-oxidation are deregulated in tumorigenesis. Recently, we found that Carnitine Palmitoyl Transferase 1 (CPT1), a crucial regulator of CS components, is expressed and dysregulated in canine mammary tumor (CMT) tissues and cells. In this study, we examined the protein expression of the three remaining enzymes of CS (Carnitine Acylcarnitine Translocase (CACT), Carnitine Palmitoyl Transferase 2 (CPT2), Carnitine O-acetyltransferase (CrAT), in canine mammary cells and tissues by Western blot and immunohistochemistry. Protein expression of the components of CS was found in normal mammary glands and a concomitant deregulation of expression in CMT tissues that inversely correlated with the degree of tumor differentiation. Moreover, the expression and a different deregulation of CS-related proteins was also observed in CF33, CMT-U27, CMT-U309, and P114 cell lines used as in vitro model. These results demonstrate for the first time the expression of CS components in CMT tissues and cancer cells; however, further studies are needed to elucidate their roles in dogs as well.
Collapse
Affiliation(s)
- Nunzio Antonio Cacciola
- Department of Veterinary Medicine and Animal Production, University of Naples “Federico II”, Via F. Delpino, 1, 80138 Naples, Italy; (M.S.); (F.S.); (M.M.); (P.M.); (B.R.)
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council (CNR), Via P. Castellino, 111, 80131 Naples, Italy; (O.P.); (S.M.)
- Correspondence: ; Tel.: +39-08-1613-2282
| | - Mariafrancesca Sgadari
- Department of Veterinary Medicine and Animal Production, University of Naples “Federico II”, Via F. Delpino, 1, 80138 Naples, Italy; (M.S.); (F.S.); (M.M.); (P.M.); (B.R.)
| | - Fabrizia Sepe
- Department of Veterinary Medicine and Animal Production, University of Naples “Federico II”, Via F. Delpino, 1, 80138 Naples, Italy; (M.S.); (F.S.); (M.M.); (P.M.); (B.R.)
| | - Orsolina Petillo
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council (CNR), Via P. Castellino, 111, 80131 Naples, Italy; (O.P.); (S.M.)
| | - Sabrina Margarucci
- Research Institute on Terrestrial Ecosystems (IRET), National Research Council (CNR), Via P. Castellino, 111, 80131 Naples, Italy; (O.P.); (S.M.)
| | - Manuela Martano
- Department of Veterinary Medicine and Animal Production, University of Naples “Federico II”, Via F. Delpino, 1, 80138 Naples, Italy; (M.S.); (F.S.); (M.M.); (P.M.); (B.R.)
| | - Paola Maiolino
- Department of Veterinary Medicine and Animal Production, University of Naples “Federico II”, Via F. Delpino, 1, 80138 Naples, Italy; (M.S.); (F.S.); (M.M.); (P.M.); (B.R.)
| | - Brunella Restucci
- Department of Veterinary Medicine and Animal Production, University of Naples “Federico II”, Via F. Delpino, 1, 80138 Naples, Italy; (M.S.); (F.S.); (M.M.); (P.M.); (B.R.)
| |
Collapse
|
165
|
Wang X, Yin G, Zhang W, Song K, Zhang L, Guo Z. Prostaglandin Reductase 1 as a Potential Therapeutic Target for Cancer Therapy. Front Pharmacol 2021; 12:717730. [PMID: 34421612 PMCID: PMC8377670 DOI: 10.3389/fphar.2021.717730] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Altered tumor metabolism is a hallmark of cancer and targeting tumor metabolism has been considered as an attractive strategy for cancer therapy. Prostaglandin Reductase 1 (PTGR1) is a rate-limiting enzyme involved in the arachidonic acid metabolism pathway and mainly responsible for the deactivation of some eicosanoids, including prostaglandins and leukotriene B4. A growing evidence suggested that PTGR1 plays a significant role in cancer and has emerged as a novel target for cancer therapeutics. In this review, we summarize the progress made in recent years toward the understanding of PTGR1 function and structure, highlight the roles of PTGR1 in cancer, and describe potential inhibitors of PTGR1. Finally, we provide some thoughts on future directions that might facilitate the PTGR1 research and therapeutics development.
