1
|
Higurashi M, Mori K, Nakagawa H, Uchida M, Ishikawa F, Shibanuma M. Respiratory complex I-mediated NAD + regeneration regulates cancer cell proliferation through the transcriptional and translational control of p21 Cip1 expression by SIRT3 and SIRT7. Mol Oncol 2025. [PMID: 39873399 DOI: 10.1002/1878-0261.13808] [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: 05/17/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 01/30/2025] Open
Abstract
The role of the electron transport chain (ETC) in cell proliferation control beyond its crucial function in supporting ATP generation has recently emerged. In this study, we found that, among the four ETC complexes, the complex I (CI)-mediated NAD+ regeneration is important for cancer cell proliferation. In cancer cells, a decrease in CI activity by RNA interference (RNAi) against NADH:ubiquinone oxidoreductase core subunit V1 (NDUFV1) arrested the cell cycle at the G1/S phase, accompanying upregulation of p21Cip1 cyclin-dependent kinase inhibitor expression. Mechanistically, a decrease in the NAD+/NADH ratio downregulated SIRT3 and SIRT7 function, which suppressed p21Cip1 expression at the translational and transcriptional levels, respectively, resulting in the upregulation of the antiproliferative molecule. Importantly, high expression levels of the core subunits of CI correlated with poor prognosis in patients with the hormone receptor(+)/human epidermal growth factor receptor 2(-) (HR+/HER2-) subtype of breast cancer. Therefore, NDUFV1 and SIRT3/7 have emerged as promising therapeutic targets against this breast cancer subtype.
Collapse
Affiliation(s)
- Masato Higurashi
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Kazunori Mori
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Hidetsugu Nakagawa
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Momoko Uchida
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | | | - Motoko Shibanuma
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| |
Collapse
|
2
|
Abukwaik R, Vera-Siguenza E, Tennant D, Spill F. p53 Orchestrates Cancer Metabolism: Unveiling Strategies to Reverse the Warburg Effect. Bull Math Biol 2024; 86:124. [PMID: 39207627 PMCID: PMC11362376 DOI: 10.1007/s11538-024-01346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Cancer cells exhibit significant alterations in their metabolism, characterised by a reduction in oxidative phosphorylation (OXPHOS) and an increased reliance on glycolysis, even in the presence of oxygen. This metabolic shift, known as the Warburg effect, is pivotal in fuelling cancer's uncontrolled growth, invasion, and therapeutic resistance. While dysregulation of many genes contributes to this metabolic shift, the tumour suppressor gene p53 emerges as a master player. Yet, the molecular mechanisms remain elusive. This study introduces a comprehensive mathematical model, integrating essential p53 targets, offering insights into how p53 orchestrates its targets to redirect cancer metabolism towards an OXPHOS-dominant state. Simulation outcomes align closely with experimental data comparing glucose metabolism in colon cancer cells with wild-type and mutated p53. Additionally, our findings reveal the dynamic capability of elevated p53 activation to fully reverse the Warburg effect, highlighting the significance of its activity levels not just in triggering apoptosis (programmed cell death) post-chemotherapy but also in modifying the metabolic pathways implicated in treatment resistance. In scenarios of p53 mutations, our analysis suggests targeting glycolysis-instigating signalling pathways as an alternative strategy, whereas targeting solely synthesis of cytochrome c oxidase 2 (SCO2) does support mitochondrial respiration but may not effectively suppress the glycolysis pathway, potentially boosting the energy production and cancer cell viability.
Collapse
Affiliation(s)
- Roba Abukwaik
- Mathematics Department, King Abdulaziz University, Rabigh, Saudi Arabia.
- School of Mathematics, University of Birmingham, Birmingham, B15 2TS, UK.
| | - Elias Vera-Siguenza
- School of Mathematics, University of Birmingham, Birmingham, B15 2TS, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Daniel Tennant
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Fabian Spill
- School of Mathematics, University of Birmingham, Birmingham, B15 2TS, UK.
| |
Collapse
|
3
|
The Correlation of HK2 Gene Expression with the Occurrence, Immune Cell Infiltration, and Prognosis of Renal Cell Carcinoma. DISEASE MARKERS 2022; 2022:1452861. [PMID: 35265223 PMCID: PMC8898847 DOI: 10.1155/2022/1452861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 11/29/2022]
Abstract
Objectives Hexokinase 2 (HK2) is one of the key factors involved in the development of several human cancers. However, its role in immune cell infiltration (ICI) and tumor development in renal cell carcinoma is not yet known. Thus, we aimed to explore its relationship with ICI, overall survival, and prognosis of renal cell carcinoma. Methods In this study, RNA-seq data from renal cancer and normal tissues were extracted from TCGA and the relationship between HK2 expression and pathological features of RCC patients was analyzed using the GEPIA and UALCAN databases. Subsequently, Western blot and qRT-PCR were performed to analyze the protein and mRNA expression of HK2 in renal cell carcinoma tissues and cell lines. Lastly, various bioinformatics tools were applied to determine the immune cell infiltration, survival, and developing prediction model. Results The analysis of RNA-seq data revealed a high expression of HK2 in renal cell carcinoma; furthermore, Western blot and qRT-PCR also showed high expression of HK2 in renal cancer tissues and cell lines. The high expression of HK2 showed a significant positive correlation with the advanced stage of the tumor, lymph node metastasis, and worst survival in renal carcinoma patients. The high expression of HK2 was further identified as an independent risk factor of RCC patients; it also showed a significant positive immune cell infiltration RCC tumor microenvironment including macrophages, B cells, neutrophils, dendritic cells, and CD8+ T cells. Conclusion the expression of HK2 is positively correlated with the immune cell infiltration and prognosis of renal cell carcinoma patients, thus playing an important role in renal cancer development.
Collapse
|
4
|
Shirvani H, Bazgir B, Rahimi M, Isanejad A, Samadi M, Sobhani V, Alizadeh AM, Arabzadeh E. Therapeutic and preventive effects of exercise training on metabolic regulators/markers in mouse colorectal cancer cells. SPORT SCIENCES FOR HEALTH 2021. [DOI: 10.1007/s11332-021-00826-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
5
|
Modesti L, Danese A, Angela Maria Vitto V, Ramaccini D, Aguiari G, Gafà R, Lanza G, Giorgi C, Pinton P. Mitochondrial Ca 2+ Signaling in Health, Disease and Therapy. Cells 2021; 10:cells10061317. [PMID: 34070562 PMCID: PMC8230075 DOI: 10.3390/cells10061317] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
The divalent cation calcium (Ca2+) is considered one of the main second messengers inside cells and acts as the most prominent signal in a plethora of biological processes. Its homeostasis is guaranteed by an intricate and complex system of channels, pumps, and exchangers. In this context, by regulating cellular Ca2+ levels, mitochondria control both the uptake and release of Ca2+. Therefore, at the mitochondrial level, Ca2+ plays a dual role, participating in both vital physiological processes (ATP production and regulation of mitochondrial metabolism) and pathophysiological processes (cell death, cancer progression and metastasis). Hence, it is not surprising that alterations in mitochondrial Ca2+ (mCa2+) pathways or mutations in Ca2+ transporters affect the activities and functions of the entire cell. Indeed, it is widely recognized that dysregulation of mCa2+ signaling leads to various pathological scenarios, including cancer, neurological defects and cardiovascular diseases (CVDs). This review summarizes the current knowledge on the regulation of mCa2+ homeostasis, the related mechanisms and the significance of this regulation in physiology and human diseases. We also highlight strategies aimed at remedying mCa2+ dysregulation as promising therapeutical approaches.
