1401
|
Yugandhar D, Nayak VL, Archana S, Shekar KC, Srivastava AK. Design, synthesis and anticancer properties of novel oxa/azaspiro[4,5]trienones as potent apoptosis inducers through mitochondrial disruption. Eur J Med Chem 2015; 101:348-57. [DOI: 10.1016/j.ejmech.2015.06.050] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 10/23/2022]
|
1402
|
Mannelli M, Rapizzi E, Fucci R, Canu L, Ercolino T, Luconi M, Young WF. 15 YEARS OF PARAGANGLIOMA: Metabolism and pheochromocytoma/paraganglioma. Endocr Relat Cancer 2015; 22:T83-90. [PMID: 26113605 DOI: 10.1530/erc-15-0215] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2015] [Indexed: 12/14/2022]
Abstract
The discovery of SDHD as a pheochromocytoma/paraganglioma susceptibility gene was the prismatic event that led to all of the subsequent work highlighting the key roles played by mitochondria in the pathogenesis of these tumors and other solid cancers. Alterations in the function of tricarboxylic acid cycle enzymes can cause accumulation of intermediate substrates and subsequent changes in cell metabolism, activation of the angiogenic pathway, increased reactive oxygen species production, DNA hypermethylation, and modification of the tumor microenvironment favoring tumor growth and aggressiveness. The elucidation of these tumorigenic mechanisms should lead to novel therapeutic targets for the treatment of the most aggressive forms of pheochromocytoma/paraganglioma.
Collapse
Affiliation(s)
- Massimo Mannelli
- Endocrinology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Viale Pieraccini 6, 50139 Florence, ItalyEndocrinology UnitCareggi Hospital, Azienda Ospedaliera Universitaria Careggi, Florence, ItalyDivision of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Elena Rapizzi
- Endocrinology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Viale Pieraccini 6, 50139 Florence, ItalyEndocrinology UnitCareggi Hospital, Azienda Ospedaliera Universitaria Careggi, Florence, ItalyDivision of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Rossella Fucci
- Endocrinology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Viale Pieraccini 6, 50139 Florence, ItalyEndocrinology UnitCareggi Hospital, Azienda Ospedaliera Universitaria Careggi, Florence, ItalyDivision of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Letizia Canu
- Endocrinology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Viale Pieraccini 6, 50139 Florence, ItalyEndocrinology UnitCareggi Hospital, Azienda Ospedaliera Universitaria Careggi, Florence, ItalyDivision of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Tonino Ercolino
- Endocrinology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Viale Pieraccini 6, 50139 Florence, ItalyEndocrinology UnitCareggi Hospital, Azienda Ospedaliera Universitaria Careggi, Florence, ItalyDivision of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Michaela Luconi
- Endocrinology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Viale Pieraccini 6, 50139 Florence, ItalyEndocrinology UnitCareggi Hospital, Azienda Ospedaliera Universitaria Careggi, Florence, ItalyDivision of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - William F Young
- Endocrinology UnitDepartment of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Viale Pieraccini 6, 50139 Florence, ItalyEndocrinology UnitCareggi Hospital, Azienda Ospedaliera Universitaria Careggi, Florence, ItalyDivision of EndocrinologyDiabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
1403
|
Berridge MV, Dong L, Neuzil J. Mitochondrial DNA in Tumor Initiation, Progression, and Metastasis: Role of Horizontal mtDNA Transfer. Cancer Res 2015. [PMID: 26224121 DOI: 10.1158/0008-5472.can-15-0859] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mitochondrial DNA (mtDNA), encoding 13 out of more than 1,000 proteins of the mitochondrial proteome, is of paramount importance for the bioenergetic machinery of oxidative phosphorylation that is required for tumor initiation, propagation, and metastasis. In stark contrast to the widely held view that mitochondria and mtDNA are retained and propagated within somatic cells of higher organisms, recent in vitro and in vivo evidence demonstrates that mitochondria move between mammalian cells. This is particularly evident in cancer where defective mitochondrial respiration can be restored and tumor-forming ability regained by mitochondrial acquisition. This paradigm shift in cancer cell biology and mitochondrial genetics, concerning mitochondrial movement between cells to meet bioenergetic needs, not only adds another layer of plasticity to the armory of cancer cells to correct damaged mitochondria, but also points to potentially new therapeutic approaches.
Collapse
Affiliation(s)
- Michael V Berridge
- Cancer Cell and Molecular Biology Group, Malaghan Institute of Medical Research, Wellington, New Zealand.
| | - Lanfeng Dong
- Mitochondria, Apoptosis and Cancer Research Group, School of Medical Science and Griffith Health Institute, Griffith University, Southport, Queensland, Australia
| | - Jiri Neuzil
- Mitochondria, Apoptosis and Cancer Research Group, School of Medical Science and Griffith Health Institute, Griffith University, Southport, Queensland, Australia. Molecular Therapy Group, Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
1404
|
Lymphocyte-specific protein tyrosine kinase (Lck) interacts with CR6-interacting factor 1 (CRIF1) in mitochondria to repress oxidative phosphorylation. BMC Cancer 2015. [PMID: 26210498 PMCID: PMC4515320 DOI: 10.1186/s12885-015-1520-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Many cancer cells exhibit reduced mitochondrial respiration as part of metabolic reprogramming to support tumor growth. Mitochondrial localization of several protein tyrosine kinases is linked to this characteristic metabolic shift in solid tumors, but remains largely unknown in blood cancer. Lymphocyte-specific protein tyrosine kinase (Lck) is a key T-cell kinase and widely implicated in blood malignancies. The purpose of our study is to determine whether and how Lck contributes to metabolic shift in T-cell leukemia through mitochondrial localization. METHODS We compared the human leukemic T-cell line Jurkat with its Lck-deficient derivative Jcam cell line. Differences in mitochondrial respiration were measured by the levels of mitochondrial membrane potential, oxygen consumption, and mitochondrial superoxide. Detailed mitochondrial structure was visualized by transmission electron microscopy. Lck localization was evaluated by subcellular fractionation and confocal microscopy. Proteomic analysis was performed to identify proteins co-precipitated with Lck in leukemic T-cells. Protein interaction was validated by biochemical co-precipitation and confocal microscopy, followed by in situ proximity ligation assay microscopy to confirm close-range (<16 nm) interaction. RESULTS Jurkat cells have abnormal mitochondrial structure and reduced levels of mitochondrial respiration, which is associated with the presence of mitochondrial Lck and lower levels of mitochondrion-encoded electron transport chain proteins. Proteomics identified CR6-interacting factor 1 (CRIF1) as the novel Lck-interacting protein. Lck association with CRIF1 in Jurkat mitochondria was confirmed biochemically and by microscopy, but did not lead to CRIF1 tyrosine phosphorylation. Consistent with the role of CRIF1 in functional mitoribosome, shRNA-mediated silencing of CRIF1 in Jcam resulted in mitochondrial dysfunction similar to that observed in Jurkat. Reduced interaction between CRIF1 and Tid1, another key component of intramitochondrial translational machinery, in Jurkat further supports the role of mitochondrial Lck as a negative regulator of CRIF1 through competitive binding. CONCLUSIONS This is the first report demonstrating the role of mitochondrial Lck in metabolic reprogramming of leukemic cells. Mechanistically, it is distinct from other reported mitochondrial protein tyrosine kinases. In a kinase-independent manner, mitochondrial Lck interferes with mitochondrial translational machinery through competitive binding to CRIF1. These findings may reveal novel approaches in cancer therapy by targeting cancer cell metabolism.
Collapse
|
1405
|
Hektoen HH, Flatmark K, Andersson Y, Dueland S, Redalen KR, Ree AH. Early increase in circulating carbonic anhydrase IX during neoadjuvant treatment predicts favourable outcome in locally advanced rectal cancer. BMC Cancer 2015. [PMID: 26205955 PMCID: PMC4513373 DOI: 10.1186/s12885-015-1557-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background Locally advanced rectal cancer (LARC) comprises heterogeneous tumours with predominant hypoxic components. The hypoxia-inducible metabolic shift causes microenvironmental acidification generated by carbonic anhydrase IX (CAIX) and facilitates metastatic progression, the dominant cause of failure in LARC. Methods Using a commercially available immunoassay, circulating CAIX was assessed in prospectively archived serial serum samples collected during combined-modality neoadjuvant treatment of LARC patients and correlated to histologic tumour response and progression-free survival (PFS). Results Patients who from their individual baseline level displayed serum CAIX increase above a threshold of 224 pg/ml (with 96 % specificity and 39 % sensitivity) after completion of short-course neoadjuvant chemotherapy (NACT) prior to long-course chemoradiotherapy and definitive surgery had significantly better 5-year PFS (94 %) than patients with below-threshold post-NACT versus baseline alteration (PFS rate of 56 %; p < 0.01). This particular CAIX parameter, ΔNACT, was significantly correlated with histologic ypT0–2 and ypN0 outcome (p < 0.01) and remained an independent PFS predictor in multivariate analysis wherein it was entered as continuous variable (p = 0.04). Conclusions Our results indicate that low ΔNACT, i.e., a weak increase in serum CAIX level following initial neoadjuvant treatment (in this case two cycles of the Nordic FLOX regimen), might be used as risk-adapted stratification to postoperative therapy or other modes of intensification of the combined-modality protocol in LARC. Trial registration ClinicalTrials.gov NCT00278694 Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1557-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helga Helseth Hektoen
- Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, Blindern, 0318, Oslo, Norway. .,Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway. .,Department of Tumour Biology, Oslo University Hospital - Norwegian Radium Hospital, P.O.Box 4950, Nydalen, 0424, Oslo, Norway.
| | - Kjersti Flatmark
- Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, Blindern, 0318, Oslo, Norway. .,Department of Tumour Biology, Oslo University Hospital - Norwegian Radium Hospital, P.O.Box 4950, Nydalen, 0424, Oslo, Norway. .,Department of Gastroenterological Surgery, Oslo University Hospital - Norwegian Radium Hospital, P.O.Box 4950, Nydalen, 0424, Oslo, Norway.
| | - Yvonne Andersson
- Department of Tumour Biology, Oslo University Hospital - Norwegian Radium Hospital, P.O.Box 4950, Nydalen, 0424, Oslo, Norway.
| | - Svein Dueland
- Department of Oncology, Oslo University Hospital - Norwegian Radium Hospital, P.O.Box 4950, Nydalen, 0424, Oslo, Norway.
| | - Kathrine Røe Redalen
- Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway.
| | - Anne Hansen Ree
- Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, Blindern, 0318, Oslo, Norway. .,Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway.
| |
Collapse
|
1406
|
Abstract
Cancer is a general name for more than 100 malignant diseases. It is postulated that all cancers start from a single abnormal cell that grows out of control. Untreated cancers can cause serious consequences and deaths. Great progress has been made in cancer research that has significantly improved our knowledge and understanding of the nature and mechanisms of the disease, but the origins of cancer are far from being well understood due to the limitations of suitable model systems and to the complexities of the disease. In view of the fact that cancers are found in various species of vertebrates and other metazoa, here, we suggest that cancer also occurs in parasitic protozoans such as Trypanosoma brucei, a blood parasite, and Toxoplasma gondii, an obligate intracellular pathogen. Without treatment, these protozoan cancers may cause severe disease and death in mammals, including humans. The simpler genomes of these single-cell organisms, in combination with their complex life cycles and fascinating life cycle differentiation processes, may help us to better understand the origins of cancers and, in particular, leukemias.