Collapse
Affiliation(s)
- Xing Wang
- Department of Breast and Thyroid Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Guobing Yin
- Department of Breast and Thyroid Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Zhang
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Kunlin Song
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Longbin Zhang
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Zufeng Guo
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
166
|
Khan A, Siddiqui S, Husain SA, Mazurek S, Iqbal MA. Phytocompounds Targeting Metabolic Reprogramming in Cancer: An Assessment of Role, Mechanisms, Pathways, and Therapeutic Relevance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:6897-6928. [PMID: 34133161 DOI: 10.1021/acs.jafc.1c01173] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The metabolism of cancer is remarkably different from that of normal cells and confers a variety of benefits, including the promotion of other cancer hallmarks. As the rewired metabolism is a near-universal property of cancer cells, efforts are underway to exploit metabolic vulnerabilities for therapeutic benefits. In the continued search for safer and effective ways of cancer treatment, structurally diverse plant-based compounds have gained substantial attention. Here, we present an extensive assessment of the role of phytocompounds in modulating cancer metabolism and attempt to make a case for the use of plant-based compounds in targeting metabolic vulnerabilities of cancer. We discuss the pharmacological interactions of phytocompounds with major metabolic pathways and evaluate the role of phytocompounds in the regulation of growth signaling and transcriptional programs involved in the metabolic transformation of cancer. Lastly, we examine the potential of these compounds in the clinical management of cancer along with limitations and challenges.
Collapse
Affiliation(s)
- Asifa Khan
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Shumaila Siddiqui
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Syed Akhtar Husain
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Sybille Mazurek
- Institute of Veterinary-Physiology and Biochemistry, University of Giessen, Giessen 35392, Germany
| | - Mohammad Askandar Iqbal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| |
Collapse
|
167
|
Life after Cell Death-Survival and Survivorship Following Chemotherapy. Cancers (Basel) 2021; 13:cancers13122942. [PMID: 34208331 PMCID: PMC8231100 DOI: 10.3390/cancers13122942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Treatment of aggressive cancers often relies on chemotherapy. This treatment has improved survival rates, but while effective at killing cancer cells, inevitably it also kills or alters the function of others. While many of the known effects are transient and resolve after treatment, as survival rates increase, so does our understanding of the long-term health costs that accompany cancer survivors. Here we provide an overview of common long-term morbidities known to be caused by conventional chemotherapy, including the risk of relapse, but more importantly, the cost of quality of life experienced, especially by those who have cancer in early life. We aim to highlight the importance of the development of targeted therapies to replace the use of conventional chemotherapy, but also that of treating the patients along with the disease to enable not only longer but also healthier life after cancer. Abstract To prevent cancer cells replacing and outnumbering their functional somatic counterparts, the most effective solution is their removal. Classical treatments rely on surgical excision, chemical or physical damage to the cancer cells by conventional interventions such as chemo- and radiotherapy, to eliminate or reduce tumour burden. Cancer treatment has in the last two decades seen the advent of increasingly sophisticated therapeutic regimens aimed at selectively targeting cancer cells whilst sparing the remaining cells from severe loss of viability or function. These include small molecule inhibitors, monoclonal antibodies and a myriad of compounds that affect metabolism, angiogenesis or immunotherapy. Our increased knowledge of specific cancer types, stratified diagnoses, genetic and molecular profiling, and more refined treatment practices have improved overall survival in a significant number of patients. Increased survival, however, has also increased the incidence of associated challenges of chemotherapy-induced morbidity, with some pathologies developing several years after termination of treatment. Long-term care of cancer survivors must therefore become a focus in itself, such that along with prolonging life expectancy, treatments allow for improved quality of life.