Collapse
Affiliation(s)
- Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Alberto Danese
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Daniela Ramaccini
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Roberta Gafà
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Giovanni Lanza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
- Correspondence: ; Tel.: +39-0532-455802
| |
Collapse
|
6
|
Schulz SVW, Schumann U, Otto S, Kirsten J, Treff G, Janni W, Huober J, Leinert E, Steinacker JM, Bizjak DA. Two-year follow-up after a six-week high-intensity training intervention study with breast cancer patients: physiological, psychological and immunological differences. Disabil Rehabil 2021; 44:4813-4820. [PMID: 33974472 DOI: 10.1080/09638288.2021.1921861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Previously we demonstrated the feasibility of a six-week-long combination of high-intensity interval endurance and strength training (HIT/HIRT) for women with nonmetastatic breast cancer leading to improvements in psychological well-being and performance. Now we report results of a 24-month follow-up. METHODS Previous intervention (IG, n = 10; 58.7 ± 8.4yrs) and control group (CG, n = 9; 58.8 ± 6.6yrs) were asked for follow-up examinations 12 (T12) and 24 months (T24) after cessation of the supervised training (POST). Medical history, mental well-being, performance and immunological variables were analyzed with respect to intervention start (PRE). RESULTS IG maximum oxygen consumption (⩒O2peak) 12%-improved POST (p = 0.05) and declined to baseline values T24, while CG ⩒O2peak increased 12% T24 (p = 0.01). IG strength (1RM) increased 31% POST (p < 0.001) and remained above baseline level T24 (p = 0.003), whereas CG 1RM slightly improved T24 (+19%, p = 0.034). IG Anxiety and Depression decreased POST and did not change until T24. IG C-reactive protein decreased POST and increased to pre-exercise levels T24. CG immunological/inflammatory/life quality markers did not change. CONCLUSIONS Six weeks of HIT/HIRT by breast cancer patients can induce similar beneficial effects like two years of convalescence, but outcomes were unstable and showed a fast backslide in aerobic capacity, activity level and in pro-inflammatory state within 12 months.IMPLICATIONS FOR REHABILITATIONHigh-intensity interval endurance and strength training (HIT/HIRT) for female breast cancer patients was shown to improve psychological well-being and performance, but long-term effects/adherence are unknown.Significant backslides in aerobic capacity, activity level as well as in the pro-inflammatory response after one and two years are observed and should be monitored.Continuous supervision and/or support of breast cancer patients before, during, and after medical care due to poor training adherence when voluntarily executed is recommended.
Collapse
Affiliation(s)
- Sebastian V W Schulz
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| | - Uwe Schumann
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| | - Stephanie Otto
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| | - Johannes Kirsten
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| | - Gunnar Treff
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Jens Huober
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Elena Leinert
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Jürgen M Steinacker
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| | - Daniel A Bizjak
- Division of Sports and Rehabilitation Medicine, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
7
|
Friedman Y, Hizi A, Avni D, Bakhanashvili M. Mitochondrial matrix-localized p53 participates in degradation of mitochondrial RNAs. Mitochondrion 2021; 58:200-212. [PMID: 33775872 DOI: 10.1016/j.mito.2021.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/11/2021] [Accepted: 03/22/2021] [Indexed: 11/18/2022]
Abstract
Mitochondrial RNA degradation plays an important role in maintenance of the mitochondria genetic integrity. Mitochondrial localization of p53 was observed in non-stressed and stressed cells. p53, as an RNA-binding protein, exerts 3'→5' exoribonuclease activity. The data suggest that in non-stressed cells, mitochondrial matrix-localized p53, with exoribonuclease activity, may play a housekeeping positive role. p53, through restriction the formation of new RNA/DNA hybrid and processing R-loop, might serve as mitochondrial R-loop suppressor. Conversely, stress-induced matrix-p53 decreases the amount of mitochondrial single-stranded RNA transcripts (including polyA- and non-polyA RNAs), thereby leading to the decline in the amount of mitochondria-encoded oxidative phosphorylation components.
Collapse
Affiliation(s)
- Yael Friedman
- Infectious Diseases Unit, Sheba Medical Center, Tel-Hashomer 5265601, Israel
| | - Amnon Hizi
- Department of Cellular and Developmental Biology, Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Dror Avni
- Lab. Mol. Cell Biology, Center for Cancer Research & Dep. of Medicine C, Sheba Medical Center, Tel Hashomer, Israel
| | - Mary Bakhanashvili
- Infectious Diseases Unit, Sheba Medical Center, Tel-Hashomer 5265601, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|
8
|
Walcott FL, Wang PY, Bryla CM, Huffstutler RD, Singh N, Pollak MN, Khincha PP, Savage SA, Mai PL, Dodd KW, Hwang PM, Fojo AT, Annunziata CM. Pilot Study Assessing Tolerability and Metabolic Effects of Metformin in Patients With Li-Fraumeni Syndrome. JNCI Cancer Spectr 2021; 4:pkaa063. [PMID: 33490865 DOI: 10.1093/jncics/pkaa063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 11/12/2022] Open
Abstract
Background Li-Fraumeni syndrome (LFS) is a highly penetrant autosomal dominant cancer predisposition disorder caused by germline TP53 pathogenic variants. Patients with LFS have increased oxidative phosphorylation capacity in skeletal muscle and oxidative stress in blood. Metformin inhibits oxidative phosphorylation, reducing available energy for cancer cell proliferation and decreasing production of reactive oxygen species that cause DNA damage. Thus, metformin may provide pharmacologic risk reduction for cancer in patients with LFS, but its safety in nondiabetic patients with germline TP53 pathogenic variants has not been documented. Methods This study assessed safety and tolerability of metformin in nondiabetic LFS patients and measured changes in metabolic profiles. Adult patients with LFS and germline TP53 variant received 14 weeks of metformin. Blood samples were obtained for measurement of serum insulin-like growth factor-1, insulin, and insulin-like growth factor binding protein 3. Hepatic mitochondrial function was assessed with fasting exhaled CO2 after ingestion of 13C-labeled methionine. Changes in serum metabolome were measured. All statistical tests were 2-sided. Results We enrolled 26 participants: 20 females and 6 males. The most common adverse events were diarrhea (50.0%) and nausea (46.2%). Lactic acidosis did not occur, and there were no changes in fasting glucose. Cumulative mean 13C exhalation was statistically significantly suppressed by metformin (P = .001). Mean levels of insulin-like growth factor binding protein 3 and insulin-like growth factor-1 were statistically significantly lowered (P = .02). Lipid metabolites and branched-chain amino acids accumulated. Conclusions Metformin was safe and tolerable in patients with LFS. It suppressed hepatic mitochondrial function as expected in these individuals. This study adds to the rationale for development of a pharmacologic risk-reduction clinical trial of metformin in LFS.
Collapse
Affiliation(s)
- Farzana L Walcott
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ping-Yuan Wang
- Cardiovascular Branch, National Heart Lung Blood Institute, Bethesda, MD, USA
| | - Christine M Bryla
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Neha Singh
- George Washington Medical School, Washington, DC, USA
| | | | - Payal P Khincha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Phuong L Mai
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin W Dodd
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Paul M Hwang
- Cardiovascular Branch, National Heart Lung Blood Institute, Bethesda, MD, USA
| | - Antonio T Fojo
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | |
Collapse
|
9
|
Pasquale V, Ducci G, Campioni G, Ventrici A, Assalini C, Busti S, Vanoni M, Vago R, Sacco E. Profiling and Targeting of Energy and Redox Metabolism in Grade 2 Bladder Cancer Cells with Different Invasiveness Properties. Cells 2020; 9:cells9122669. [PMID: 33322565 PMCID: PMC7764708 DOI: 10.3390/cells9122669] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Bladder cancer is one of the most prevalent deadly diseases worldwide. Grade 2 tumors represent a good window of therapeutic intervention, whose optimization requires high resolution biomarker identification. Here we characterize energy metabolism and cellular properties associated with spreading and tumor progression of RT112 and 5637, two Grade 2 cancer cell lines derived from human bladder, representative of luminal-like and basal-like tumors, respectively. The two cell lines have similar proliferation rates, but only 5637 cells show efficient lateral migration. In contrast, RT112 cells are more prone to form spheroids. RT112 cells produce more ATP by glycolysis and OXPHOS, present overall higher metabolic plasticity and are less sensitive than 5637 to nutritional perturbation of cell proliferation and migration induced by treatment with 2-deoxyglucose and metformin. On the contrary, spheroid formation is less sensitive to metabolic perturbations in 5637 than RT112 cells. The ability of metformin to reduce, although with different efficiency, cell proliferation, sphere formation and migration in both cell lines, suggests that OXPHOS targeting could be an effective strategy to reduce the invasiveness of Grade 2 bladder cancer cells.