Collapse
|
1407
|
Phan L, Chou PC, Velazquez-Torres G, Samudio I, Parreno K, Huang Y, Tseng C, Vu T, Gully C, Su CH, Wang E, Chen J, Choi HH, Fuentes-Mattei E, Shin JH, Shiang C, Grabiner B, Blonska M, Skerl S, Shao Y, Cody D, Delacerda J, Kingsley C, Webb D, Carlock C, Zhou Z, Hsieh YC, Lee J, Elliott A, Ramirez M, Bankson J, Hazle J, Wang Y, Li L, Weng S, Rizk N, Wen YY, Lin X, Wang H, Wang H, Zhang A, Xia X, Wu Y, Habra M, Yang W, Pusztai L, Yeung SC, Lee MH. The cell cycle regulator 14-3-3σ opposes and reverses cancer metabolic reprogramming. Nat Commun 2015; 6:7530. [PMID: 26179207 PMCID: PMC4507299 DOI: 10.1038/ncomms8530] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/18/2015] [Indexed: 12/16/2022] Open
Abstract
Extensive reprogramming of cellular energy metabolism is a hallmark of cancer. Despite its importance, the molecular mechanism controlling this tumour metabolic shift remains not fully understood. Here we show that 14-3-3σ regulates cancer metabolic reprogramming and protects cells from tumorigenic transformation. 14-3-3σ opposes tumour-promoting metabolic programmes by enhancing c-Myc poly-ubiquitination and subsequent degradation. 14-3-3σ demonstrates the suppressive impact on cancer glycolysis, glutaminolysis, mitochondrial biogenesis and other major metabolic processes of tumours. Importantly, 14-3-3σ expression levels predict overall and recurrence-free survival rates, tumour glucose uptake and metabolic gene expression in breast cancer patients. Thus, these results highlight that 14-3-3σ is an important regulator of tumour metabolism, and loss of 14-3-3σ expression is critical for cancer metabolic reprogramming. We anticipate that pharmacologically elevating the function of 14-3-3σ in tumours could be a promising direction for targeted anticancer metabolism therapy development in future.
Collapse
Affiliation(s)
- Liem Phan
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Ping-Chieh Chou
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Guermarie Velazquez-Torres
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Ismael Samudio
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenneth Parreno
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yaling Huang
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Chieh Tseng
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Thuy Vu
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Chris Gully
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Chun-Hui Su
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Edward Wang
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Jian Chen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hyun-Ho Choi
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Enrique Fuentes-Mattei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ji-Hyun Shin
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Christine Shiang
- 1] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA. [2] Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Brian Grabiner
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Marzenna Blonska
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephen Skerl
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yiping Shao
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dianna Cody
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jorge Delacerda
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Charles Kingsley
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Douglas Webb
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Colin Carlock
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| | - Zhongguo Zhou
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yun-Chih Hsieh
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jaehyuk Lee
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Elliott
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marc Ramirez
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jim Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John Hazle
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yongxing Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lei Li
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaofan Weng
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nibal Rizk
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Ye Wen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xin Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hua Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aijun Zhang
- Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Xuefeng Xia
- Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Yun Wu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mouhammed Habra
- Department of Endocrinology Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Yang
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lajos Pusztai
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sai-Ching Yeung
- 1] Department of Endocrinology Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mong-Hong Lee
- 1] Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. [2] Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA
| |
Collapse
|
1408
|
Kontro H, Cannino G, Rustin P, Dufour E, Kainulainen H. DAPIT Over-Expression Modulates Glucose Metabolism and Cell Behaviour in HEK293T Cells. PLoS One 2015; 10:e0131990. [PMID: 26161955 PMCID: PMC4498893 DOI: 10.1371/journal.pone.0131990] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 06/09/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Diabetes Associated Protein in Insulin-sensitive Tissues (DAPIT) is a subunit of mitochondrial ATP synthase and has also been found to associate with the vacuolar H+-ATPase. Its expression is particularly high in cells with elevated aerobic metabolism and in epithelial cells that actively transport nutrients and ions. Deletion of DAPIT is known to induce loss of mitochondrial ATP synthase but the effects of its over-expression are obscure. RESULTS In order to study the consequences of high expression of DAPIT, we constructed a transgenic cell line that constitutively expressed DAPIT in human embryonal kidney cells, HEK293T. Enhanced DAPIT expression decreased mtDNA content and mitochondrial mass, and saturated respiratory chain by decreasing H+-ATP synthase activity. DAPIT over-expression also increased mitochondrial membrane potential and superoxide level, and translocated the transcription factors hypoxia inducible factor 1α (Hif1α) and β-catenin to the nucleus. Accordingly, cells over-expressing DAPIT used more glucose and generated a larger amount of lactate compared to control cells. Interestingly, these changes were associated with an epithelial to mesenchymal (EMT)-like transition by changing E-cadherin to N-cadherin and up-regulating several key junction/adhesion proteins. At physiological level, DAPIT over-expression slowed down cell growth by G1 arrest and migration, and enhanced cell detachment. Several cancers also showed an increase in genomic copy number of Usmg5 (gene encoding DAPIT), thereby providing strong correlative evidence for DAPIT possibly having oncogenic function in cancers. CONCLUSIONS DAPIT over-expression thus appears to modulate mitochondrial functions and alter cellular regulations, promote anaerobic metabolism and induce EMT-like transition. We propose that DAPIT over-expression couples the changes in mitochondrial metabolism to physiological and pathophysiological regulations, and suggest it could play a critical role in H+-ATP synthase dysfunctions.
Collapse
Affiliation(s)
- Heidi Kontro
- Tampere Centre for Child Health Research, University of Tampere, Tampere, Finland
| | - Giuseppe Cannino
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
| | - Pierre Rustin
- INSERM UMR 1141, Paris, France; Université Paris 7, Paris, France
| | - Eric Dufour
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
| | - Heikki Kainulainen
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
1409
|
McKenzie M, Chiotis M, Hroudová J, Lopez Sanchez MIG, Lim SC, Cook MJ, McKelvie P, Cotton RGH, Murphy M, St John JC, Trounce IA. Capture of somatic mtDNA point mutations with severe effects on oxidative phosphorylation in synaptosome cybrid clones from human brain. Hum Mutat 2015; 35:1476-84. [PMID: 25219341 DOI: 10.1002/humu.22694] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 09/03/2014] [Indexed: 01/13/2023]
Abstract
Mitochondrial DNA (mtDNA) is replicated throughout life in postmitotic cells, resulting in higher levels of somatic mutation than in nuclear genes. However, controversy remains as to the importance of low-level mtDNA somatic mutants in cancerous and normal human tissues. To capture somatic mtDNA mutations for functional analysis, we generated synaptosome cybrids from synaptic endings isolated from fresh hippocampus and cortex brain biopsies. We analyzed the whole mtDNA genome from 120 cybrid clones derived from four individual donors by chemical cleavage of mismatch and Sanger sequencing, scanning around two million base pairs. Seventeen different somatic point mutations were identified, including eight coding region mutations, four of which result in frameshifts. Examination of one cybrid clone with a novel m.2949_2953delCTATT mutation in MT-RNR2 (which encodes mitochondrial 16S rRNA) revealed a severe disruption of mtDNA-encoded protein translation. We also performed functional studies on a homoplasmic nonsense mutation in MT-ND1, previously reported in oncocytomas, and show that both ATP generation and the stability of oxidative phosphorylation complex I are disrupted. As the mtDNA remains locked against direct genetic manipulation, we demonstrate that the synaptosome cybrid approach can capture biologically relevant mtDNA mutants in vitro to study effects on mitochondrial respiratory chain function.
Collapse
Affiliation(s)
- Matthew McKenzie
- Centre for Genetic Diseases, MIMR-PHI Institute of Medical Research, Monash University, Clayton, Victoria, 3168, Australia; Monash University, Clayton, Victoria, 3168, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1410
|
Srinivasan S, Guha M, Dong DW, Whelan KA, Ruthel G, Uchikado Y, Natsugoe S, Nakagawa H, Avadhani NG. Disruption of cytochrome c oxidase function induces the Warburg effect and metabolic reprogramming. Oncogene 2015; 35:1585-95. [PMID: 26148236 PMCID: PMC4703574 DOI: 10.1038/onc.2015.227] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 04/30/2015] [Accepted: 05/10/2015] [Indexed: 02/01/2023]
Abstract
Defects in mitochondrial oxidative phosphorylation complexes, altered bioenergetics and metabolic shift are often seen in cancers. Here we show a role for the dysfunction of electron transport chain component, cytochrome c oxidase (CcO) in cancer progression. We show that genetic silencing of the CcO complex by shRNA expression and loss of CcO activity in multiple cell types from the mouse and human sources resulted in metabolic shift to glycolysis, loss of anchorage dependent growth and acquired invasive phenotypes. Disruption of CcO complex caused loss of transmembrane potential and induction of Ca2+/Calcineurin-mediated retrograde signaling. Propagation of this signaling, includes activation of PI3-kinase, IGF1R and Akt, Ca2+ sensitive transcription factors and also, TGFβ1, MMP16, periostin that are involved in oncogenic progression. Whole genome expression analysis showed up regulation of genes involved in cell signaling, extracellular matrix interactions, cell morphogenesis, cell motility and migration. The transcription profiles reveal extensive similarity to retrograde signaling initiated by partial mtDNA depletion, though distinct differences are observed in signaling induced by CcO dysfunction. The possible CcO dysfunction as a biomarker for cancer progression was supported by data showing that esophageal tumors from human patients show reduced CcO subunits IVi1 and Vb in regions that were previously shown to be hypoxic core of the tumors. Our results show that mitochondrial electron transport chain defect initiates a retrograde signaling. These results suggest that a defect in CcO complex can potentially induce tumor progression.
