51
|
Cai Z, Zhao JS, Li JJ, Peng DN, Wang XY, Chen TL, Qiu YP, Chen PP, Li WJ, Xu LY, Li EM, Tam JPM, Qi RZ, Jia W, Xie D. A combined proteomics and metabolomics profiling of gastric cardia cancer reveals characteristic dysregulations in glucose metabolism. Mol Cell Proteomics 2010; 9:2617-28. [PMID: 20699381 DOI: 10.1074/mcp.m110.000661] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastric cardia cancer (GCC), which occurs at the gastric-esophageal boundary, is one of the most malignant tumors. Despite its high mortality and morbidity, the molecular mechanism of initiation and progression of this disease is largely unknown. In this study, using proteomics and metabolomics approaches, we found that the level of several enzymes and their related metabolic intermediates involved in glucose metabolism were deregulated in GCC. Among these enzymes, two subunits controlling pyruvic acid efflux, lactate dehydrogenase A (LDHA) and pyruvate dehydrogenase B (PDHB), were further analyzed in vitro. Either down-regulation of LDH subunit LDHA or overexpression of PDH subunit PDHB could force pyruvic acid into the Krebs cycle rather than the glycolysis process in AGS gastric cancer cells, which inhibited cell growth and cell migration. Our results reflect an important glucose metabolic signature, especially the dysregulation of pyruvic acid efflux in the development of GCC. Forced transition from glycolysis to the Krebs cycle had an inhibitory effect on GCC progression, providing potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Zhen Cai
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Lu YY, Chen TS, Wang XP, Li L. Single-cell analysis of dihydroartemisinin-induced apoptosis through reactive oxygen species-mediated caspase-8 activation and mitochondrial pathway in ASTC-a-1 cells using fluorescence imaging techniques. JOURNAL OF BIOMEDICAL OPTICS 2010; 15:046028. [PMID: 20799830 DOI: 10.1117/1.3481141] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Dihydroartemisinin (DHA), a front-line antimalarial herbal compound, has been shown to possess promising anticancer activity with low toxicity. We have previously reported that DHA induced caspase-3-dependent apoptosis in human lung adenocarcinoma cells. However, the cellular target and molecular mechanism of DHA-induced apoptosis is still poorly defined. We use confocal fluorescence microscopy imaging, fluorescence resonance energy transfer, and fluorescence recovery after photobleaching techniques to explore the roles of DHA-elicited reactive oxygen species (ROS) in the DHA-induced Bcl-2 family proteins activation, mitochondrial dysfunction, caspase cascade, and cell death. Cell Counting Kit-8 assay and flow cytometry analysis showed that DHA induced ROS-mediated apoptosis. Confocal imaging analysis in a single living cell and Western blot assay showed that DHA triggered ROS-dependent Bax translocation, mitochondrial membrane depolarization, alteration of mitochondrial morphology, cytochrome c release, caspase-9, caspase-8, and caspase-3 activation, indicating the coexistence of ROS-mediated mitochondrial and death receptor pathway. Collectively, our findings demonstrate for the first time that DHA induces cell apoptosis by triggering ROS-mediated caspase-8/Bid activation and the mitochondrial pathway, which provides some novel insights into the application of DHA as a potential anticancer drug and a new therapeutic strategy by targeting ROS signaling in lung adenocarcinoma therapy in the future.
Collapse
Affiliation(s)
- Ying-Ying Lu
- South China Normal University, Institute of Laser Life Science, MOE Key Laboratory of Laser Life Science, Guangzhou, China
| | | | | | | |
Collapse
|
53
|
Formentini L, Martínez-Reyes I, Cuezva JM. The mitochondrial bioenergetic capacity of carcinomas. IUBMB Life 2010; 62:554-60. [DOI: 10.1002/iub.352] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
54
|
Amplification of 8q21 in breast cancer is independent of MYC and associated with poor patient outcome. Mod Pathol 2010; 23:603-10. [PMID: 20139910 DOI: 10.1038/modpathol.2010.5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Copy number gains involving the long arm of chromosome 8, including high-level amplifications at 8q21 and 8q24, have been frequently reported in breast cancer. Although the role of the MYC gene as the driver of the 8q24 amplicon is well established, the significance of the 8q21 amplicon is less clear. The breast cancer cell line SK-BR-3 contains three separate 8q21 amplicons, the distal two of which correspond to putative target genes TPD52 and WWP1. To understand the effect of proximal 8q21 amplification on breast cancer phenotype and patient prognosis, we analyzed 8q21 copy number changes using fluorescence in situ hybridization (FISH) in a tissue microarray containing more than 2000 breast cancers. Amplification at 8q21 was found in 3% of tumors, and was associated with medullary type (P<0.03), high tumor grade (P<0.0001), high Ki67 labeling index (P<0.05), amplification of MYC (P<0.0001), HER2, MDM2, and CCND1 (P<0.05 each), as well as the total number of gene amplifications (P<0.0001). 8q21 copy number gains were significantly related to unfavorable patient outcome in univariate analysis. However, multivariate Cox regression analysis did not reveal an independent prognostic value of 8q21 amplification. The position of our FISH probe and data of a previously performed high-resolution CGH study in the breast cancer cell line SK-BR-3 involve TCEB1 and TMEM70 as new possible candidate oncogenes at 8q21 in breast cancer.