Collapse
|
168
|
Chen Y, Tian Y, Jin G, Cui Z, Guo W, Zhang X, Liu X. Lobetyolin inhibits the proliferation of breast cancer cells via ASCT2 down-regulation-induced apoptosis. Hum Exp Toxicol 2021; 40:2074-2086. [PMID: 34075790 DOI: 10.1177/09603271211021476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study aimed to investigate the anti-cancer effect of lobetyolin on breast cancer cells. Lobetyolin was incubated with MDA-MB-231 and MDA-MB-468 breast cancer cells for 24 h. Glucose uptake and the mRNA expression of GLUT4 (SLC2A4), HK2 and PKM2 were detected to assess the effect of lobetyolin on glucose metabolism. Glutamine uptake and the mRNA expression of ASCT2 (SLC1A5), GLS1, GDH and GLUL were measured to assess the effect of lobetyolin on glutamine metabolism. Annexin V/PI double staining and Hoechst 33342 staining were used to investigate the effect of lobetyolin on cell apoptosis. Immunoblot was employed to estimate the effect of lobetyolin on the expression of proliferation-related markers and apoptosis-related markers. SLC1A5 knockdown with specific siRNA was performed to study the role of ASCT2 played in the anti-cancer effect of lobetyolin on MDA-MB-231 and MDA-MB-468 breast cancer cells. C-MYC knockdown with specific siRNA was performed to study the role of c-Myc played in lobetyolin-induced ASCT2 down-regulation. Myr-AKT overexpression was performed to investigate the role of AKT/GSK3β signaling played in lobetyolin-induced down-regulation of c-Myc and ASCT2. The results showed that lobetyolin inhibited the proliferation of both MDA-MB-231 and MDA-MB-468 breast cancer cells. Lobetyolin disrupted glutamine uptake via down-regulating ASCT2. SLC1A5 knockdown attenuated the anti-cancer effect of lobetyolin. C-MYC knockdown attenuated lobetyolin-caused down-regulation of ASCT2 and Myr-AKT overexpression reversed lobetyolin-caused down-regulation of both c-Myc and ASCT2. In conclusion, the present work suggested that lobetyolin exerted anti-cancer effect via ASCT2 down-regulation-induced apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Yansong Chen
- Deptment of Radiotherapy & Oncology, 105860The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Oncological Surgery, 74540The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ye Tian
- Deptment of Radiotherapy & Oncology, 105860The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Gongsheng Jin
- Department of Oncological Surgery, 74540The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhen Cui
- Department of Oncological Surgery, 74540The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Wei Guo
- Department of Oncological Surgery, 74540The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaojing Zhang
- Department of Oncological Surgery, 74540The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xianfu Liu
- Department of Oncological Surgery, 74540The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
169
|
Giacomini I, Gianfanti F, Desbats MA, Orso G, Berretta M, Prayer-Galetti T, Ragazzi E, Cocetta V. Cholesterol Metabolic Reprogramming in Cancer and Its Pharmacological Modulation as Therapeutic Strategy. Front Oncol 2021; 11:682911. [PMID: 34109128 PMCID: PMC8181394 DOI: 10.3389/fonc.2021.682911] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Cholesterol is a ubiquitous sterol with many biological functions, which are crucial for proper cellular signaling and physiology. Indeed, cholesterol is essential in maintaining membrane physical properties, while its metabolism is involved in bile acid production and steroid hormone biosynthesis. Additionally, isoprenoids metabolites of the mevalonate pathway support protein-prenylation and dolichol, ubiquinone and the heme a biosynthesis. Cancer cells rely on cholesterol to satisfy their increased nutrient demands and to support their uncontrolled growth, thus promoting tumor development and progression. Indeed, transformed cells reprogram cholesterol metabolism either by increasing its uptake and de novo biosynthesis, or deregulating the efflux. Alternatively, tumor can efficiently accumulate cholesterol into lipid droplets and deeply modify the activity of key cholesterol homeostasis regulators. In light of these considerations, altered pathways of cholesterol metabolism might represent intriguing pharmacological targets for the development of exploitable strategies in the context of cancer therapy. Thus, this work aims to discuss the emerging evidence of in vitro and in vivo studies, as well as clinical trials, on the role of cholesterol pathways in the treatment of cancer, starting from already available cholesterol-lowering drugs (statins or fibrates), and moving towards novel potential pharmacological inhibitors or selective target modulators.