Collapse
Affiliation(s)
- S Srinivasan
- Department of Biomedical Sciences, The Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, Philadelphia, PA, USA
| | - M Guha
- Department of Biomedical Sciences, The Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, Philadelphia, PA, USA
| | - D W Dong
- Department of Biomedical Sciences, The Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, Philadelphia, PA, USA
| | - K A Whelan
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - G Ruthel
- Department of Biomedical Sciences, The Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, Philadelphia, PA, USA
| | - Y Uchikado
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medicine, Kagoshima University, Kagoshima, Japan
| | - S Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medicine, Kagoshima University, Kagoshima, Japan
| | - H Nakagawa
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - N G Avadhani
- Department of Biomedical Sciences, The Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, Philadelphia, PA, USA
| |
Collapse
|
1411
|
PI3K therapy reprograms mitochondrial trafficking to fuel tumor cell invasion. Proc Natl Acad Sci U S A 2015; 112:8638-43. [PMID: 26124089 DOI: 10.1073/pnas.1500722112] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Molecular therapies are hallmarks of "personalized" medicine, but how tumors adapt to these agents is not well-understood. Here we show that small-molecule inhibitors of phosphatidylinositol 3-kinase (PI3K) currently in the clinic induce global transcriptional reprogramming in tumors, with activation of growth factor receptors, (re)phosphorylation of Akt and mammalian target of rapamycin (mTOR), and increased tumor cell motility and invasion. This response involves redistribution of energetically active mitochondria to the cortical cytoskeleton, where they support membrane dynamics, turnover of focal adhesion complexes, and random cell motility. Blocking oxidative phosphorylation prevents adaptive mitochondrial trafficking, impairs membrane dynamics, and suppresses tumor cell invasion. Therefore, "spatiotemporal" mitochondrial respiration adaptively induced by PI3K therapy fuels tumor cell invasion, and may provide an important antimetastatic target.
Collapse
|
1412
|
Alternative approaches to Hsp90 modulation for the treatment of cancer. Future Med Chem 2015; 6:1587-605. [PMID: 25367392 DOI: 10.4155/fmc.14.89] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hsp90 is responsible for the conformational maturation of newly synthesized polypeptides (client proteins) and the re-maturation of denatured proteins via the Hsp90 chaperone cycle. Inhibition of the Hsp90 N-terminus has emerged as a clinically relevant strategy for anticancer chemotherapeutics due to the involvement of clients in a variety of oncogenic pathways. Several immunophilins, co-chaperones and partner proteins are also necessary for Hsp90 chaperoning activity. Alternative strategies to inhibit Hsp90 function include disruption of the C-terminal dimerization domain and the Hsp90 heteroprotein complex. C-terminal inhibitors and Hsp90 co-chaperone disruptors prevent cancer cell proliferation similar to N-terminal inhibitors and destabilize client proteins without induction of heat shock proteins. Herein, current Hsp90 inhibitors, the chaperone cycle, and regulation of this cycle will be discussed.
Collapse
|
1413
|
RSC Chromatin-Remodeling Complex Is Important for Mitochondrial Function in Saccharomyces cerevisiae. PLoS One 2015; 10:e0130397. [PMID: 26086550 PMCID: PMC4472808 DOI: 10.1371/journal.pone.0130397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/19/2015] [Indexed: 11/19/2022] Open
Abstract
RSC (Remodel the Structure of Chromatin) is an ATP-dependent chromatin remodeling complex essential for the growth of Saccharomyces cerevisiae. RSC exists as two distinct isoforms that share core subunits including the ATPase subunit Nps1/Sth1 but contain either Rsc1or Rsc2. Using the synthetic genetic array (SGA) of the non-essential null mutation method, we screened for mutations exhibiting synthetic growth defects in combination with the temperature-sensitive mutant, nps1-105, and found connections between mitochondrial function and RSC. rsc mutants, including rsc1Δ, rsc2Δ, and nps1-13, another temperature-sensitive nps1 mutant, exhibited defective respiratory growth; in addition, rsc2Δ and nps1-13 contained aggregated mitochondria. The rsc2Δ phenotypes were relieved by RSC1 overexpression, indicating that the isoforms play a redundant role in respiratory growth. Genome-wide expression analysis in nps1-13 under respiratory conditions suggested that RSC regulates the transcription of some target genes of the HAP complex, a transcriptional activator of respiratory gene expression. Nps1 physically interacted with Hap4, the transcriptional activator moiety of the HAP complex, and overexpression of HAP4 alleviated respiratory defects in nps1-13, suggesting that RSC plays pivotal roles in mitochondrial gene expression and shares a set of target genes with the HAP complex.
Collapse
|
1414
|
Chen C, Chen Y, Guan MX. A peep into mitochondrial disorder: multifaceted from mitochondrial DNA mutations to nuclear gene modulation. Protein Cell 2015; 6:862-70. [PMID: 26084519 PMCID: PMC4656216 DOI: 10.1007/s13238-015-0175-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/17/2015] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial genome is responsible for multiple human diseases in a maternal inherited pattern, yet phenotypes of patients in a same pedigree frequently vary largely. Genes involving in epigenetic modification, RNA processing, and other biological pathways, rather than “threshold effect” and environmental factors, provide more specific explanation to the aberrant phenotype. Thus, the double hit theory, mutations both in mitochondrial DNA and modifying genes aggravating the symptom, throws new light on mitochondrial dysfunction processes. In addition, mitochondrial retrograde signaling pathway that leads to reconfiguration of cell metabolism to adapt defects in mitochondria may as well play an active role. Here we review selected examples of modifier genes and mitochondrial retrograde signaling in mitochondrial disorders, which refine our understanding and will guide the rational design of clinical therapies.
Collapse
Affiliation(s)
- Chao Chen
- School of Medicine, Institute of Genetics, Zhejiang University, Hangzhou, 310058, China
| | - Ye Chen
- School of Medicine, Institute of Genetics, Zhejiang University, Hangzhou, 310058, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310058, China.
| | - Min-Xin Guan
- School of Medicine, Institute of Genetics, Zhejiang University, Hangzhou, 310058, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
1415
|
Truitt ML, Conn CS, Shi Z, Pang X, Tokuyasu T, Coady AM, Seo Y, Barna M, Ruggero D. Differential Requirements for eIF4E Dose in Normal Development and Cancer. Cell 2015; 162:59-71. [PMID: 26095252 DOI: 10.1016/j.cell.2015.05.049] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/19/2015] [Accepted: 04/28/2015] [Indexed: 12/18/2022]
Abstract
eIF4E, the major cap-binding protein, has long been considered limiting for translating the mammalian genome. However, the eIF4E dose requirement at an organismal level remains unexplored. By generating an Eif4e haploinsufficient mouse, we found that a 50% reduction in eIF4E expression, while compatible with normal development and global protein synthesis, significantly impeded cellular transformation. Genome-wide translational profiling uncovered a translational program induced by oncogenic transformation and revealed a critical role for the dose of eIF4E, specifically in translating a network of mRNAs enriched for a unique 5' UTR signature. In particular, we demonstrate that the dose of eIF4E is essential for translating mRNAs that regulate reactive oxygen species, fueling transformation and cancer cell survival in vivo. Our findings indicate eIF4E is maintained at levels in excess for normal development that are hijacked by cancer cells to drive a translational program supporting tumorigenesis.
Collapse
Affiliation(s)
- Morgan L Truitt
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Crystal S Conn
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Zhen Shi
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Xiaming Pang
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Taku Tokuyasu
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alison M Coady
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Youngho Seo
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Maria Barna
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA.
| | - Davide Ruggero
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
1416
|
Tower J. Mitochondrial maintenance failure in aging and role of sexual dimorphism. Arch Biochem Biophys 2015; 576:17-31. [PMID: 25447815 PMCID: PMC4409928 DOI: 10.1016/j.abb.2014.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/08/2014] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Abstract
Gene expression changes during aging are partly conserved across species, and suggest that oxidative stress, inflammation and proteotoxicity result from mitochondrial malfunction and abnormal mitochondrial-nuclear signaling. Mitochondrial maintenance failure may result from trade-offs between mitochondrial turnover versus growth and reproduction, sexual antagonistic pleiotropy and genetic conflicts resulting from uni-parental mitochondrial transmission, as well as mitochondrial and nuclear mutations and loss of epigenetic regulation. Aging phenotypes and interventions are often sex-specific, indicating that both male and female sexual differentiation promote mitochondrial failure and aging. Studies in mammals and invertebrates implicate autophagy, apoptosis, AKT, PARP, p53 and FOXO in mediating sex-specific differences in stress resistance and aging. The data support a model where the genes Sxl in Drosophila, sdc-2 in Caenorhabditis elegans, and Xist in mammals regulate mitochondrial maintenance across generations and in aging. Several interventions that increase life span cause a mitochondrial unfolded protein response (UPRmt), and UPRmt is also observed during normal aging, indicating hormesis. The UPRmt may increase life span by stimulating mitochondrial turnover through autophagy, and/or by inhibiting the production of hormones and toxic metabolites. The data suggest that metazoan life span interventions may act through a common hormesis mechanism involving liver UPRmt, mitochondrial maintenance and sexual differentiation.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2910, United States.
| |
Collapse
|
1417
|
Pietrocola F, Galluzzi L, Bravo-San Pedro JM, Madeo F, Kroemer G. Acetyl coenzyme A: a central metabolite and second messenger. Cell Metab 2015; 21:805-21. [PMID: 26039447 DOI: 10.1016/j.cmet.2015.05.014] [Citation(s) in RCA: 899] [Impact Index Per Article: 99.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acetyl-coenzyme A (acetyl-CoA) is a central metabolic intermediate. The abundance of acetyl-CoA in distinct subcellular compartments reflects the general energetic state of the cell. Moreover, acetyl-CoA concentrations influence the activity or specificity of multiple enzymes, either in an allosteric manner or by altering substrate availability. Finally, by influencing the acetylation profile of several proteins, including histones, acetyl-CoA controls key cellular processes, including energy metabolism, mitosis, and autophagy, both directly and via the epigenetic regulation of gene expression. Thus, acetyl-CoA determines the balance between cellular catabolism and anabolism by simultaneously operating as a metabolic intermediate and as a second messenger.