Collapse
Affiliation(s)
- Valentina Pasquale
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Giacomo Ducci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Gloria Campioni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Adria Ventrici
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
| | - Chiara Assalini
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
| | - Stefano Busti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| | - Riccardo Vago
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Hospital, 20132 Milan, Italy;
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (V.P.); (G.D.); (G.C.); (A.V.); (S.B.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
- Correspondence: (M.V.); (R.V.); (E.S.); Tel.: +39-02-6448-3525 (M.V.); +39-02-2643-5664 (R.V.); +39-02-6448-3379 (E.S.)
| |
Collapse
|
10
|
Beneficial Molecular Adaptations In BRCA-Mutation Carriers By Combined HIT/HIRT Intervention: Results From A Pilot Study. Cancers (Basel) 2020; 12:cancers12061526. [PMID: 32532068 PMCID: PMC7352264 DOI: 10.3390/cancers12061526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/02/2020] [Accepted: 06/06/2020] [Indexed: 12/30/2022] Open
Abstract
Based on growing evidence that breast cancer (BRCA) also plays a pivotal role in the regulation of skeletal muscle metabolism and the response to anti-oxidative stress, we examined the influence of regular exercise in human BRCA mutation carriers on their BRCA1 gene/protein expression and inflammatory/oxidative response. Sixteen BRCA-mutation carriers were assigned to an intervention (IG) or control group (CG). IG received a combination of high-intensity interval endurance (HIT) and strength training (HIRT) for six weeks, whereas CG received a low-intensity activity program. Before (T0) and at the end of the intervention (T1), muscle biopsy, physiological performance, blood withdrawal and anthropometry were obtained. Parameters included: Muscle BRCA1 gene/protein expression, inflammatory/oxidative stress, anti-oxidative capacity, peak oxygen capacity (VO2peak) and 1-repetition maximum (1-RM) at six different training machines. VO2peak and 1-RM of IG were increased at T1 compared to T0, whereas CG performance, physiological and molecular parameters remained unchanged. IG showed increased BRCA1 protein concentration as well as anti-oxidative capacity, whereas gene expression was unaltered. IG inflammatory and oxidative damage did not differ between time points. Combined HIT/HIRT increases aerobic and strength performance of BRCA-mutation carriers with up regulated BRCA1 protein expression and improved anti-oxidative status without showing an increased inflammatory response.
Collapse
|
11
|
SLMP53-1 Inhibits Tumor Cell Growth through Regulation of Glucose Metabolism and Angiogenesis in a P53-Dependent Manner. Int J Mol Sci 2020; 21:ijms21020596. [PMID: 31963392 PMCID: PMC7013701 DOI: 10.3390/ijms21020596] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 01/10/2023] Open
Abstract
The Warburg effect is an emerging hallmark of cancer, which has the tumor suppressor p53 as its major regulator. Herein, we unveiled that p53 activation by (S)-tryptophanol-derived oxazoloisoindolinone (SLMP53-1) mediated the reprograming of glucose metabolism in cancer cells and xenograft human tumor tissue, interfering with angiogenesis and migration. Particularly, we showed that SLMP53-1 regulated glycolysis by downregulating glucose transporter 1 (GLUT1), hexokinase-2 (HK2), and phosphofructokinase-2 isoform 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase-3 (PFKFB3) (key glycolytic enzymes), while upregulating the mitochondrial markers synthesis of cytochrome c oxidase 2 (SCO2), cytochrome c oxidase subunit 4 (COX4), and OXPHOS mitochondrial complexes. SLMP53-1 also downregulated the monocarboxylate transporter 4 (MCT4), causing the subsequent reduction of lactate export by cancer cells. Besides the acidification of the extracellular environment, SLMP53-1 further increased E-cadherin and reduced metalloproteinase-9 (MMP-9) expression levels in both cancer cells and xenograft human tumor tissue, which suggested the interference of SLMP53-1 in extracellular matrix remodeling and epithelial-to-mesenchymal transition. Consistently, SLMP53-1 depleted angiogenesis, decreasing endothelial cell tube formation and vascular endothelial growth factor (VEGF) expression levels. SLMP53-1 also exhibited synergistic growth inhibitory activity in combination with the metabolic modulator dichloroacetic acid. These data reinforce the promising application of the p53-activating agent SLMP53-1 in cancer therapy, by targeting p53-mediated pathways of growth and dissemination.
Collapse
|
12
|
Ahmed MA, O'Callaghan C, Chang ED, Jiang H, Vassilopoulos A. Context-Dependent Roles for SIRT2 and SIRT3 in Tumor Development Upon Calorie Restriction or High Fat Diet. Front Oncol 2020; 9:1462. [PMID: 31970087 PMCID: PMC6960403 DOI: 10.3389/fonc.2019.01462] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/05/2019] [Indexed: 12/27/2022] Open
Abstract
Calorie restriction (CR) is considered one of the most robust ways to extend life span and reduce the risk of age-related diseases, including cancer, as shown in many different organisms, whereas opposite effects have been associated with high fat diets (HFDs). Despite the proven contribution of sirtuins in mediating the effects of CR in longevity, the involvement of these nutrient sensors, specifically, in the diet-induced effects on tumorigenesis has yet to be elucidated. Previous studies focusing on SIRT1, do not support a critical role for this sirtuin family member in CR-mediated cancer prevention. However, the contribution of other family members which exhibit strong deacetylase activity is unexplored. To fill this gap, we aimed at investigating the role of SIRT2 and SIRT3 in mediating the anti and pro-tumorigenic effect of CR and HFD, respectively. Our results provide strong evidence supporting distinct, context-dependent roles played by these two family members. SIRT2 is indispensable for the protective effect of CR against tumorigenesis. On the contrary, SIRT3 exhibited oncogenic properties in the context of HFD-induced tumorigenesis, suggesting that SIRT3 inhibition may mitigate the cancer-promoting effects of HFD. Given the different functions regulated by SIRT2 and SIRT3, unraveling downstream targets/pathways involved may provide opportunities to develop new strategies for cancer prevention.
Collapse
Affiliation(s)
- Mohamed A Ahmed
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Radiation Biology Department, National Center for Radiation Research and Technology, Cairo, Egypt
| | - Carol O'Callaghan
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Elliot D Chang
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Haiyan Jiang
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Athanassios Vassilopoulos
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
13
|
Cardial Tobias G, Lucas Penteado Gomes J, Paula Renó Soci U, Fernandes T, Menezes de Oliveira E. A Landscape of Epigenetic Regulation by MicroRNAs to the Hallmarks of Cancer and Cachexia: Implications of Physical Activity to Tumor Regression. Epigenetics 2019. [DOI: 10.5772/intechopen.84847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
14
|
Blandino G, Valenti F, Sacconi A, Di Agostino S. Wild type- and mutant p53 proteins in mitochondrial dysfunction: emerging insights in cancer disease. Semin Cell Dev Biol 2019; 98:105-117. [PMID: 31112799 DOI: 10.1016/j.semcdb.2019.05.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
Deregulated cell metabolism is one of the cancer hallmarks. Mitochondrial DNA mutations and enzyme defects, aberrant tumor suppressor or oncogenic activities cause mitochondrial dysfunction leading to deregulated cellular energetics. The tumor suppressor protein, p53 is a tetrameric transcription factor that in response to diverse genotoxic and non-genotoxic insults activates a plethora of target genes to preserve genome integrity. In the last two decades the discovery of cytoplasmic p53 localization focused intense research on its extra-nuclear functions. The ability of p53 to induce apoptosis acting directly at mitochondria and the related mechanisms of p53 localization and translocation in the cytoplasm have been investigated. A role of cytoplasmic p53 in autophagy, pentose phosphate pathway, fatty acid synthesis and oxidation, and drug response has been proposed. TP53 gene is mutated in more than half of human cancers. In parallel to loss of tumor suppressive functions, mutant p53 proteins often gain new tumorigenic activities (GOF, gain of function). It has been recently shown that mutant p53 proteins mediate metabolic changes thereby promoting cancer development and metastases. Here we review the contribution of either wild-type p53 or mutant p53 proteins to the fine-tuning of mitochondrial metabolism of both normal and cancer cells. Greater knowledge at the mechanistic level might provide insights to develop new cancer therapeutic approaches.