Collapse
Affiliation(s)
- Isabella Giacomini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Federico Gianfanti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, VIMM, Padova, Italy
| | | | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Tommaso Prayer-Galetti
- Department of Surgery, Oncology and Gastroenterology - Urology, University of Padova, Padova, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
170
|
Gorelick AN, Kim M, Chatila WK, La K, Hakimi AA, Berger MF, Taylor BS, Gammage PA, Reznik E. Respiratory complex and tissue lineage drive recurrent mutations in tumour mtDNA. Nat Metab 2021; 3:558-570. [PMID: 33833465 PMCID: PMC9304985 DOI: 10.1038/s42255-021-00378-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/03/2021] [Indexed: 12/21/2022]
Abstract
Mitochondrial DNA (mtDNA) encodes protein subunits and translational machinery required for oxidative phosphorylation (OXPHOS). Using repurposed whole-exome sequencing data, in the present study we demonstrate that pathogenic mtDNA mutations arise in tumours at a rate comparable to those in the most common cancer driver genes. We identify OXPHOS complexes as critical determinants shaping somatic mtDNA mutation patterns across tumour lineages. Loss-of-function mutations accumulate at an elevated rate specifically in complex I and often arise at specific homopolymeric hotspots. In contrast, complex V is depleted of all non-synonymous mutations, suggesting that impairment of ATP synthesis and mitochondrial membrane potential dissipation are under negative selection. Common truncating mutations and rarer missense alleles are both associated with a pan-lineage transcriptional programme, even in cancer types where mtDNA mutations are comparatively rare. Pathogenic mutations of mtDNA are associated with substantial increases in overall survival of colorectal cancer patients, demonstrating a clear functional relationship between genotype and phenotype. The mitochondrial genome is therefore frequently and functionally disrupted across many cancers, with major implications for patient stratification, prognosis and therapeutic development.
Collapse
Affiliation(s)
- Alexander N Gorelick
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Minsoo Kim
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Walid K Chatila
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Konnor La
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York, NY, USA
| | - A Ari Hakimi
- Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael F Berger
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barry S Taylor
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Payam A Gammage
- CRUK Beatson Institute, Glasgow, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | - Ed Reznik
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
171
|
Lu C, Qiao P, Sun Y, Ren C, Yu Z. Positive regulation of PFKFB3 by PIM2 promotes glycolysis and paclitaxel resistance in breast cancer. Clin Transl Med 2021; 11:e400. [PMID: 33931981 PMCID: PMC8087946 DOI: 10.1002/ctm2.400] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is one of the most common female malignancies in the world. Chemotherapeutic resistance is the major cause of BC therapy failure, leading to tumor recurrence and metastasis. Studies have illustrated the close relationship between glycolysis and BC progression and drug resistance. The key glycolysis regulator, PFKFB3 makes a difference during BC progression and drug resistance. However, the mechanism remains to be unknown. METHODS Mass spectrometry analyses were used to found that PIM2 was a potential new binding protein of PFKFB3. Co-immunoprecipitated and western blot were used to verify the interaction between PIM2 and PFKFB3 in BC and the molecular mechanism by which PIM2 phosphorylates PFKFB3 in regulating the protein function. PFKFB3 mutant forms were used to demonstrate the need for PFKFB3 in BC drug resistance. RESULTS We identified that PIM2 is a new binding protein of PFKFB3. We used biochemical methods to determine that PIM2 can directly bind and change the phosphorylation of PFKFB3 at Ser478 to enhance PFKFB3 protein stability through the ubiquitin-proteasome pathway. Importantly, phosphorylation of PFKFB3 at Ser478 promoted glycolysis, BC cell growth, and paclitaxel resistance together with PIM2 in vitro and in vivo. CONCLUSION Our study demonstrates that PIM2 mediates PFKFB3 phosphorylation thus regulates glycolysis and paclitaxel resistance to promote tumor progression in BC and provides preclinical evidence for targeting PFKFB3 as a new strategy in BC treatment to battle paclitaxel resistance.