Collapse
Affiliation(s)
- Federico Pietrocola
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - José Manuel Bravo-San Pedro
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria.
| | - Guido Kroemer
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.
| |
Collapse
|
1418
|
Hierarchical targeted hepatocyte mitochondrial multifunctional chitosan nanoparticles for anticancer drug delivery. Biomaterials 2015; 52:240-50. [DOI: 10.1016/j.biomaterials.2015.02.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 01/27/2015] [Accepted: 02/01/2015] [Indexed: 02/06/2023]
|
1419
|
Li BB, Jia YX, Zhu PC, Chew RJ, Li Y, Tan NS, Leung PH. Highly selective anti-cancer properties of ester functionalized enantiopure dinuclear gold(I)-diphosphine. Eur J Med Chem 2015; 98:250-5. [DOI: 10.1016/j.ejmech.2015.05.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/15/2015] [Accepted: 05/18/2015] [Indexed: 01/22/2023]
|
1420
|
Cunniff B, Newick K, Nelson KJ, Wozniak AN, Beuschel S, Leavitt B, Bhave A, Butnor K, Koenig A, Chouchani ET, James AM, Haynes AC, Lowther WT, Murphy MP, Shukla A, Heintz NH. Disabling Mitochondrial Peroxide Metabolism via Combinatorial Targeting of Peroxiredoxin 3 as an Effective Therapeutic Approach for Malignant Mesothelioma. PLoS One 2015; 10:e0127310. [PMID: 26011724 PMCID: PMC4444329 DOI: 10.1371/journal.pone.0127310] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 04/14/2015] [Indexed: 01/01/2023] Open
Abstract
Dysregulation of signaling pathways and energy metabolism in cancer cells enhances production of mitochondrial hydrogen peroxide that supports tumorigenesis through multiple mechanisms. To counteract the adverse effects of mitochondrial peroxide many solid tumor types up-regulate the mitochondrial thioredoxin reductase 2 - thioredoxin 2 (TRX2) - peroxiredoxin 3 (PRX3) antioxidant network. Using malignant mesothelioma cells as a model, we show that thiostrepton (TS) irreversibly disables PRX3 via covalent crosslinking of peroxidatic and resolving cysteine residues in homodimers, and that targeting the oxidoreductase TRX2 with the triphenylmethane gentian violet (GV) potentiates adduction by increasing levels of disulfide-bonded PRX3 dimers. Due to the fact that activity of the PRX3 catalytic cycle dictates the rate of adduction by TS, immortalized and primary human mesothelial cells are significantly less sensitive to both compounds. Moreover, stable knockdown of PRX3 reduces mesothelioma cell proliferation and sensitivity to TS. Expression of catalase in shPRX3 mesothelioma cells restores defects in cell proliferation but not sensitivity to TS. In a SCID mouse xenograft model of human mesothelioma, administration of TS and GV together reduced tumor burden more effectively than either agent alone. Because increased production of mitochondrial hydrogen peroxide is a common phenotype of malignant cells, and TS and GV are well tolerated in mammals, we propose that targeting PRX3 is a feasible redox-dependent strategy for managing mesothelioma and other intractable human malignancies.
Collapse
Affiliation(s)
- Brian Cunniff
- University of Vermont, College of Medicine, Department of Pathology, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
- * E-mail:
| | - Kheng Newick
- University of Pennsylvania School of Medicine, Division of Pulmonary, Thoracic Oncology Research Laboratory, Philadelphia, PA, 19147, United States of America
| | - Kimberly J. Nelson
- Wake Forest School of Medicine, Department of Biochemistry, Medical Center Boulevard, Winston-Salem, NC, 27157, United States of America
| | - Alexandra N. Wozniak
- University of Vermont, College of Medicine, Department of Pathology, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| | - Stacie Beuschel
- University of Vermont, College of Medicine, Department of Pathology, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| | - Bruce Leavitt
- University of Vermont, College of Medicine, Department of Surgery, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| | - Anant Bhave
- University of Vermont, College of Medicine, Department of Radiology, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| | - Kelly Butnor
- University of Vermont, College of Medicine, Department of Pathology, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| | - Andreas Koenig
- University of Vermont, Department of Immunology medicine, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| | - Edward T. Chouchani
- Medical Research Council, Mitochondrial Biology Unit, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 2QQ, United Kingdom
| | - Andrew M. James
- Medical Research Council, Mitochondrial Biology Unit, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Alexina C. Haynes
- Wake Forest School of Medicine, Department of Biochemistry, Medical Center Boulevard, Winston-Salem, NC, 27157, United States of America
| | - W. Todd Lowther
- Wake Forest School of Medicine, Department of Biochemistry, Medical Center Boulevard, Winston-Salem, NC, 27157, United States of America
| | - Michael P. Murphy
- Medical Research Council, Mitochondrial Biology Unit, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Arti Shukla
- University of Vermont, College of Medicine, Department of Pathology, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| | - Nicholas H. Heintz
- University of Vermont, College of Medicine, Department of Pathology, 149 Beaumont Ave, Burlington, VT, 05405, United States of America
| |
Collapse
|
1421
|
Kurelac I, de Biase D, Calabrese C, Ceccarelli C, Ng CKY, Lim R, MacKay A, Weigelt B, Porcelli AM, Reis-Filho JS, Tallini G, Gasparre G. High-resolution genomic profiling of thyroid lesions uncovers preferential copy number gains affecting mitochondrial biogenesis loci in the oncocytic variants. Am J Cancer Res 2015; 5:1954-1971. [PMID: 26269756 PMCID: PMC4529616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/10/2015] [Indexed: 06/04/2023] Open
Abstract
Oncocytic change is the result of aberrant mitochondrial hyperplasia, which may occur in both neoplastic and non-neoplastic cells and is not infrequent in the thyroid. Despite being a well-characterized histologic phenotype, the molecular causes underlying such a distinctive cellular change are poorly understood. To identify potential genetic causes for the oncocytic phenotype in thyroid, we analyzed copy number alterations in a set of oncocytic (n=21) and non-oncocytic (n=20) thyroid lesions by high-resolution microarray-based comparative genomic hybridization (aCGH). Each group comprised lesions of diverse histologic types, including hyperplastic nodules, adenomas and carcinomas. Unsupervised hierarchical clustering of categorical aCGH data resulted in two distinct branches, one of which was significantly enriched for samples with the oncocytic phenotype, regardless of histologic type. Analysis of aCGH events showed that the oncocytic group harbored a significantly higher number of genes involved in copy number gains, when compared to that of conventional thyroid lesions. Functional annotation demonstrated an enrichment for copy number gains that affect genes encoding activators of mitochondrial biogenesis in oncocytic cases but not in their non-oncocytic counterparts. Taken together, our data suggest that genomic alterations may represent additional/alternative mechanisms underlying the development of the oncocytic phenotype in the thyroid.
Collapse
Affiliation(s)
- Ivana Kurelac
- Medical Genetics Unit, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, S. Orsola-Malpighi HospitalBologna, Italy
| | - Dario de Biase
- Department of Clinical, Diagnostic and Experimental Medicine (DIMES), University of Bologna, Section of Anatomic Pathology at Bellaria HospitalBologna, Italy
| | - Claudia Calabrese
- Medical Genetics Unit, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, S. Orsola-Malpighi HospitalBologna, Italy
| | - Claudio Ceccarelli
- Department of Clinical, Diagnostic and Experimental Medicine (DIMES), University of Bologna, S. Orsola-Malpighi HospitalBologna, Italy
| | - Charlotte KY Ng
- Department of Pathology, Memorial Sloan Kettering Cancer CenterNew York, NY, USA
| | - Raymond Lim
- Department of Pathology, Memorial Sloan Kettering Cancer CenterNew York, NY, USA
| | - Alan MacKay
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer ResearchLondon, UK
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer CenterNew York, NY, USA
| | - Anna Maria Porcelli
- Department of Farmacy and Biotechnology (FABIT), University of BolognaBologna, Italy
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer CenterNew York, NY, USA
| | - Giovanni Tallini
- Department of Clinical, Diagnostic and Experimental Medicine (DIMES), University of Bologna, Section of Anatomic Pathology at Bellaria HospitalBologna, Italy
| | - Giuseppe Gasparre
- Medical Genetics Unit, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, S. Orsola-Malpighi HospitalBologna, Italy
| |
Collapse
|
1422
|
Yin K. Positive correlation between expression level of mitochondrial serine hydroxymethyltransferase and breast cancer grade. Onco Targets Ther 2015; 8:1069-74. [PMID: 25999742 PMCID: PMC4437609 DOI: 10.2147/ott.s82433] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Metabolic reprogramming plays an essential role in supporting the survival and proliferation of cancer cells. Serine hydroxymethyltransferase (SHMT) directs serine to the metabolism of one-carbon unit and the synthesis of thymidilate as a key factor in this metabolic shift. Although the mitochondrial isoform of SHMT (SHMT2) has been proven to be a crucial factor in the serine/glycine metabolism in several cancer cell types, the expression pattern of SHMT2 and the correlation of expression level of SHMT2 and other clinicopathological parameters in clinical breast cancer remain to be explored. In this research, 76 breast cancer patients who underwent modified radical mastectomy were enrolled for immunohistochemical analysis of the expression level of SHMT2 in their cancerous breast tissues for comparison with that in matching, distant noncancerous tissues. The results showed that SHMT2 was not expressed in the distant noncancerous cells. In contrast, SHMT2 protein could be stained in all breast cancer samples at varying degrees. Higher level of SHMT2 was expressed in grade III breast cancer cells than that those in grade I–II (P<0.05). In conclusion, SHMT2 was highly expressed in breast cancer cells, and the expression level of SHMT2 was positively correlated with breast cancer grade, suggesting that SHMT2 could be a target for anticancer therapies.
Collapse
Affiliation(s)
- Ke Yin
- Department of Thyroid and Breast Surgery, Ningbo First Hospital, Ningbo, People's Republic of China
| |
Collapse
|
1423
|
Sun N, Johnson J, Stack MS, Szajko J, Sander C, Rebuyon R, Deatsch A, Easton J, Tanner CE, Ruggiero ST. Nanoparticle analysis of cancer cells by light transmission spectroscopy. Anal Biochem 2015; 484:58-65. [PMID: 25981981 DOI: 10.1016/j.ab.2015.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 04/21/2015] [Accepted: 05/05/2015] [Indexed: 01/21/2023]
Abstract
We have measured the optical properties of cancer and normal whole cells and lysates using light transmission spectroscopy (LTS). LTS provides both the optical extinction coefficient in the wavelength range from 220 to 1100nm and (by spectral inversion using a Mie model) the particle distribution density in the size range from 1 to 3000nm. Our current work involves whole cells and lysates of cultured human oral cells in liquid suspension. We found systematic differences in the optical extinction between cancer and normal whole cells and lysates, which translate to different particle size distributions (PSDs) for these materials. Specifically, we found that cancer cells have distinctly lower concentrations of nanoparticles with diameters less than 100nm and have higher concentrations of particles with diameters from 100 to 1000nm-results that hold for both whole cells and lysates. We also found a power-law dependence of particle density with diameter over several orders of magnitude.