Collapse
Affiliation(s)
- Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Department of Diagnostic Research and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy.
| | - Fabio Valenti
- Oncogenomic and Epigenetic Unit, Department of Diagnostic Research and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Andrea Sacconi
- Oncogenomic and Epigenetic Unit, Department of Diagnostic Research and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Silvia Di Agostino
- Oncogenomic and Epigenetic Unit, Department of Diagnostic Research and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, 00144, Italy.
| |
Collapse
|
15
|
Abstract
SIGNIFICANCE Hexokinases are key enzymes that are responsible for the first reaction of glycolysis, but they also moonlight other cellular processes, including mitochondrial redox signaling regulation. Modulation of hexokinase activity and spatiotemporal location by reactive oxygen and nitrogen species as well as other gasotransmitters serves as the basis for a unique, underexplored method of tight and flexible regulation of these fundamental enzymes. Recent Advances: Redox modifications of thiols serve as a molecular code that enables the precise and complex regulation of hexokinases. Redox regulation of hexokinases is also used by multiple parasites to cause widespread and severe diseases, including malaria, Chagas disease, and sleeping sickness. Redox-active molecules affect each other, and the moonlighting activity of hexokinases provides another feedback loop that affects the cellular redox status and is hijacked in malignantly transformed cells. CRITICAL ISSUES Several compounds affect the redox status of hexokinases in vivo. These include the dehydroascorbic acid (oxidized form of vitamin C), pyrrolidinium porrolidine-1-carbodithioate (contraceptive), peroxynitrite (product of ethanol metabolism), alloxan (a glucose analog), and isobenzothiazolinone ebselen. However, very limited information is available regarding which amino acid residues in hexokinases are affected by redox signaling. Except in cases of monogenic diabetes, direct evidence is absent for disease phenotypes that are associated with variations within motifs that are susceptible to redox signaling. FUTURE DIRECTIONS Further studies should address the propensity of hexokinases and their disease-associated variants to participate in redox regulation. Robust and straightforward proteomic methods are needed to understand the context and consequences of hexokinase-mediated redox regulation in health and disease.
Collapse
Affiliation(s)
- Petr Heneberg
- Third Faculty of Medicine, Charles University , Prague, Czech Republic
| |
Collapse
|
16
|
Labuschagne CF, Zani F, Vousden KH. Control of metabolism by p53 - Cancer and beyond. Biochim Biophys Acta Rev Cancer 2018; 1870:32-42. [PMID: 29883595 PMCID: PMC6102416 DOI: 10.1016/j.bbcan.2018.06.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 12/18/2022]
Abstract
p53 is an important tumour suppressor gene, with loss of p53 contributing to the development of most human cancers. However, the activation of p53 in response to stress signals underpins a role for p53 in diverse aspects of health and disease. Activities of p53 that regulate metabolism can play a role in maintaining homeostasis and protecting cells from damage - so preventing disease development. By contrast, either loss or over-activation of p53 can contribute to numerous metabolic pathologies, including aging, obesity and diabetes.
Collapse
Affiliation(s)
| | - Fabio Zani
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Karen H Vousden
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
17
|
Zhao Y, Zhang L, Wu Y, Dai Q, Zhou Y, Li Z, Yang L, Guo Q, Lu N. Selective anti-tumor activity of wogonin targeting the Warburg effect through stablizing p53. Pharmacol Res 2018; 135:49-59. [PMID: 30031170 DOI: 10.1016/j.phrs.2018.07.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/15/2018] [Accepted: 07/12/2018] [Indexed: 01/11/2023]
Abstract
Most cancer cells generate energy through aerobic glycolysis to enable their rapid growth and proliferation, which is a phenomenon known as Warburg effect. Inhibition of aerobic glycolysis reduces lactate and ATP generation in cancer cells, and ultimately kills tumor cells. Increasing evidence suggests that wogonin, a flavonoid isolated from Scutellaria baicalensis Georgi, exhibits potent anti-tumor effects in vivo and in vitro. However, the role of wogonin in the aerobic glycolysis of tumor cells has not yet been elucidated. In this study, the effect of wogonin on glucose uptake, lactate generation and ATP content is assessed in colon, ovarian and hepatocellular cancer cells. The results indicate that wogonin reduces glycolysis and cell proliferation in cancer cells expressing wild-type p53 but not mutated p53. Wogonin increases the expression of p53 and p53-inducible glycolysis and apoptosis regulator (TIGAR), while decreases glucose transporter 1 (GLUT1) and some key glycolytic enzymes. Expressing wild-type and mutant-type p53 in HCT116 p53-/- cells proved that the inhibitory effect of wogonin on glycolysis in cancer cells is dependent on wild type p53. Mechanistically, wogonin induced the phosphorylation and acetylation of p53 and inhibited the expression of MDM2 to enhance the stability of p53. Furthermore, wogonin suppressed the growth and glycolysis of transplanted wild-type p53 expressing A2780 cells on nude mice, but did not affect mutant-type p53 expressing HT-29 cells. In conclusion, these findings explain the broad anti-tumor effect of wogonin, and offer a novel avenue for the therapeutic strategy in cancer.
Collapse
Affiliation(s)
- Yikai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Lulu Zhang
- Department of Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China
| | - Yifan Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Qinsheng Dai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Zhiyu Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Lin Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China; Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China; Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| |
Collapse
|
18
|
Huang J, Long Z, Lin W, Liao X, Xie Y, Liu L, Ma W. Integrative omics analysis of p53-dependent regulation of metabolism. FEBS Lett 2018; 592:380-393. [PMID: 29323703 DOI: 10.1002/1873-3468.12968] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 12/08/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022]
Abstract
Accumulated evidence in the last decade implies that regulation of metabolism by p53 represents a reviving mechanism vital to prevent tumorigenesis. To gain a more in-depth understanding of metabolic regulation by baseline levels of p53, we employed both metabolomics and transcriptomics analysis with human colon cancer cell-line HCT116 depleted of p53. Metabolomics analyses with UPLC/quadrupole time-of-flight mass spectrometry identified 283 significantly changed metabolites including 138 important metabolites. Transcriptomics analysis with microarray revealed 1317 differentially expressed genes. By integrated analysis of both omics data, we found nucleotides metabolism and sulfur-related metabolism are of great importance. Our study provided a pilot comprehensive view of the metabolism regulated by p53 and suggests several potential p53 targets in metabolism for further study.
Collapse
Affiliation(s)
- Jiajun Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, MUST, China
| | - Ze Long
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, MUST, China
| | - Wanjun Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, MUST, China
| | - Xiaolin Liao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, MUST, China
| | - Ying Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, MUST, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, MUST, China
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, MUST, China
| |
Collapse
|
19
|
Maqueda M, Roca E, Brotons D, Soria JM, Perera A. Affected pathways and transcriptional regulators in gene expression response to an ultra-marathon trail: Global and independent activity approaches. PLoS One 2017; 12:e0180322. [PMID: 29028836 PMCID: PMC5640184 DOI: 10.1371/journal.pone.0180322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/14/2017] [Indexed: 12/18/2022] Open
Abstract
Gene expression (GE) analyses on blood samples from marathon and half-marathon runners have reported significant impacts on the immune and inflammatory systems. An ultra-marathon trail (UMT) represents a greater effort due to its more testing conditions. For the first time, we report the genome-wide GE profiling in a group of 16 runners participating in an 82 km UMT competition. We quantified their differential GE profile before and after the race using HuGene2.0st microarrays (Affymetrix Inc., California, US). The results obtained were decomposed by means of an independent component analysis (ICA) targeting independent expression modes. We observed significant differences in the expression levels of 5,084 protein coding genes resulting in an overrepresentation of 14% of the human biological pathways from the Kyoto Encyclopedia of Genes and Genomes database. These were mainly clustered on terms related with protein synthesis repression, altered immune system and infectious diseases related mechanisms. In a second analysis, 27 out of the 196 transcriptional regulators (TRs) included in the Open Regulatory Annotation database were overrepresented. Among these TRs, we identified transcription factors from the hypoxia-inducible factors (HIF) family EPAS1 (p< 0.01) and HIF1A (p<0.001), and others jointly described in the gluconeogenesis program such as HNF4 (p< 0.001), EGR1 (p<0.001), CEBPA (p< 0.001) and a highly specific TR, YY1 (p<0.01). The five independent components, obtained from ICA, further revealed a down-regulation of 10 genes distributed in the complex I, III and V from the electron transport chain. This mitochondrial activity reduction is compatible with HIF-1 system activation. The vascular endothelial growth factor (VEGF) pathway, known to be regulated by HIF, also emerged (p<0.05). Additionally, and related to the brain rewarding circuit, the endocannabinoid signalling pathway was overrepresented (p<0.05).