Collapse
Affiliation(s)
- Chao Lu
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityWeifangShandong ProvinceP. R. China
| | - Pengyun Qiao
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityWeifangShandong ProvinceP. R. China
| | - Yonghong Sun
- Department of PathologyAffiliated Hospital of Weifang Medical UniversityWeifangShandong ProvinceP. R. China
| | - Chune Ren
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityWeifangShandong ProvinceP. R. China
| | - Zhenhai Yu
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityWeifangShandong ProvinceP. R. China
| |
Collapse
|
172
|
Ruiz-Iglesias A, Mañes S. The Importance of Mitochondrial Pyruvate Carrier in Cancer Cell Metabolism and Tumorigenesis. Cancers (Basel) 2021; 13:cancers13071488. [PMID: 33804985 PMCID: PMC8037430 DOI: 10.3390/cancers13071488] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The characteristic metabolic hallmark of cancer cells is the massive catabolism of glucose by glycolysis, even under aerobic conditions—the so-called Warburg effect. Although energetically unfavorable, glycolysis provides “building blocks” to sustain the unlimited growth of malignant cells. Aberrant glycolysis is also responsible for lactate accumulation and acidosis in the tumor milieu, which fosters hypoxia and immunosuppression. One of the mechanisms used by cancer cells to increase glycolytic flow is the negative regulation of the proteins that conform the mitochondrial pyruvate carrier (MPC) complex, which transports pyruvate into the mitochondrial matrix to be metabolized in the tricarboxylic acid (TCA) cycle. Evidence suggests that MPC downregulation in tumor cells impacts many aspects of tumorigenesis, including cancer cell-intrinsic (proliferation, invasiveness, stemness, resistance to therapy) and -extrinsic (angiogenesis, anti-tumor immune activity) properties. In many cancers, but not in all, MPC downregulation is associated with poor survival. MPC regulation is therefore central to tackling glycolysis in tumors. Abstract Pyruvate is a key molecule in the metabolic fate of mammalian cells; it is the crossroads from where metabolism proceeds either oxidatively or ends with the production of lactic acid. Pyruvate metabolism is regulated by many enzymes that together control carbon flux. Mitochondrial pyruvate carrier (MPC) is responsible for importing pyruvate from the cytosol to the mitochondrial matrix, where it is oxidatively phosphorylated to produce adenosine triphosphate (ATP) and to generate intermediates used in multiple biosynthetic pathways. MPC activity has an important role in glucose homeostasis, and its alteration is associated with diabetes, heart failure, and neurodegeneration. In cancer, however, controversy surrounds MPC function. In some cancers, MPC upregulation appears to be associated with a poor prognosis. However, most transformed cells undergo a switch from oxidative to glycolytic metabolism, the so-called Warburg effect, which, amongst other possibilities, is induced by MPC malfunction or downregulation. Consequently, impaired MPC function might induce tumors with strong proliferative, migratory, and invasive capabilities. Moreover, glycolytic cancer cells secrete lactate, acidifying the microenvironment, which in turn induces angiogenesis, immunosuppression, and the expansion of stromal cell populations supporting tumor growth. This review examines the latest findings regarding the tumorigenic processes affected by MPC.
Collapse
|
173
|
A Novel Therapeutic Target, BACH1, Regulates Cancer Metabolism. Cells 2021; 10:cells10030634. [PMID: 33809182 PMCID: PMC8001775 DOI: 10.3390/cells10030634] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
BTB domain and CNC homology 1 (BACH1) is a transcription factor that is highly expressed in tumors including breast and lung, relative to their non-tumor tissues. BACH1 is known to regulate multiple physiological processes including heme homeostasis, oxidative stress response, senescence, cell cycle, and mitosis. In a tumor, BACH1 promotes invasion and metastasis of cancer cells, and the expression of BACH1 presents a poor outcome for cancer patients including breast and lung cancer patients. Recent studies identified novel functional roles of BACH1 in the regulation of metabolic pathways in cancer cells. BACH1 inhibits mitochondrial metabolism through transcriptional suppression of mitochondrial membrane genes. In addition, BACH1 suppresses activity of pyruvate dehydrogenase (PDH), a key enzyme that converts pyruvate to acetyl-CoA for the citric acid (TCA) cycle through transcriptional activation of pyruvate dehydrogenase kinase (PDK). Moreover, BACH1 increases glucose uptake and lactate secretion through the expression of metabolic enzymes involved such as hexokinase 2 (HK2) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) for aerobic glycolysis. Pharmacological or genetic inhibition of BACH1 could reprogram by increasing mitochondrial metabolism, subsequently rendering metabolic vulnerability of cancer cells against mitochondrial respiratory inhibition. Furthermore, inhibition of BACH1 decreased antioxidant-induced glycolysis rates as well as reduced migration and invasion of cancer cells, suggesting BACH1 as a potentially useful cancer therapeutic target.