Collapse
Affiliation(s)
- N Sun
- Department of Physics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - J Johnson
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - M S Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - J Szajko
- F Cubed, South Bend, IN 46617, USA
| | - C Sander
- Department of Physics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - R Rebuyon
- Department of Physics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - A Deatsch
- Department of Physics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - J Easton
- Ivy Tech, South Bend, IN 46601, USA
| | - C E Tanner
- Department of Physics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - S T Ruggiero
- Department of Physics, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
1424
|
Abstract
Recent advances in mitochondrial biology have revealed the high diversity and complexity of proteolytic enzymes that regulate mitochondrial function. We have classified mitochondrial proteases, or mitoproteases, on the basis of their function and location, and defined the human mitochondrial degradome as the complete set of mitoproteases that are encoded by the human genome. In addition to their nonspecific degradative functions, mitoproteases perform highly regulated proteolytic reactions that are important in mitochondrial function, integrity and homeostasis. These include protein synthesis, quality control, mitochondrial biogenesis and dynamics, mitophagy and apoptosis. Impaired or dysregulated function of mitoproteases is associated with ageing and with many pathological conditions such as neurodegenerative disorders, metabolic syndromes and cancer. A better understanding of the mitochondrial proteolytic landscape and its modulation may contribute to improving human lifespan and 'healthspan'.
Collapse
|
1425
|
MYC oncogene overexpression drives renal cell carcinoma in a mouse model through glutamine metabolism. Proc Natl Acad Sci U S A 2015; 112:6539-44. [PMID: 25964345 DOI: 10.1073/pnas.1507228112] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The MYC oncogene is frequently mutated and overexpressed in human renal cell carcinoma (RCC). However, there have been no studies on the causative role of MYC or any other oncogene in the initiation or maintenance of kidney tumorigenesis. Here, we show through a conditional transgenic mouse model that the MYC oncogene, but not the RAS oncogene, initiates and maintains RCC. Desorption electrospray ionization-mass-spectrometric imaging was used to obtain chemical maps of metabolites and lipids in the mouse RCC samples. Gene expression analysis revealed that the mouse tumors mimicked human RCC. The data suggested that MYC-induced RCC up-regulated the glutaminolytic pathway instead of the glycolytic pathway. The pharmacologic inhibition of glutamine metabolism with bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide impeded MYC-mediated RCC tumor progression. Our studies demonstrate that MYC overexpression causes RCC and points to the inhibition of glutamine metabolism as a potential therapeutic approach for the treatment of this disease.
Collapse
|
1426
|
Ayakannu T, Taylor AH, Willets JM, Konje JC. The evolving role of the endocannabinoid system in gynaecological cancer. Hum Reprod Update 2015; 21:517-35. [PMID: 25958409 DOI: 10.1093/humupd/dmv022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/09/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The 'endocannabinoid system' (ECS), comprising endogenous ligands (endocannabinoids) and their regulating enzymes, together with the cannabinoid receptors, has attracted a great deal of attention because it affects not only all facets of human reproduction, from gametogenesis through to parturition and beyond, but also targets key mechanisms affecting some hallmarks of cancer. Recent evidence showing that cannabinoid receptors play a very important role in the development of malignancies outside of the reproductive organs suggests a similar role for the ECS in the establishment or continued development of gynaecological malignancy. METHODS Primary papers and review articles, and primary sources within these papers, up to December 2014, on the evolving role of the ECS in cancer, with a special focus on gynaecological cancers, were obtained by Medline and PubMed searches using the search terms: 'cancer', 'cannabinoid', 'endocannabinoid', 'gynaecology' and 'malignancy'. Non-English manuscripts were excluded. RESULTS More than 2100 sources were obtained from which only 112 were specifically important to the topic. Analysis of those articles supports a role of the ECS in gynaecological cancers but leaves many gaps in our knowledge that need to be filled. How some of the relevant receptors are activated and cause changes in cell phenotypes that progress to malignancy remains undiscovered and an area for future research. Increasing evidence suggests that malignant transformation within the female genital tract could be accompanied by deregulation of components of the ECS, acting through rather complex cannabinoid receptor-dependent and receptor-independent mechanisms. CONCLUSIONS The paucity of studies in this area suggests that research using animal models is needed to evaluate endocannabinoid signalling in cancer networks. Future randomized clinical studies should reveal whether endocannabinoids or their derivatives prove to be useful therapeutic targets for gynaecological and other cancers.
Collapse
Affiliation(s)
- Thangesweran Ayakannu
- Endocannabinoid Research Group, Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester LE2 7LX, UK
| | - Anthony H Taylor
- Endocannabinoid Research Group, Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester LE2 7LX, UK Biosciences, School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham NG1 4BU, UK
| | - Jonathan M Willets
- Endocannabinoid Research Group, Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester LE2 7LX, UK
| | - Justin C Konje
- Endocannabinoid Research Group, Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester LE2 7LX, UK Department of Obstetrics and Gynaecology, Sidra Medical and Research Centre, Doha P.O. Box 26999, Qatar
| |
Collapse
|
1427
|
Pathak RK, Kolishetti N, Dhar S. Targeted nanoparticles in mitochondrial medicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 7:315-29. [PMID: 25348382 PMCID: PMC4397104 DOI: 10.1002/wnan.1305] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/13/2014] [Accepted: 09/02/2014] [Indexed: 12/12/2022]
Abstract
Mitochondria, the so-called 'energy factory of cells' not only produce energy but also contribute immensely in cellular mortality management. Mitochondrial dysfunctions result in various diseases including but not limited to cancer, atherosclerosis, and neurodegenerative diseases. In the recent years, targeting mitochondria emerged as an attractive strategy to control mitochondrial dysfunction-related diseases. Despite the desire to direct therapeutics to the mitochondria, the actual task is more difficult due to the highly complex nature of the mitochondria. The potential benefits of integrating nanomaterials with properties such as biodegradability, magnetization, and fluorescence into a single object of nanoscale dimensions can lead to the development of hybrid nanomedical platforms for targeting therapeutics to the mitochondria. Only a handful of nanoparticles based on metal oxides, gold nanoparticles, dendrons, carbon nanotubes, and liposomes were recently engineered to target mitochondria. Most of these materials face tremendous challenges when administered in vivo due to their limited biocompatibility. Biodegradable polymeric nanoparticles emerged as eminent candidates for effective drug delivery. In this review, we highlight the current advancements in the development of biodegradable nanoparticle platforms as effective targeting tools for mitochondrial medicine.
Collapse
Affiliation(s)
- Rakesh K. Pathak
- NanoTherapeutics Research Laboratory Department of Chemistry University of Georgia, Athens, GA 30602
| | - Nagesh Kolishetti
- NanoTherapeutics Research Laboratory Department of Chemistry University of Georgia, Athens, GA 30602
- PartiKula LLC, 7777 Davie Rd., Hollywood, FL 33024
| | - Shanta Dhar
- NanoTherapeutics Research Laboratory Department of Chemistry University of Georgia, Athens, GA 30602
| |
Collapse
|
1428
|
Affiliation(s)
- Keigi Fujiwara
- Department of Cardiology, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 2121 West Holcombe Blvd., Houston, TX 77030, USA
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Room 543D, Philadelphia, PA 19107, USA
| |
Collapse
|
1429
|
Wan S, Wu Y, Zhou X, Chen Y, An J, Yu X, Zhang H, Yang H, Xing J. Polymorphisms in Genes of Tricarboxylic Acid Cycle Key Enzymes Are Associated with Early Recurrence of Hepatocellular Carcinoma. PLoS One 2015; 10:e0124471. [PMID: 25894340 PMCID: PMC4404327 DOI: 10.1371/journal.pone.0124471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/01/2015] [Indexed: 02/06/2023] Open
Abstract
Alterations of activity and expression in tricarboxylic acid (TCA) cycle key enzymes have been indicated in several malignancies, including hepatocellular carcinoma (HCC). They play an important role in the progression of cancer. However, the impact of single nucleotide polymorphisms (SNPs) in genes encoding these key enzymes on the recurrence of HCC has not been investigated. In this study, we genotyped 17 SNPs in genes encoding TCA cycle key enzymes and analyzed their association with recurrence-free survival (RFS) in a cohort of 492 Chinese HCC patients by Cox proportional hazard model and survival tree analysis. We identified 7 SNPs in SDHC, SDHD, FH, and IDH2 genes to be significantly associated with the RFS of HCC patients. Moreover, all these SNPs were associated with the early recurrence (within 2 years after surgery) risk of diseases. Cumulative effect analysis showed that these SNPs exhibited a dose-dependent effect on the overall and early recurrence. Further stratified analysis suggested that number of risk genotypes modified the protective effect on HCC recurrence conferred by transcatheter arterial chemoembolization treatment. Finally, the survival tree analysis revealed that SNP rs10789859 in SDHD gene was the primary factor contributing to HCC recurrence in our population. To the best of our knowledge, we for the first time observed the association between SNPs in genes encoding TCA cycle key enzymes and HCC recurrence risk. Further observational and functional studies are needed to validate our findings and generalize its clinical usage.
Collapse
Affiliation(s)
- Shaogui Wan
- Institute of Pharmacy, Pharmaceutical College of Henan University, Kaifeng, Henan, China
| | - Yousheng Wu
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xingchun Zhou
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yibing Chen
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiaze An
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaohe Yu
- Department of Interventional Radiology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Huiqing Zhang
- Department of Pain treatment, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hushan Yang
- Division of Population Science, Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail: (HY); (JX)
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- * E-mail: (HY); (JX)
| |
Collapse
|
1430
|
Williams SB, Ye Y, Huang M, Chang DW, Kamat AM, Pu X, Dinney CP, Wu X. Mitochondrial DNA Content as Risk Factor for Bladder Cancer and Its Association with Mitochondrial DNA Polymorphisms. Cancer Prev Res (Phila) 2015; 8:607-13. [PMID: 25896234 DOI: 10.1158/1940-6207.capr-14-0414] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/01/2015] [Indexed: 01/05/2023]
Abstract
Mitochondrial DNA (mtDNA) content has been shown to be associated with cancer susceptibility. We identified 926 bladder cancer patients and compared these with 926 healthy controls frequency matched on age, gender, and ethnicity. Patients diagnosed with bladder cancer had significantly decreased mtDNA content when compared with control subjects (median, 0.98 vs. 1.04, P < 0.001). Low mtDNA content (i.e., less than the median in control subjects) was associated with a statistically significant increased risk of bladder cancer, when compared with high mtDNA content [Odds ratio (OR), 1.37; 95% confidence interval (CI), 1.13-1.66; P < 0.001). In a trend analysis, a statistically significant dose-response relationship was detected between lower mtDNA content and increasing risk of bladder cancer (Ptrend <0.001). When stratified by host characteristics, advanced age (>65 years), male sex and positive smoking history were significantly associated with low mtDNA content and increased risk of bladder cancer. We identified two unique mtDNA polymorphisms significantly associated with risk of bladder cancer: mitot10464c (OR, 1.39; 95% CI, 1.00-1.93; P = 0.048) and mitoa4918g (OR, 1.40; 95% CI, 1.00-1.95; P = 0.049). Analysis of the joint effect of low mtDNA content and unfavorable mtDNA polymorphisms revealed a 2.5-fold increased risk of bladder cancer (OR, 2.50; 95% CI, 1.60-3.94; P < 0.001). Significant interaction was observed between mitoa4918g and mtDNA content (Pinteraction = 0.028). Low mtDNA content was associated with increased risk of bladder cancer and we identified new susceptibility mtDNA alleles associated with increased risk that require further investigation into the biologic underpinnings of bladder carcinogenesis.