Collapse
Affiliation(s)
- Maria Maqueda
- Department of ESAII, Center for Biomedical Engineering Research, Universitat Politècnica de Catalunya, Barcelona, Catalonia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Catalonia, Spain
- * E-mail:
| | - Emma Roca
- Summit 2014 S.L., Centelles, Barcelona, Catalonia Spain
- Department of Electronic Engineering, Center for Biomedical Engineering Research, Universitat Politècnica de Catalunya, Barcelona, Catalonia, Spain
| | | | - Jose Manuel Soria
- Unit of Genomics of Complex Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Alexandre Perera
- Department of ESAII, Center for Biomedical Engineering Research, Universitat Politècnica de Catalunya, Barcelona, Catalonia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Catalonia, Spain
| |
Collapse
|
20
|
San-Millán I, Brooks GA. Reexamining cancer metabolism: lactate production for carcinogenesis could be the purpose and explanation of the Warburg Effect. Carcinogenesis 2017; 38:119-133. [PMID: 27993896 PMCID: PMC5862360 DOI: 10.1093/carcin/bgw127] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 12/08/2016] [Indexed: 12/15/2022] Open
Abstract
Herein, we use lessons learned in exercise physiology and metabolism to propose that augmented lactate production (‘lactagenesis’), initiated by gene mutations, is the reason and purpose of the Warburg Effect and that dysregulated lactate metabolism and signaling are the key elements in carcinogenesis. Lactate-producing (‘lactagenic’) cancer cells are characterized by increased aerobic glycolysis and excessive lactate formation, a phenomenon described by Otto Warburg 93 years ago, which still remains unexplained. After a hiatus of several decades, interest in lactate as a player in cancer has been renewed. In normal physiology, lactate, the obligatory product of glycolysis, is an important metabolic fuel energy source, the most important gluconeogenic precursor, and a signaling molecule (i.e. a ‘lactormone’) with major regulatory properties. In lactagenic cancers, oncogenes and tumor suppressor mutations behave in a highly orchestrated manner, apparently with the purpose of increasing glucose utilization for lactagenesis purposes and lactate exchange between, within and among cells. Five main steps are identified (i) increased glucose uptake, (ii) increased glycolytic enzyme expression and activity, (iii) decreased mitochondrial function, (iv) increased lactate production, accumulation and release and (v) upregulation of monocarboxylate transporters MTC1 and MCT4 for lactate exchange. Lactate is probably the only metabolic compound involved and necessary in all main sequela for carcinogenesis, specifically: angiogenesis, immune escape, cell migration, metastasis and self-sufficient metabolism. We hypothesize that lactagenesis for carcinogenesis is the explanation and purpose of the Warburg Effect. Accordingly, therapies to limit lactate exchange and signaling within and among cancer cells should be priorities for discovery.
Collapse
Affiliation(s)
- Iñigo San-Millán
- Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine, Aurora, CO 80045, USA.,Physiology Laboratory, CU Sports Medicine and Performance Center, Boulder, CO 80309, USA and
| | - George A Brooks
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
21
|
A Review on the Role of Physical Activity in Cancer Prevention: Middle East Reports. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2017. [DOI: 10.5812/ijcm.8992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Krasich R, Copeland WC. DNA polymerases in the mitochondria: A critical review of the evidence. FRONT BIOSCI-LANDMRK 2017; 22:692-709. [PMID: 27814640 PMCID: PMC5485829 DOI: 10.2741/4510] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since 1970, the DNA polymerase gamma (PolG) has been known to be the DNA polymerase responsible for replication and repair of mitochondrial DNA, and until recently it was generally accepted that this was the only polymerase present in mitochondria. However, recent data has challenged that opinion, as several polymerases are now proposed to have activity in mitochondria. To date, their exact role of these other DNA polymerases is unclear and the amount of evidence supporting their role in mitochondria varies greatly. Further complicating matters, no universally accepted standards have been set for definitive proof of the mitochondrial localization of a protein. To gain an appreciation of these newly proposed DNA polymerases in the mitochondria, we review the evidence and standards needed to establish the role of a polymerase in the mitochondria. Employing PolG as an example, we established a list of criteria necessary to verify the existence and function of new mitochondrial proteins. We then apply this criteria towards several other putative mitochondrial polymerases. While there is still a lot left to be done in this exciting new direction, it is clear that PolG is not acting alone in mitochondria, opening new doors for potential replication and repair mechanisms.
Collapse
Affiliation(s)
- Rachel Krasich
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - William C Copeland
- Genome Integrity and Structural Biology Laboratory, NIEHS, National Institutes of Health, 111 T.W. Alexander Dr., Bldg. 101, Rm. E316, Research Triangle Park, NC 27709,
| |
Collapse
|
23
|
Wong S, Napoli E, Krakowiak P, Tassone F, Hertz-Picciotto I, Giulivi C. Role of p53, Mitochondrial DNA Deletions, and Paternal Age in Autism: A Case-Control Study. Pediatrics 2016; 137:peds.2015-1888. [PMID: 27033107 PMCID: PMC4811307 DOI: 10.1542/peds.2015-1888] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The tumor suppressor p53 responds to a variety of environmental stressors by regulating cell cycle arrest, apoptosis, senescence, DNA repair, bioenergetics and mitochondrial DNA (mtDNA) copy number maintenance. Developmental abnormalities have been reported in p53-deficient mice, and altered p53 and p53-associated pathways in autism (AU). Furthermore, via the Pten-p53 crosstalk, Pten haploinsufficient-mice have autisticlike behavior accompanied by brain mitochondrial dysfunction with accumulation of mtDNA deletions. METHODS mtDNA copy number and deletions, and p53 gene copy ratios were evaluated in peripheral blood monocytic cells from children aged 2-5 years with AU (n = 66), race-, gender-, and age-matched typically neurodeveloping children (n = 46), and both parents from each diagnostic group, recruited by the Childhood Autism Risk from Genes and Environment study at the University of California, Davis. RESULTS mtDNA deletions and higher p53 gene copy ratios were more common in children with AU and their fathers. The incidence of mtDNA deletions in fathers of children with AU was increased 1.9-fold over fathers of typically neurodeveloping children, suggesting a role for deficient DNA repair capacity not driven by paternal age. Deletions in mtDNA and altered p53 gene copy ratios seem to result from genetics (children with severity scores ≥8) and/or act in concert with environmental factors (children with 6-7 severity scores). CONCLUSIONS Given pro- and antioxidant activities of p53, and associations of genomic instability with disorders other than AU, our study suggests a link between DNA repair capacity, genomic instability in the 17p13.1 region influenced by environmental triggers, and AU diagnosis.
Collapse
Affiliation(s)
- Sarah Wong
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine
| | | | - Flora Tassone
- Biochemistry and Molecular Medicine, School of Medicine, and,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Davis, California
| | - Irva Hertz-Picciotto
- Departments of Public Health Sciences and,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Davis, California
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Davis, California
| |
Collapse
|
24
|
TP53 mutation, mitochondria and cancer. Curr Opin Genet Dev 2016; 38:16-22. [PMID: 27003724 DOI: 10.1016/j.gde.2016.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/09/2016] [Accepted: 02/22/2016] [Indexed: 12/20/2022]
Abstract
Under normal conditions, basal levels of wild-type p53 promote mitochondrial function through multiple mechanisms. Remarkably, some missense mutations of p53, in contrast to the null state, can result in the retention of its metabolic activities. These effects are particularly prominent in the mitochondria and demonstrate a functional role for mutant p53 in cancer metabolism. This review summarizes accumulating data on the mechanisms by which p53 missense mutations can regulate mitochondrial metabolism and promote the viability and survival of both normal and cancer cells, thus acting as a double edged sword for the host. Greater understanding of these mechanisms may provide insights for developing new treatment or preventive strategies against cancer.