Collapse
|
174
|
Broadfield LA, Duarte JAG, Schmieder R, Broekaert D, Veys K, Planque M, Vriens K, Karasawa Y, Napolitano F, Fujita S, Fujii M, Eto M, Holvoet B, Vangoitsenhoven R, Fernandez-Garcia J, Van Elsen J, Dehairs J, Zeng J, Dooley J, Rubio RA, van Pelt J, Grünewald TGP, Liston A, Mathieu C, Deroose CM, Swinnen JV, Lambrechts D, di Bernardo D, Kuroda S, De Bock K, Fendt SM. Fat Induces Glucose Metabolism in Nontransformed Liver Cells and Promotes Liver Tumorigenesis. Cancer Res 2021; 81:1988-2001. [PMID: 33687947 DOI: 10.1158/0008-5472.can-20-1954] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/27/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022]
Abstract
Hepatic fat accumulation is associated with diabetes and hepatocellular carcinoma (HCC). Here, we characterize the metabolic response that high-fat availability elicits in livers before disease development. After a short term on a high-fat diet (HFD), otherwise healthy mice showed elevated hepatic glucose uptake and increased glucose contribution to serine and pyruvate carboxylase activity compared with control diet (CD) mice. This glucose phenotype occurred independently from transcriptional or proteomic programming, which identifies increased peroxisomal and lipid metabolism pathways. HFD-fed mice exhibited increased lactate production when challenged with glucose. Consistently, administration of an oral glucose bolus to healthy individuals revealed a correlation between waist circumference and lactate secretion in a human cohort. In vitro, palmitate exposure stimulated production of reactive oxygen species and subsequent glucose uptake and lactate secretion in hepatocytes and liver cancer cells. Furthermore, HFD enhanced the formation of HCC compared with CD in mice exposed to a hepatic carcinogen. Regardless of the dietary background, all murine tumors showed similar alterations in glucose metabolism to those identified in fat exposed nontransformed mouse livers, however, particular lipid species were elevated in HFD tumor and nontumor-bearing HFD liver tissue. These findings suggest that fat can induce glucose-mediated metabolic changes in nontransformed liver cells similar to those found in HCC. SIGNIFICANCE: With obesity-induced hepatocellular carcinoma on a rising trend, this study shows in normal, nontransformed livers that fat induces glucose metabolism similar to an oncogenic transformation.
Collapse
Affiliation(s)
- Lindsay A Broadfield
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - João André Gonçalves Duarte
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Roberta Schmieder
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Dorien Broekaert
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Koen Veys
- Department of Oncology (KU Leuven) and Center for Cancer Biology (VIB), Laboratory of Angiogenesis and Vascular Metabolism, Leuven, Belgium
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Kim Vriens
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Yasuaki Karasawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan.,Department of Neurosurgery, University of Tokyo Hospital, Tokyo, Japan.,Department of Rehabilitation, University of Tokyo Hospital, Tokyo, Japan
| | - Francesco Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics Laboratory and High Content Screening Facility, Naples, Italy
| | - Suguru Fujita
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Masashi Fujii
- Department of Mathematical and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Miki Eto
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Bryan Holvoet
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Belgium
| | | | - Juan Fernandez-Garcia
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Joke Van Elsen
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, Leuven, Belgium
| | - Jia Zeng
- School of Life Science, Hunan University of Science and Technology, Xiangtan, Hunan, China
| | - James Dooley
- Department of Microbiology and Immunology, KU Leuven; and Translational Immunology Laboratory, Leuven, Belgium
| | - Rebeca Alba Rubio
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, Munich, Germany
| | - Jos van Pelt
- Department of Oncology, Laboratory of Clinical Digestive Oncology, KU, Leuven, Belgium
| | - Thomas G P Grünewald
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, Munich, Germany.,Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Adrian Liston
- Department of Microbiology and Immunology, KU Leuven; and Translational Immunology Laboratory, Leuven, Belgium
| | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg KU Leuven, Leuven, Belgium
| | - Christophe M Deroose
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Belgium
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, Leuven, Belgium