Collapse
Affiliation(s)
- Stephen B Williams
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuanqing Ye
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maosheng Huang
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David W Chang
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ashish M Kamat
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xia Pu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Colin P Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
1431
|
Kumar S, Lombard DB. Mitochondrial sirtuins and their relationships with metabolic disease and cancer. Antioxid Redox Signal 2015; 22:1060-77. [PMID: 25545135 PMCID: PMC4389911 DOI: 10.1089/ars.2014.6213] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Maintenance of metabolic homeostasis is critical for cellular and organismal health. Proper regulation of mitochondrial functions represents a crucial element of overall metabolic homeostasis. Mitochondrial sirtuins (SIRT3, SIRT4, and SIRT5) play pivotal roles in promoting this homeostasis by regulating numerous aspects of mitochondrial metabolism in response to environmental stressors. RECENT ADVANCES New work has illuminated multiple links between mitochondrial sirtuins and cancer. SIRT5 has been shown to regulate the recently described post-translational modifications succinyl-lysine, malonyl-lysine, and glutaryl-lysine. An understanding of these modifications is still in its infancy. Enumeration of SIRT3 and SIRT5 targets via advanced proteomic techniques promises to dramatically enhance insight into functions of these proteins. CRITICAL ISSUES In this review, we highlight the roles of mitochondrial sirtuins and their targets in cellular and organismal metabolic homeostasis. Furthermore, we discuss emerging roles for mitochondrial sirtuins in suppressing and/or promoting tumorigenesis, depending on the cellular and molecular context. FUTURE DIRECTIONS Currently, hundreds of potential SIRT3 and SIRT5 molecular targets have been identified in proteomic experiments. Future studies will need to validate the major targets of these enzymes, and elucidate how acetylation and/or acylation modulate their functionality. A great deal of interest exists in targeting sirtuins pharmacologically; this endeavor will require development of sirtuin-specific modulators (activators and inhibitors) as potential treatments for cancer and metabolic disease.
Collapse
Affiliation(s)
- Surinder Kumar
- 1 Department of Pathology, University of Michigan , Ann Arbor, Michigan
| | | |
Collapse
|
1432
|
Pajuelo-Reguera D, Alán L, Olejár T, Ježek P. Dichloroacetate stimulates changes in the mitochondrial network morphology via partial mitophagy in human SH-SY5Y neuroblastoma cells. Int J Oncol 2015; 46:2409-18. [PMID: 25846762 DOI: 10.3892/ijo.2015.2953] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/11/2015] [Indexed: 11/06/2022] Open
Abstract
Dichloroacetate (DCA) is beneficial in cancer therapy because it induces apoptosis and decreases cancer growth in vitro and in vivo without affecting non-cancer cells. DCA stimulates the activity of the enzyme pyruvate dehydrogenase by inhibiting pyruvate dehydrogenase kinase. Consequently, DCA promotes oxidative phosphorylation after glycolysis. Therefore, DCA produces changes in energy metabolism that could affect the mitochondrial network and mitophagy. This investigation determined the effects of DCA treatment on mitophagy in human neuroblastoma SH-SY5Y cells. SH-SY5Y cells were cultured and distributed into 3 groups: control, NH4Cl and chloroquine. Each group was treated with DCA at 0, 5, 30 and 60 mM for 16 h. Samples were analyzed for cell viability, mtDNA copy number, mitochondrial network morphology and expression of key proteins involved in mitochondrial dynamics, such as LC3b, FIS1, OPA1, PARKIN and PINK1. In all groups, DCA caused a decrease in cell viability, an induction of autophagy in a dose-dependent manner and a decrease in Tim23, FIS1 and PARKIN protein expression, leading to profound morphological changes in the mitochondrial network resulting in shorter and more fragmented filaments. However, TFAM protein levels remained unchanged. Similarly, the mitochondrial copy number was not significantly different among the treatment groups. In conclusion, DCA induces mitophagy and remodeling of the mitochondrial network. In this remodeling, DCA induces a decrease in the expression of key proteins involved in protein degradation and mitochondrial dynamics but does not significantly affect the mtDNA density. Blocking late phase autophagy increases the effects of DCA, suggesting that autophagy protects the cell, at least partially, against DCA.
Collapse
Affiliation(s)
- David Pajuelo-Reguera
- Department of Membrane Transport Biophysics, No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Lukáš Alán
- Department of Membrane Transport Biophysics, No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tomáš Olejár
- Department of Membrane Transport Biophysics, No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Petr Ježek
- Department of Membrane Transport Biophysics, No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
1433
|
Gold nanoparticle-enhanced photodynamic therapy: effects of surface charge and mitochondrial targeting. Ther Deliv 2015; 6:307-21. [DOI: 10.4155/tde.14.115] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: The authors aimed to further improve the efficiency and selectivity of gold nanoparticle (Au NP)-assisted photodynamic therapy by modulating the surface charge of Au NPs and delivering Au NPs particularly to mitochondria of breast cancer cells. Methods: Solid gold nanospheres (˜50 nm) with negative and positive surface charge were synthesized respectively, and mitochondria-targeting Au NPs were prepared by conjugating with triphenylphosphonium molecules. Conclusion: Positively charged Au NPs were preferably taken up by breast cancer cells. Combination of positive surface charge with mitochondria-targeting domain onto Au NPs allowed their accumulation in the mitochondria of breast cancer cells to significantly elevate reactive oxygen species formation in 5-aminolevulinic-acid-enabled photodynamic therapy and improve selective destruction of breast cancer cells.
Collapse
|
1434
|
Abstract
Quinol oxidation in the catalytic quinol oxidation site (Qo site) of cytochrome (cyt) bc1 complexes is the key step of the Q cycle mechanism, which laid the ground for Mitchell’s chemiosmotic theory of energy conversion. Bifurcated electron transfer upon quinol oxidation enables proton uptake and release on opposite membrane sides, thus generating a proton gradient that fuels ATP synthesis in cellular respiration and photosynthesis. The Qo site architecture formed by cyt b and Rieske iron–sulfur protein (ISP) impedes harmful bypass reactions. Catalytic importance is assigned to four residues of cyt b formerly described as PEWY motif in the context of mitochondrial complexes, which we now denominate Qo motif as comprehensive evolutionary sequence analysis of cyt b shows substantial natural variance of the motif with phylogenetically specific patterns. In particular, the Qo motif is identified as PEWY in mitochondria, α- and ε-Proteobacteria, Aquificae, Chlorobi, Cyanobacteria, and chloroplasts. PDWY is present in Gram-positive bacteria, Deinococcus–Thermus and haloarchaea, and PVWY in β- and γ-Proteobacteria. PPWF only exists in Archaea. Distinct patterns for acidophilic organisms indicate environment-specific adaptations. Importantly, the presence of PDWY and PEWY is correlated with the redox potential of Rieske ISP and quinone species. We propose that during evolution from low to high potential electron-transfer systems in the emerging oxygenic atmosphere, cyt bc1 complexes with PEWY as Qo motif prevailed to efficiently use high potential ubiquinone as substrate, whereas cyt b with PDWY operate best with low potential Rieske ISP and menaquinone, with the latter being the likely composition of the ancestral cyt bc1 complex.
Collapse
Affiliation(s)
- Wei-Chun Kao
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Germany
- Faculty of Biology, University of Freiburg, Germany
| | - Carola Hunte
- Institute for Biochemistry and Molecular Biology, ZBMZ, BIOSS Centre for Biological Signalling Studies, University of Freiburg, Germany
- *Corresponding author: E-mail:
| |
Collapse
|
1435
|
Abstract
It has been over 20 years since the discovery that caveolar lipid rafts function as signalling organelles. Lipid rafts create plasma membrane heterogeneity, and caveolae are the most extensively studied subset of lipid rafts. A newly emerging paradigm is that changes in caveolae also generate tumour metabolic heterogeneity. Altered caveolae create a catabolic tumour microenvironment, which supports oxidative mitochondrial metabolism in cancer cells and which contributes to dismal survival rates for cancer patients. In this Review, we discuss the role of caveolae in tumour progression, with a special emphasis on their metabolic and cell signalling effects, and their capacity to transform the tumour microenvironment.
Collapse
Affiliation(s)
- Ubaldo E Martinez-Outschoorn
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | - Federica Sotgia
- 1] Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK. [2] Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, Manchester M20 4BX, UK
| | - Michael P Lisanti
- 1] Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK. [2] Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
1436
|
How do changes in the mtDNA and mitochondrial dysfunction influence cancer and cancer therapy? Challenges, opportunities and models. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 764:16-30. [DOI: 10.1016/j.mrrev.2015.01.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 12/28/2022]
|
1437
|
Beloueche-Babari M, Box C, Arunan V, Parkes HG, Valenti M, De Haven Brandon A, Jackson LE, Eccles SA, Leach MO. Acquired resistance to EGFR tyrosine kinase inhibitors alters the metabolism of human head and neck squamous carcinoma cells and xenograft tumours. Br J Cancer 2015; 112:1206-14. [PMID: 25742484 PMCID: PMC4385966 DOI: 10.1038/bjc.2015.86] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/30/2015] [Accepted: 02/04/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Acquired resistance to molecularly targeted therapeutics is a key challenge in personalised cancer medicine, highlighting the need for identifying the underlying mechanisms and early biomarkers of relapse, in order to guide subsequent patient management. METHODS Here we use human head and neck squamous cell carcinoma (HNSCC) models and nuclear magnetic resonance (NMR) spectroscopy to assess the metabolic changes that follow acquired resistance to EGFR tyrosine kinase inhibitors (TKIs), and which could serve as potential metabolic biomarkers of drug resistance. RESULTS Comparison of NMR metabolite profiles obtained from control (CAL(S)) and EGFR TKI-resistant (CAL(R)) cells grown as 2D monolayers, 3D spheroids or xenograft tumours in athymic mice revealed a number of differences between the sensitive and drug-resistant models. In particular, we observed elevated levels of glycerophosphocholine (GPC) in CAL(R) relative to CAL(S) monolayers, spheroids and tumours, independent of the growth rate or environment. In addition, there was an increase in alanine, aspartate and creatine+phosphocreatine in resistant spheroids and xenografts, and increased levels of lactate, branched-chain amino acids and a fall in phosphoethanolamine only in xenografts. The xenograft lactate build-up was associated with an increased expression of the glucose transporter GLUT-1, whereas the rise in GPC was attributed to inhibition of GPC phosphodiesterase. Reduced glycerophosphocholine (GPC) and phosphocholine were observed in a second HNSCC model probably indicative of a different drug resistance mechanism. CONCLUSIONS Our studies reveal metabolic signatures associated not only with acquired EGFR TKI resistance but also growth pattern, microenvironment and contributing mechanisms in HNSCC models. These findings warrant further investigation as metabolic biomarkers of disease relapse in the clinic.