Collapse
|
25
|
Weighted gene co-expression network analysis of pneumocytes under exposure to a carcinogenic dose of chloroprene. Life Sci 2016; 151:339-347. [PMID: 26916823 DOI: 10.1016/j.lfs.2016.02.074] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 02/16/2016] [Accepted: 02/20/2016] [Indexed: 02/06/2023]
Abstract
AIMS Occupational exposure to chloroprene via inhalation may lead to acute toxicity and chronic pulmonary diseases, including lung cancer. Currently, most research is focused on epidemiological studies of chloroprene production workers. The specific molecular mechanism of carcinogenesis by chloroprene in lung tissues still remains obscure, and specific candidate therapeutic targets for lung cancer are lacking. The present study identifies specific gene modules and valuable hubs associated with lung cancer. MAIN METHODS We downloaded the dataset GSE40795 from the Gene Expression Omnibus (GEO) and divided the dataset into the non-carcinogenic dose chloroprene exposed mice group and the carcinogenic dose chloroprene exposed mice group. With a systemic biological view, we discovered significantly altered gene modules between the two groups and identified hub genes in the carcinogenic dose exposed group using weighted co-expression network analysis (WGCNA). KEY FINDINGS A total of 2434 differentially expressed genes were identified. Twelve gene modules with multiple biological activities were related to the carcinogenesis of chloroprene in lung tissue. Seven hub genes that were critical for the carcinogenesis of chloroprene in lung tissue were ultimately identified, including Cftr, Hip1, Tbl1x, Ephx1, Cbr3, Antxr2 and Ccnd2. They were implicated in inflammatory response, cell transformation, gene transcription regulation, phase II detoxification, angiogenesis, cell adhesion, motility and the cell cycle. SIGNIFICANCE The seven hub genes may become valuable candidates for risk assessment biomarkers and therapeutic targets in lung cancer.
Collapse
|
26
|
Park JH, Zhuang J, Li J, Hwang PM. p53 as guardian of the mitochondrial genome. FEBS Lett 2016; 590:924-34. [PMID: 26780878 DOI: 10.1002/1873-3468.12061] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/22/2015] [Accepted: 01/04/2016] [Indexed: 12/16/2022]
Abstract
Participating in the repair of nuclear DNA is one mechanism by which p53 suppresses tumorigenesis, but there is growing evidence that p53 also helps maintain the mitochondrial genome through its translocation into mitochondria and interactions with mtDNA repair proteins. Because of the susceptibility of mtDNA to oxidative damage and replication errors, it is vital to protect mtDNA genomic stability to preserve health and fitness. Here, we focus on reviewing the evidence for the involvement of p53 in maintaining the integrity of mtDNA through its activities in both the nucleus and the mitochondria.
Collapse
Affiliation(s)
- Ji-Hoon Park
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Zhuang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Li
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul M Hwang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Walcott FL, Patel J, Lubet R, Rodriguez L, Calzone KA. Hereditary cancer syndromes as model systems for chemopreventive agent development. Semin Oncol 2016; 43:134-145. [PMID: 26970132 PMCID: PMC10433689 DOI: 10.1053/j.seminoncol.2015.09.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Research in chemoprevention has undergone a shift in emphasis for pragmatic reasons from large, phase III randomized studies to earlier phase studies focused on safety, mechanisms, and utilization of surrogate endpoints such as biomarkers instead of cancer incidence. This transition permits trials to be conducted in smaller populations and at substantially reduced costs while still yielding valuable information. This article will summarize some of the current chemoprevention challenges and the justification for the use of animal models to facilitate identification and testing of chemopreventive agents as illustrated though four inherited cancer syndromes. Preclinical models of inherited cancer syndromes serve as prototypical systems in which chemopreventive agents can be developed for ultimate application to both the sporadic and inherited cancer settings.
Collapse
Affiliation(s)
- Farzana L Walcott
- National Institutes of Health, National Cancer Institute, Division of Cancer Prevention, Bethesda, MD, USA.
| | - Jigar Patel
- National Institutes of Health, National Cancer Institute, Division of Cancer Prevention, Bethesda, MD, USA
| | - Ronald Lubet
- Consultant to National Institutes of Health, National Cancer Institute, Division of Cancer Prevention, Chemopreventive Agent Development Research Group, Bethesda, MD, USA
| | - Luz Rodriguez
- National Institutes of Health, National Cancer Institute, Division of Cancer Prevention, Gastrointestinal & Other Cancers Research, Bethesda, MD, USA
| | - Kathleen A Calzone
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Genetics Branch, Bethesda, MD, USA.
| |
Collapse
|
28
|
Qu D, Xu XM, Zhang M, Jiang TS, Zhang Y, Li SQ. Cbl participates in shikonin-induced apoptosis by negatively regulating phosphoinositide 3-kinase/protein kinase B signaling. Mol Med Rep 2015; 12:1305-13. [PMID: 25815461 DOI: 10.3892/mmr.2015.3510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 02/11/2015] [Indexed: 11/06/2022] Open
Abstract
Shikonin, a naturally occurring naphthoquinone, exhibits anti-tumorigenic activity. However, its precise mechanisms of action have remained elusive. In the present study, the involvement in the action of shikonin of the ubiquitin ligases Cbl-b and c-Cbl, which are negative regulators of phosphoinositide 3-kinase (PI3K) activation, was investigated. Shikonin was observed to reduce cell viability and induce apoptosis and G2/M phase arrest in lung cancer cells. In addition, shikonin increased the protein levels of B-cell lymphoma 2 (Bcl-2)-associated X and p53 and reduced those of Bcl-2. Additionally, shikonin inhibited PI3k/Akt activity and upregulated Cbl protein expression. In addition, a specific inhibitor of PI3K, LY294002, was observed to have a synergistic effect on the proliferation inhibition and apoptotic induction of A549 cells with shikonin. In conclusion, the results of the present study suggested that Cbl proteins promote shikonin-induced apoptosis by negatively regulating PI3K/Akt signaling in lung cancer cells.
Collapse
Affiliation(s)
- Dan Qu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xiao-Man Xu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Meng Zhang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ting-Shu Jiang
- Department of Respiratory Medicine, Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Yi Zhang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Sheng-Qi Li
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
29
|
Bouchard C, Antunes-Correa LM, Ashley EA, Franklin N, Hwang PM, Mattsson CM, Negrao CE, Phillips SA, Sarzynski MA, Wang PY, Wheeler MT. Personalized preventive medicine: genetics and the response to regular exercise in preventive interventions. Prog Cardiovasc Dis 2014; 57:337-46. [PMID: 25559061 DOI: 10.1016/j.pcad.2014.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Regular exercise and a physically active lifestyle have favorable effects on health. Several issues related to this theme are addressed in this report. A comment on the requirements of personalized exercise medicine and in-depth biological profiling along with the opportunities that they offer is presented. This is followed by a brief overview of the evidence for the contributions of genetic differences to the ability to benefit from regular exercise. Subsequently, studies showing that mutations in TP53 influence exercise capacity in mice and humans are succinctly described. The evidence for effects of exercise on endothelial function in health and disease also is covered. Finally, changes in cardiac and skeletal muscle in response to exercise and their implications for patients with cardiac disease are summarized. Innovative research strategies are needed to define the molecular mechanisms involved in adaptation to exercise and to translate them into useful clinical and public health applications.