| | - Diether Lambrechts
- Department of Human Genetics, Laboratory of Translational Genetics, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine (TIGEM), System Biology and Bioinformatics Laboratory and High Content Screening Facility, Naples, Italy.,Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Shinya Kuroda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Katrien De Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zurich, Zurich, Switzerland
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium. .,Department of Oncology, Laboratory of Cellular Metabolism and Metabolic Regulation, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| |
Collapse
|
175
|
Reimunde P, Pensado-López A, Carreira Crende M, Lombao Iglesias V, Sánchez L, Torrecilla-Parra M, Ramírez CM, Anfray C, Torres Andón F. Cellular and Molecular Mechanisms Underlying Glioblastoma and Zebrafish Models for the Discovery of New Treatments. Cancers (Basel) 2021; 13:1087. [PMID: 33802571 PMCID: PMC7961726 DOI: 10.3390/cancers13051087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common of all brain malignant tumors; it displays a median survival of 14.6 months with current complete standard treatment. High heterogeneity, aggressive and invasive behavior, the impossibility of completing tumor resection, limitations for drug administration and therapeutic resistance to current treatments are the main problems presented by this pathology. In recent years, our knowledge of GBM physiopathology has advanced significantly, generating relevant information on the cellular heterogeneity of GBM tumors, including cancer and immune cells such as macrophages/microglia, genetic, epigenetic and metabolic alterations, comprising changes in miRNA expression. In this scenario, the zebrafish has arisen as a promising animal model to progress further due to its unique characteristics, such as transparency, ease of genetic manipulation, ethical and economic advantages and also conservation of the major brain regions and blood-brain-barrier (BBB) which are similar to a human structure. A few papers described in this review, using genetic and xenotransplantation zebrafish models have been used to study GBM as well as to test the anti-tumoral efficacy of new drugs, their ability to interact with target cells, modulate the tumor microenvironment, cross the BBB and/or their toxicity. Prospective studies following these lines of research may lead to a better diagnosis, prognosis and treatment of patients with GBM.
Collapse
Affiliation(s)
- Pedro Reimunde
- Department of Medicine, Campus de Oza, Universidade da Coruña, 15006 A Coruña, Spain
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Martín Carreira Crende
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Vanesa Lombao Iglesias
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Marta Torrecilla-Parra
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Cristina M. Ramírez
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Clément Anfray
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| |
Collapse
|
176
|
Grasmann G, Mondal A, Leithner K. Flexibility and Adaptation of Cancer Cells in a Heterogenous Metabolic Microenvironment. Int J Mol Sci 2021; 22:1476. [PMID: 33540663 PMCID: PMC7867260 DOI: 10.3390/ijms22031476] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
The metabolic microenvironment, comprising all soluble and insoluble nutrients and co-factors in the extracellular milieu, has a major impact on cancer cell proliferation and survival. A large body of evidence from recent studies suggests that tumor cells show a high degree of metabolic flexibility and adapt to variations in nutrient availability. Insufficient vascular networks and an imbalance of supply and demand shape the metabolic tumor microenvironment, which typically contains a lower concentration of glucose compared to normal tissues. The present review sheds light on the recent literature on adaptive responses in cancer cells to nutrient deprivation. It focuses on the utilization of alternative nutrients in anabolic metabolic pathways in cancer cells, including soluble metabolites and macromolecules and outlines the role of central metabolic enzymes conferring metabolic flexibility, like gluconeogenesis enzymes. Moreover, a conceptual framework for potential therapies targeting metabolically flexible cancer cells is presented.
Collapse
Affiliation(s)
- Gabriele Grasmann
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (G.G.); (A.M.)
| | - Ayusi Mondal
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (G.G.); (A.M.)
| | - Katharina Leithner
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (G.G.); (A.M.)
- BioTechMed-Graz, A-8010 Graz, Austria
| |
Collapse
|