Collapse
Affiliation(s)
- M Beloueche-Babari
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - C Box
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, UK
| | - V Arunan
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - H G Parkes
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - M Valenti
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, UK
| | - A De Haven Brandon
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, UK
| | - L E Jackson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - S A Eccles
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, UK
| | - M O Leach
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| |
Collapse
|
1438
|
Fayzulin RZ, Perez M, Kozhukhar N, Spadafora D, Wilson GL, Alexeyev MF. A method for mutagenesis of mouse mtDNA and a resource of mouse mtDNA mutations for modeling human pathological conditions. Nucleic Acids Res 2015; 43:e62. [PMID: 25820427 PMCID: PMC4482060 DOI: 10.1093/nar/gkv140] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/10/2015] [Indexed: 12/23/2022] Open
Abstract
Mutations in human mitochondrial DNA (mtDNA) can cause mitochondrial disease and have been associated with neurodegenerative disorders, cancer, diabetes and aging. Yet our progress toward delineating the precise contributions of mtDNA mutations to these conditions is impeded by the limited availability of faithful transmitochondrial animal models. Here, we report a method for the isolation of mutations in mouse mtDNA and its implementation for the generation of a collection of over 150 cell lines suitable for the production of transmitochondrial mice. This method is based on the limited mutagenesis of mtDNA by proofreading-deficient DNA-polymerase γ followed by segregation of the resulting highly heteroplasmic mtDNA population by means of intracellular cloning. Among generated cell lines, we identify nine which carry mutations affecting the same amino acid or nucleotide positions as in human disease, including a mutation in the ND4 gene responsible for 70% of Leber Hereditary Optic Neuropathies (LHON). Similar to their human counterparts, cybrids carrying the homoplasmic mouse LHON mutation demonstrated reduced respiration, reduced ATP content and elevated production of mitochondrial reactive oxygen species (ROS). The generated resource of mouse mtDNA mutants will be useful both in modeling human mitochondrial disease and in understanding the mechanisms of ROS production mediated by mutations in mtDNA.
Collapse
Affiliation(s)
- Rafik Z Fayzulin
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, AL 36688, USA
| | - Michael Perez
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Natalia Kozhukhar
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Domenico Spadafora
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Glenn L Wilson
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, AL 36688, USA
| | - Mikhail F Alexeyev
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
1439
|
Carlson EA, Marquez RT, Du F, Wang Y, Xu L, Yan SS. Overexpression of 17β-hydroxysteroid dehydrogenase type 10 increases pheochromocytoma cell growth and resistance to cell death. BMC Cancer 2015; 15:166. [PMID: 25879199 PMCID: PMC4384325 DOI: 10.1186/s12885-015-1173-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/06/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND 17β-hydroxysteroid dehydrogenase type 10 (HSD10) has been shown to play a protective role in cells undergoing stress. Upregulation of HSD10 under nutrient-limiting conditions leads to recovery of a homeostatic state. Across disease states, increased HSD10 levels can have a profound and varied impact, such as beneficial in Parkinson's disease and harmful in Alzheimer's disease. Recently, HSD10 overexpression has been observed in some prostate and bone cancers, consistently correlating with poor patient prognosis. As the role of HSD10 in cancer remains underexplored, we propose that cancer cells utilize this enzyme to promote cancer cell survival under cell death conditions. METHODS The proliferative effect of HSD10 was examined in transfected pheochromocytoma cells by growth curve analysis and a xenograft model. Fluctuations in mitochondrial bioenergetics were evaluated by electron transport chain complex enzyme activity assays and energy production. Additionally, the effect of HSD10 on pheochromocytoma resistance to cell death was investigated using TUNEL staining, MTT, and complex IV enzyme activity assays. RESULTS In this study, we examined the tumor-promoting effect of HSD10 in pheochromocytoma cells. Overexpression of HSD10 increased pheochromocytoma cell growth in both in vitro cell culture and an in vivo xenograft mouse model. The increases in respiratory enzymes and energy generation observed in HSD10-overexpressing cells likely supported the accelerated growth rate observed. Furthermore, cells overexpressing HSD10 were more resistant to oxidative stress-induced perturbation. CONCLUSIONS Our findings demonstrate that overexpression of HSD10 accelerates pheochromocytoma cell growth, enhances cell respiration, and increases cellular resistance to cell death induction. This suggests that blockade of HSD10 may halt and/or prevent cancer growth, thus providing a promising novel target for cancer patients as a screening or therapeutic option.
Collapse
Affiliation(s)
- Emily A Carlson
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, 66047, USA. .,Higuchi Biosciences Center, University of Kansas, Lawrence, KS, 66047, USA.
| | - Rebecca T Marquez
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA.
| | - Fang Du
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, 66047, USA. .,Higuchi Biosciences Center, University of Kansas, Lawrence, KS, 66047, USA.
| | - Yongfu Wang
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, 66047, USA. .,Higuchi Biosciences Center, University of Kansas, Lawrence, KS, 66047, USA.
| | - Liang Xu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA.
| | - Shirley ShiDu Yan
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, 66047, USA. .,Higuchi Biosciences Center, University of Kansas, Lawrence, KS, 66047, USA.
| |
Collapse
|
1440
|
Wahrheit J, Nonnenmacher Y, Sperber S, Heinzle E. High-throughput respiration screening of single mitochondrial substrates using permeabilized CHO cells highlights control of mitochondria metabolism. Eng Life Sci 2015. [DOI: 10.1002/elsc.201400175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Judith Wahrheit
- Biochemical Engineering Institute; Saarland University; Saarbrücken Germany
| | | | - Saskia Sperber
- Biochemical Engineering Institute; Saarland University; Saarbrücken Germany
| | - Elmar Heinzle
- Biochemical Engineering Institute; Saarland University; Saarbrücken Germany
| |
Collapse
|
1441
|
Zhang S, Yang C, Yang Z, Zhang D, Ma X, Mills G, Liu Z. Homeostasis of redox status derived from glucose metabolic pathway could be the key to understanding the Warburg effect. Am J Cancer Res 2015; 5:1265-1280. [PMID: 26101696 PMCID: PMC4473309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/06/2015] [Indexed: 06/04/2023] Open
Abstract
Glucose metabolism in mitochondria through oxidative phosphorylation (OXPHOS) for generation of adenosine triphosphate (ATP) is vital for cell function. However, reactive oxygen species (ROS), a by-product from OXPHOS, is a major source of endogenously produced toxic stressors on the genome. In fact, ATP could be efficiently produced in a high throughput manner without ROS generation in cytosol through glycolysis, which could be a unique and critical metabolic pathway to prevent spontaneous mutation during DNA replication. Therefore glycolysis is dominant in robust proliferating cells. Indeed, aerobic glycolysis, or the Warburg effect, in normal proliferating cells is an example of homeostasis of redox status by transiently shifting metabolic flux from OXPHOS to glycolysis to avoid ROS generation during DNA synthesis and protect genome integrity. The process of maintaining redox homeostasis is driven by genome wide transcriptional clustering with mitochondrial retrograde signaling and coupled with the glucose metabolic pathway and cell division cycle. On the contrary, the Warburg effect in cancer cells is the results of the alteration of redox status from a reprogramed glucose metabolic pathway caused by the dysfunctional OXPHOS. Mutations in mitochondrial DNA (mtDNA) and nuclear DNA (nDNA) disrupt mitochondrial structural integrity, leading to reduced OXPHOS capacity, sustained glycolysis and excessive ROS leak, all of which are responsible for tumor initiation, progression and metastasis. A "plumbing model" is used to illustrate how redox status could be regulated through glucose metabolic pathway and provide a new insight into the understanding of the Warburg effect in both normal and cancer cells.
Collapse
Affiliation(s)
- Shiwu Zhang
- Department of Pathology, Tianjin Union Medical CenterTianjin, People’s Republic of China
| | - Chuanwei Yang
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, TX, 77030, USA
- Breast Medical Oncology, The University of Texas MD Anderson Cancer CenterHouston, TX, 77030, USA
| | - Zhenduo Yang
- Department of Pathology, Tianjin Union Medical CenterTianjin, People’s Republic of China
| | - Dan Zhang
- Department of Pathology, Tianjin Union Medical CenterTianjin, People’s Republic of China
| | - Xiaoping Ma
- Department of Integrative Biology and Pharmacology, The University of Texas Medical SchoolHouston, TX 77030, USA
| | - Gordon Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, TX, 77030, USA
| | - Zesheng Liu
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, TX, 77030, USA
| |
Collapse
|
1442
|
Hajrezaie M, Paydar M, Looi CY, Moghadamtousi SZ, Hassandarvish P, Salga MS, Karimian H, Shams K, Zahedifard M, Majid NA, Ali HM, Abdulla MA. Apoptotic effect of novel Schiff based CdCl₂(C₁₄H₂₁N₃O₂) complex is mediated via activation of the mitochondrial pathway in colon cancer cells. Sci Rep 2015; 5:9097. [PMID: 25764970 PMCID: PMC4649862 DOI: 10.1038/srep09097] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 01/21/2015] [Indexed: 01/06/2023] Open
Abstract
The development of metal-based agents has had a tremendous role in the present progress in cancer chemotherapy. One well-known example of metal-based agents is Schiff based metal complexes, which hold great promise for cancer therapy. Based on the potential of Schiff based complexes for the induction of apoptosis, this study aimed to examine the cytotoxic and apoptotic activity of a CdCl2(C14H21N3O2) complex on HT-29 cells. The complex exerted a potent suppressive effect on HT-29 cells with an IC50 value of 2.57 ± 0.39 after 72 h of treatment. The collapse of the mitochondrial membrane potential and the elevated release of cytochrome c from the mitochondria to the cytosol indicate the involvement of the intrinsic pathway in the induction of apoptosis. The role of the mitochondria-dependent apoptotic pathway was further proved by the significant activation of the initiator caspase-9 and the executioner caspases-3 and -7. In addition, the activation of caspase-8, which is associated with the suppression of NF-κB translocation to the nucleus, also revealed the involvement of the extrinsic pathway in the induced apoptosis. The results suggest that the CdCl2(C14H21N3O2) complex is able to induce the apoptosis of colon cancer cells and is a potential candidate for future cancer studies.