Collapse
Affiliation(s)
- Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| | | | - Euan A Ashley
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Nina Franklin
- Department of Physical Therapy, Department of Medicine, Integrative Physiology Laboratory, University of Illinois at Chicago, Chicago, IL, USA
| | - Paul M Hwang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - C Mikael Mattsson
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA; The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Carlos E Negrao
- Heart Institute (InCor), Medical School, University of Sao Paulo, Sao Paulo, Brazil; School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Shane A Phillips
- Department of Physical Therapy, Department of Medicine, Integrative Physiology Laboratory, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark A Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Ping-Yuan Wang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Matthew T Wheeler
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
30
|
Pan LZ, Ahn DG, Sharif T, Clements D, Gujar SA, Lee PWK. The NAD+ synthesizing enzyme nicotinamide mononucleotide adenylyltransferase 2 (NMNAT-2) is a p53 downstream target. Cell Cycle 2014; 13:1041-8. [PMID: 24552824 DOI: 10.4161/cc.28128] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
NAD(+) metabolism plays key roles not only in energy production but also in diverse cellular physiology. Aberrant NAD(+) metabolism is considered a hallmark of cancer. Recently, the tumor suppressor p53, a major player in cancer signaling pathways, has been implicated as an important regulator of cellular metabolism. This notion led us to examine whether p53 can regulate NAD(+) biosynthesis in the cell. Our search resulted in the identification of nicotinamide mononucleotide adenylyltransferase 2 (NMNAT-2), a NAD(+) synthetase, as a novel downstream target gene of p53. We show that NMNAT-2 expression is induced upon DNA damage in a p53-dependent manner. Two putative p53 binding sites were identified within the human NMNAT-2 gene, and both were found to be functional in a p53-dependent manner. Furthermore, knockdown of NMNAT-2 significantly reduces cellular NAD(+) levels and protects cells from p53-dependent cell death upon DNA damage, suggesting an important functional role of NMNAT-2 in p53-mediated signaling. Our demonstration that p53 modulates cellular NAD(+) synthesis is congruent with p53's emerging role as a key regulator of metabolism and related cell fate.
Collapse
Affiliation(s)
- Lu-Zhe Pan
- Department of Microbiology and Immunology; Dalhousie University; Halifax, Nova Scotia, Canada
| | - Dae-Gyun Ahn
- Department of Microbiology and Immunology; Dalhousie University; Halifax, Nova Scotia, Canada
| | - Tanveer Sharif
- Department of Microbiology and Immunology; Dalhousie University; Halifax, Nova Scotia, Canada
| | - Derek Clements
- Department of Pathology; Dalhousie University; Halifax, Nova Scotia, Canada
| | - Shashi A Gujar
- Department of Microbiology and Immunology; Dalhousie University; Halifax, Nova Scotia, Canada; Strategy & Organizational Performance; IWK Health Centre; Halifax, Nova Scotia, Canada
| | - Patrick W K Lee
- Department of Microbiology and Immunology; Dalhousie University; Halifax, Nova Scotia, Canada; Department of Pathology; Dalhousie University; Halifax, Nova Scotia, Canada
| |
Collapse
|
31
|
Tang WH, Stitham J, Jin Y, Liu R, Lee SH, Du J, Atteya G, Gleim S, Spollett G, Martin K, Hwa J. Aldose reductase-mediated phosphorylation of p53 leads to mitochondrial dysfunction and damage in diabetic platelets. Circulation 2014; 129:1598-609. [PMID: 24474649 DOI: 10.1161/circulationaha.113.005224] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Platelet abnormalities are well-recognized complications of diabetes mellitus. Mitochondria play a central role in platelet metabolism and activation. Mitochondrial dysfunction is evident in diabetes mellitus. The molecular pathway for hyperglycemia-induced mitochondrial dysfunction in platelets in diabetes mellitus is unknown. METHODS AND RESULTS Using both human and humanized mouse models, we report that hyperglycemia-induced aldose reductase activation and subsequent reactive oxygen species production lead to increased p53 phosphorylation (Ser15), which promotes mitochondrial dysfunction, damage, and rupture by sequestration of the antiapoptotic protein Bcl-xL. In a glucose dose-dependent manner, severe mitochondrial damage leads to loss of mitochondrial membrane potential and platelet apoptosis (cytochrome c release, caspase 3 activation, and phosphatidylserine exposure). Although platelet hyperactivation, mitochondrial dysfunction, aldose reductase activation, reactive oxygen species production, and p53 phosphorylation are all induced by hyperglycemia, we demonstrate that platelet apoptosis and hyperactivation are 2 distinct states that depend on the severity of the hyperglycemia and mitochondrial damage. Combined, both lead to increased thrombus formation in a mouse blood stasis model. CONCLUSIONS Aldose reductase contributes to diabetes-mediated mitochondrial dysfunction and damage through the activation of p53. The degree of mitochondrial dysfunction and damage determines whether hyperactivity (mild damage) or apoptosis (severe damage) will ensue. These signaling components provide novel therapeutic targets for thrombotic complications in diabetes mellitus.
Collapse
Affiliation(s)
- Wai Ho Tang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (W.H.T., J.S., Y.J., R.L., S.H.L., J.D., G.A., S.G., K.M., J.H.) and Section of Endocrinology and Metabolism, Department of Internal Medicine (G.S.), Yale University School of Medicine, New Haven, CT
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Complex Systems Biology of Networks: The Riddle and the Challenge. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
33
|
Seyfried TN, Flores RE, Poff AM, D'Agostino DP. Cancer as a metabolic disease: implications for novel therapeutics. Carcinogenesis 2013; 35:515-27. [PMID: 24343361 PMCID: PMC3941741 DOI: 10.1093/carcin/bgt480] [Citation(s) in RCA: 318] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Emerging evidence indicates that cancer is primarily a metabolic disease involving disturbances in energy production through respiration and fermentation. The genomic instability observed in tumor cells and all other recognized hallmarks of cancer are considered downstream epiphenomena of the initial disturbance of cellular energy metabolism. The disturbances in tumor cell energy metabolism can be linked to abnormalities in the structure and function of the mitochondria. When viewed as a mitochondrial metabolic disease, the evolutionary theory of Lamarck can better explain cancer progression than can the evolutionary theory of Darwin. Cancer growth and progression can be managed following a whole body transition from fermentable metabolites, primarily glucose and glutamine, to respiratory metabolites, primarily ketone bodies. As each individual is a unique metabolic entity, personalization of metabolic therapy as a broad-based cancer treatment strategy will require fine-tuning to match the therapy to an individual’s unique physiology.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA and
| | | | | | | |
Collapse
|
34
|
Bartlett JD, Close GL, Drust B, Morton JP. The Emerging Role of p53 in Exercise Metabolism. Sports Med 2013; 44:303-9. [DOI: 10.1007/s40279-013-0127-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
35
|
Skrzypek K, Tertil M, Golda S, Ciesla M, Weglarczyk K, Collet G, Guichard A, Kozakowska M, Boczkowski J, Was H, Gil T, Kuzdzal J, Muchova L, Vitek L, Loboda A, Jozkowicz A, Kieda C, Dulak J. Interplay between heme oxygenase-1 and miR-378 affects non-small cell lung carcinoma growth, vascularization, and metastasis. Antioxid Redox Signal 2013; 19:644-60. [PMID: 23617628 PMCID: PMC3740397 DOI: 10.1089/ars.2013.5184] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AIMS Heme oxygenase-1 (HO-1, HMOX1) can prevent tumor initiation; while in various tumors, it has been demonstrated to promote growth, angiogenesis, and metastasis. Here, we investigated whether HMOX1 can modulate microRNAs (miRNAs) and regulate human non-small cell lung carcinoma (NSCLC) development. RESULTS Stable HMOX1 overexpression in NSCLC NCI-H292 cells up-regulated tumor-suppressive miRNAs, whereas it significantly diminished the expression of oncomirs and angiomirs. The most potently down-regulated was miR-378. HMOX1 also up-regulated p53, down-regulated angiopoietin-1 (Ang-1) and mucin-5AC (MUC5AC), reduced proliferation, migration, and diminished angiogenic potential. Carbon monoxide was a mediator of HMOX1 effects on proliferation, migration, and miR-378 expression. In contrast, stable miR-378 overexpression decreased HMOX1 and p53; while enhanced expression of MUC5AC, vascular endothelial growth factor (VEGF), interleukin-8 (IL-8), and Ang-1, and consequently increased proliferation, migration, and stimulation of endothelial cells. Adenoviral delivery of HMOX1 reversed miR-378 effect on the proliferation and migration of cancer cells. In vivo, HMOX1 overexpressing tumors were smaller, less vascularized and oxygenated, and less metastatic. Overexpression of miR-378 exerted opposite effects. Accordingly, in patients with NSCLC, HMOX1 expression was lower in metastases to lymph nodes than in primary tumors. INNOVATION AND CONCLUSION In vitro and in vivo data indicate that the interplay between HMOX1 and miR-378 significantly modulates NSCLC progression and angiogenesis, suggesting miR-378 as a new therapeutic target. REBOUND TRACK: This work was rejected during standard peer review and rescued by Rebound Peer Review (Antioxid Redox Signal 16, 293-296, 2012) with the following serving as open reviewers: James F. George, Mahin D. Maines, Justin C. Mason, and Yasufumi Sato.