Collapse
Affiliation(s)
- Maryam Hajrezaie
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mohammadjavad Paydar
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Pouya Hassandarvish
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Hamed Karimian
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Keivan Shams
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Maryam Zahedifard
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hapipah Mohd Ali
- Department of Chemistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
1443
|
Tan K, Fujimoto M, Takii R, Takaki E, Hayashida N, Nakai A. Mitochondrial SSBP1 protects cells from proteotoxic stresses by potentiating stress-induced HSF1 transcriptional activity. Nat Commun 2015; 6:6580. [PMID: 25762445 PMCID: PMC4558571 DOI: 10.1038/ncomms7580] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 02/09/2015] [Indexed: 12/13/2022] Open
Abstract
Heat-shock response is an adaptive response to proteotoxic stresses including heat shock, and is regulated by heat-shock factor 1 (HSF1) in mammals. Proteotoxic stresses challenge all subcellular compartments including the mitochondria. Therefore, there must be close connections between mitochondrial signals and the activity of HSF1. Here, we show that heat shock triggers nuclear translocation of mitochondrial SSBP1, which is involved in replication of mitochondrial DNA, in a manner dependent on the mitochondrial permeability transition pore ANT–VDAC1 complex and direct interaction with HSF1. HSF1 recruits SSBP1 to the promoters of genes encoding cytoplasmic/nuclear and mitochondrial chaperones. HSF1–SSBP1 complex then enhances their induction by facilitating the recruitment of a chromatin-remodelling factor BRG1, and supports cell survival and the maintenance of mitochondrial membrane potential against proteotoxic stresses. These results suggest that the nuclear translocation of mitochondrial SSBP1 is required for the regulation of cytoplasmic/nuclear and mitochondrial proteostasis against proteotoxic stresses. Heat shock induces proteotoxic stress, and the cellular response is mediated by heat-shock factor 1 (HSF1). Here, Tan et al. show that following heat shock, mitochondrial SSBP1 translocates to the nucleus and binds HSF1 to enhance the expression of chaperones and support the maintenance of mitochondrial function.
Collapse
Affiliation(s)
- Ke Tan
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Mitsuaki Fujimoto
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Ryosuke Takii
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Eiichi Takaki
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Naoki Hayashida
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| | - Akira Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan
| |
Collapse
|
1444
|
Mechanisms by which different functional states of mitochondria define yeast longevity. Int J Mol Sci 2015; 16:5528-54. [PMID: 25768339 PMCID: PMC4394491 DOI: 10.3390/ijms16035528] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/05/2015] [Accepted: 03/05/2015] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial functionality is vital to organismal physiology. A body of evidence supports the notion that an age-related progressive decline in mitochondrial function is a hallmark of cellular and organismal aging in evolutionarily distant eukaryotes. Studies of the baker’s yeast Saccharomyces cerevisiae, a unicellular eukaryote, have led to discoveries of genes, signaling pathways and chemical compounds that modulate longevity-defining cellular processes in eukaryotic organisms across phyla. These studies have provided deep insights into mechanistic links that exist between different traits of mitochondrial functionality and cellular aging. The molecular mechanisms underlying the essential role of mitochondria as signaling organelles in yeast aging have begun to emerge. In this review, we discuss recent progress in understanding mechanisms by which different functional states of mitochondria define yeast longevity, outline the most important unanswered questions and suggest directions for future research.
Collapse
|
1445
|
Yang L, Li N, Pan W, Yu Z, Tang B. Real-Time Imaging of Mitochondrial Hydrogen Peroxide and pH Fluctuations in Living Cells Using a Fluorescent Nanosensor. Anal Chem 2015; 87:3678-84. [DOI: 10.1021/ac503975x] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Limin Yang
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Na Li
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Wei Pan
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Zhengze Yu
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| |
Collapse
|
1446
|
|
1447
|
Bhat TA, Kumar S, Chaudhary AK, Yadav N, Chandra D. Restoration of mitochondria function as a target for cancer therapy. Drug Discov Today 2015; 20:635-43. [PMID: 25766095 DOI: 10.1016/j.drudis.2015.03.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/16/2015] [Accepted: 03/03/2015] [Indexed: 12/15/2022]
Abstract
Defective oxidative phosphorylation has a crucial role in the attenuation of mitochondrial function, which confers therapy resistance in cancer. Various factors, including endogenous heat shock proteins (HSPs) and exogenous agents such as dichloroacetate, restore respiratory and other physiological functions of mitochondria in cancer cells. Functional mitochondria might ultimately lead to the restoration of apoptosis in cancer cells that are refractory to current anticancer agents. Here, we summarize the key reasons contributing to mitochondria dysfunction in cancer cells and how restoration of mitochondrial function could be exploited for cancer therapeutics.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
1448
|
Lloyd RE, Keatley K, Littlewood DTJ, Meunier B, Holt WV, An Q, Higgins SC, Polyzoidis S, Stephenson KF, Ashkan K, Fillmore HL, Pilkington GJ, McGeehan JE. Identification and functional prediction of mitochondrial complex III and IV mutations associated with glioblastoma. Neuro Oncol 2015; 17:942-52. [PMID: 25731774 PMCID: PMC4474231 DOI: 10.1093/neuonc/nov020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/23/2015] [Indexed: 12/30/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common primary brain tumor in adults, with a dismal prognosis. Treatment is hampered by GBM's unique biology, including differential cell response to therapy. Although several mitochondrial abnormalities have been identified, how mitochondrial DNA (mtDNA) mutations contribute to GBM biology and therapeutic response remains poorly described. We sought to determine the spectrum of functional complex III and IV mtDNA mutations in GBM. Methods The complete mitochondrial genomes of 10 GBM cell lines were obtained using next-generation sequencing and combined with another set obtained from 32 GBM tissues. Three-dimensional structural mapping and analysis of all the nonsynonymous mutations identified in complex III and IV proteins was then performed to investigate functional importance. Results Over 200 mutations were identified in the mtDNAs, including a significant proportion with very low mutational loads. Twenty-five were nonsynonymous mutations in complex III and IV, 9 of which were predicted to be functional and affect mitochondrial respiratory chain activity. Most of the functional candidates were GBM specific and not found in the general population, and 2 were present in the germ-line. Patient-specific maps reveal that 43% of tumors carry at least one functional candidate. Conclusions We reveal that the spectrum of GBM-associated mtDNA mutations is wider than previously thought, as well as novel structural-functional links between specific mtDNA mutations, abnormal mitochondria, and the biology of GBM. These results could provide tangible new prognostic indicators as well as targets with which to guide the development of patient-specific mitochondrially mediated chemotherapeutic approaches.
Collapse
Affiliation(s)
- Rhiannon E Lloyd
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Kathleen Keatley
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - D Timothy J Littlewood
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Brigitte Meunier
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - William V Holt
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Qian An
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Samantha C Higgins
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Stavros Polyzoidis
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Katie F Stephenson
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Keyoumars Ashkan
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Helen L Fillmore
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - Geoffrey J Pilkington
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| | - John E McGeehan
- Brain Tumour Research Centre (R.E.L., K.K., S.C.H., K.F.S., H.L.F., G.J.P.), Molecular Biophysics Laboratories (K.K., J.E.M.), Epigenetics and Developmental Biology Laboratories (Q.A.), Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK; Department of Life Sciences, Natural History Museum, London, UK (D.T.J.L.); Institut de Biologie Intégrative de la Cellule, Paris-Saclay University, CEA, CNRS, Université Paris-Sud, Gif sur Yvette, France (B.M.); Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK (W.V.H.); Department of Neurosurgery, Kings College Hospital, London, UK (S.P., K.A.)
| |
Collapse
|
1449
|
Sechi S, Frappaolo A, Belloni G, Colotti G, Giansanti MG. The multiple cellular functions of the oncoprotein Golgi phosphoprotein 3. Oncotarget 2015; 6:3493-506. [PMID: 25691054 PMCID: PMC4414131 DOI: 10.18632/oncotarget.3051] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/07/2015] [Indexed: 12/13/2022] Open
Abstract
The highly conserved Golgi phosphoprotein 3 (GOLPH3) protein, a component of Trans-Golgi Network (TGN), has been defined as a "first-in-class Golgi oncoprotein" and characterized as a Phosphatidylinositol 4-phosphate [PI(4)P] effector at the Golgi. GOLPH3 is commonly amplified in several solid tumors. Furthermore this protein has been associated with poor prognosis in many cancers. Highly conserved from yeast to humans, GOLPH3 provides an essential function in vesicle trafficking and Golgi structure. Recent data have also implicated this oncoprotein in regulation of cytokinesis, modulation of mitochondrial mass and cellular response to DNA damage. A minute dissection of the molecular pathways that require GOLPH3 protein will be helpful to develop new therapeutic cancer strategies.
Collapse
Affiliation(s)
- Stefano Sechi
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| | - Anna Frappaolo
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| | - Giorgio Belloni
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| | - Gianni Colotti
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, 00185 Roma, Italy
| | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| |
Collapse
|
1450
|
Targeting mitochondria metabolism for cancer therapy. Nat Chem Biol 2015; 11:9-15. [PMID: 25517383 DOI: 10.1038/nchembio.1712] [Citation(s) in RCA: 980] [Impact Index Per Article: 108.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/04/2014] [Indexed: 01/02/2023]
Abstract
Mitochondria have a well-recognized role in the production of ATP and the intermediates needed for macromolecule biosynthesis, such as nucleotides. Mitochondria also participate in the activation of signaling pathways. Overall, accumulating evidence now suggests that mitochondrial bioenergetics, biosynthesis and signaling are required for tumorigenesis. Thus, emerging studies have begun to demonstrate that mitochondrial metabolism is potentially a fruitful arena for cancer therapy. In this Perspective, we highlight recent developments in targeting mitochondrial metabolism for the treatment of cancer.
Collapse
|