Collapse
Affiliation(s)
- Klaudia Skrzypek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Wei L, Dai Q, Zhou Y, Zou M, Li Z, Lu N, Guo Q. Oroxylin A sensitizes non-small cell lung cancer cells to anoikis via glucose-deprivation-like mechanisms: c-Src and hexokinase II. Biochim Biophys Acta Gen Subj 2013; 1830:3835-45. [PMID: 23500080 DOI: 10.1016/j.bbagen.2013.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 03/05/2013] [Accepted: 03/06/2013] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cellular metabolism, particularly glycolysis, is altered during the metastatic process and is highly associated with tumor progression and apoptosis resistance. Oroxylin A, a natural plant flavonoid, exhibits chemopreventive and therapeutic anti-inflammatory and anticancer potential. However, the anticancer effects of oroxylin A on non-small cell lung carcinoma (NSCLC) remain poorly understood. METHODS In vitro studies were performed using 2D and 3D conditions. The effects on anoikis-sensitization and glycolysis-inhibition of oroxylin A in human non-small cell lung cancer A549 cells were examined. In vivo murine lung metastasis experiments were utilized to assess the anti-metastatic capacity of oroxylin A. RESULTS ROS-mediated activation of c-Src following detachment caused anoikis resistance in A549 cells. Oroxylin A sensitized A549 cells to anoikis by inactivating the c-Src/AKT/HK II pathway in addition to inducing the dissociation of HK II from mitochondria. Prior to sensitizing A549 cells to anoikis, oroxylin A decreased the ATP level and inhibited glycolysis. Furthermore, oroxylin A inhibited lung metastasis of A549 cells in vivo in nude mice. CONCLUSIONS Oroxylin A sensitized anoikis, which underlies distinct glucose-deprivation-like mechanisms that involved c-Src and HK II. GENERAL SIGNIFICANCE The findings in this study indicated that oroxylin A could potentially be utilized in the development of improved metastatic cancer treatments.
Collapse
Affiliation(s)
- Libin Wei
- China Pharmaceutical University, Nanjing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Wang PY, Ma W, Park JY, Celi FS, Arena R, Choi JW, Ali QA, Tripodi DJ, Zhuang J, Lago CU, Strong LC, Talagala SL, Balaban RS, Kang JG, Hwang PM. Increased oxidative metabolism in the Li-Fraumeni syndrome. N Engl J Med 2013; 368:1027-32. [PMID: 23484829 PMCID: PMC4123210 DOI: 10.1056/nejmoa1214091] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
There is growing evidence that alterations in metabolism may contribute to tumorigenesis. Here, we report on members of families with the Li-Fraumeni syndrome who carry germline mutations in TP53, the gene encoding the tumor-suppressor protein p53. As compared with family members who are not carriers and with healthy volunteers, family members with these mutations have increased oxidative phosphorylation of skeletal muscle. Basic experimental studies of tissue samples from patients with the Li-Fraumeni syndrome and a mouse model of the syndrome support this in vivo finding of increased mitochondrial function. These results suggest that p53 regulates bioenergetic homeostasis in humans. (Funded by the National Heart, Lung, and Blood Institute and the National Institutes of Health; ClinicalTrials.gov number, NCT00406445.).
Collapse
Affiliation(s)
- Ping-Yuan Wang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhuang J, Ma W, Lago CU, Hwang PM. Metabolic regulation of oxygen and redox homeostasis by p53: lessons from evolutionary biology? Free Radic Biol Med 2012; 53:1279-85. [PMID: 22841759 PMCID: PMC3444283 DOI: 10.1016/j.freeradbiomed.2012.07.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 07/20/2012] [Accepted: 07/20/2012] [Indexed: 12/31/2022]
Abstract
The genetic links between p53 and metabolic processes such as oxidative phosphorylation are being studied with increasing interest given that cellular metabolism seems to play an important role in tumorigenesis. This review focuses on how p53 regulation of various metabolic genes may influence redox homeostasis, as the genome is constantly susceptible to oxidative damage, a consequence of living in an aerobic environment. Because p53-like genetic sequences are also found in life forms that may not necessarily benefit from tumor suppression, an evolutionary introduction is given in an attempt to understand why p53 might regulate a basic cellular activity such as metabolism. The presented epidemiologic and experimental data suggest that one reason may be for the homeostatic regulation of oxygen, the essential substrate for reactive oxygen species generation.
Collapse
Affiliation(s)
- Jie Zhuang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
39
|
Antico Arciuch VG, Elguero ME, Poderoso JJ, Carreras MC. Mitochondrial regulation of cell cycle and proliferation. Antioxid Redox Signal 2012; 16:1150-80. [PMID: 21967640 PMCID: PMC3315176 DOI: 10.1089/ars.2011.4085] [Citation(s) in RCA: 317] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 10/03/2011] [Accepted: 10/03/2011] [Indexed: 01/01/2023]
Abstract
Eukaryotic mitochondria resulted from symbiotic incorporation of α-proteobacteria into ancient archaea species. During evolution, mitochondria lost most of the prokaryotic bacterial genes and only conserved a small fraction including those encoding 13 proteins of the respiratory chain. In this process, many functions were transferred to the host cells, but mitochondria gained a central role in the regulation of cell proliferation and apoptosis, and in the modulation of metabolism; accordingly, defective organelles contribute to cell transformation and cancer, diabetes, and neurodegenerative diseases. Most cell and transcriptional effects of mitochondria depend on the modulation of respiratory rate and on the production of hydrogen peroxide released into the cytosol. The mitochondrial oxidative rate has to remain depressed for cell proliferation; even in the presence of O₂, energy is preferentially obtained from increased glycolysis (Warburg effect). In response to stress signals, traffic of pro- and antiapoptotic mitochondrial proteins in the intermembrane space (B-cell lymphoma-extra large, Bcl-2-associated death promoter, Bcl-2 associated X-protein and cytochrome c) is modulated by the redox condition determined by mitochondrial O₂ utilization and mitochondrial nitric oxide metabolism. In this article, we highlight the traffic of the different canonical signaling pathways to mitochondria and the contributions of organelles to redox regulation of kinases. Finally, we analyze the dynamics of the mitochondrial population in cell cycle and apoptosis.
Collapse
Affiliation(s)
| | - María Eugenia Elguero
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
| | - Juan José Poderoso
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- Department of Internal Medicine, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - María Cecilia Carreras
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
- Department of Clinical Biochemistry, INFIBIOC and School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW There is an inverse relationship between cancer incidence and cardiorespiratory fitness in large population studies. Mechanistic insights into these observations may strengthen the rationale for encouraging exercise fitness in the clinics for cancer prevention and may promote the development of new preventive strategies. RECENT FINDINGS Studying the multifaceted activities of p53, a critical tumor suppressor gene, has revealed various cellular pathways necessary for adapting to environmental stresses. Genetic connections are being made between p53 and an increasing number of metabolic activities such as oxidative phosphorylation, glycolysis and fatty acid oxidation. In-vivo mouse models show that p53 plays an important role in determining both basal aerobic exercise capacity and its improvement by training. SUMMARY The genetic pathways by which p53 regulates metabolism and exercise may help explain significant epidemiologic observations connecting cardiorespiratory fitness and cancer. Further understanding of these molecular pathways through human translational studies may promote the development of new cancer preventive strategies.
Collapse
|