Collapse
|
55
|
Circu ML, Aw TY. Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic Biol Med 2010; 48:749-62. [PMID: 20045723 PMCID: PMC2823977 DOI: 10.1016/j.freeradbiomed.2009.12.022] [Citation(s) in RCA: 2384] [Impact Index Per Article: 170.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 12/21/2009] [Accepted: 12/27/2009] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are products of normal metabolism and xenobiotic exposure, and depending on their concentration, ROS can be beneficial or harmful to cells and tissues. At physiological low levels, ROS function as "redox messengers" in intracellular signaling and regulation, whereas excess ROS induce oxidative modification of cellular macromolecules, inhibit protein function, and promote cell death. Additionally, various redox systems, such as the glutathione, thioredoxin, and pyridine nucleotide redox couples, participate in cell signaling and modulation of cell function, including apoptotic cell death. Cell apoptosis is initiated by extracellular and intracellular signals via two main pathways, the death receptor- and the mitochondria-mediated pathways. Various pathologies can result from oxidative stress-induced apoptotic signaling that is consequent to ROS increases and/or antioxidant decreases, disruption of intracellular redox homeostasis, and irreversible oxidative modifications of lipid, protein, or DNA. In this review, we focus on several key aspects of ROS and redox mechanisms in apoptotic signaling and highlight the gaps in knowledge and potential avenues for further investigation. A full understanding of the redox control of apoptotic initiation and execution could underpin the development of therapeutic interventions targeted at oxidative stress-associated disorders.
Collapse
Affiliation(s)
- Magdalena L Circu
- Department of Molecular & Cellular Physiology, Louisiana University Health Sciences Center, Shreveport, LA 71130, USA
| | | |
Collapse
|
56
|
Abstract
Down-regulation of β-F1-ATPase (the catalytic subunit of the mitochondrial H+-ATP synthase) is a hallmark of many human tumours. The expression level of β-F1-ATPase provides a marker of the prognosis of cancer patients, as well as of the tumour response to chemotherapy. However, the mechanisms that participate in down-regulating its expression in human tumours remain unknown. In the present study, we have investigated the expression of β-F1-ATPase mRNA (termed β-mRNA) in breast, colon and lung adenocarcinomas and squamous carcinomas of the lung. Despite the down-regulation of the protein, tumour β-mRNA levels remained either unchanged (breast and lung adenocarcinomas) or significantly increased (colon and squamous lung carcinomas) when compared with paired normal tissues, suggesting a specific translation-masking event for β-mRNA in human cancer. Consistently, we show using cell-free translation assays that a large fraction (~70%) of protein extracts derived from breast and lung adenocarcinomas specifically repress the translation of β-mRNA. We show that the 3′UTR (3′ untranslated region) of human β-mRNA is a relevant cis-acting element required for efficient translation of the transcript. However, an RNA chimaera bearing the 3′UTR of human β-mRNA does not recapitulate the inhibitory effect of tumour extracts on β-mRNA translation. Overall, the findings of the present study support the hypothesis that down-regulation of the bioenergetic activity of mitochondria in human tumours is exerted by translation silencing of β-mRNA.
Collapse
|
57
|
Morán M, Rivera H, Sánchez-Aragó M, Blázquez A, Merinero B, Ugalde C, Arenas J, Cuezva JM, Martín MA. Mitochondrial bioenergetics and dynamics interplay in complex I-deficient fibroblasts. Biochim Biophys Acta Mol Basis Dis 2010; 1802:443-53. [PMID: 20153825 DOI: 10.1016/j.bbadis.2010.02.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 02/03/2010] [Accepted: 02/08/2010] [Indexed: 11/17/2022]
Abstract
BACKGROUND Complex I (CI) deficiency is the most frequent cause of OXPHOS disorders. Recent studies have shown increases in reactive oxygen species (ROS) production and mitochondrial network disturbances in patients' fibroblasts harbouring mutations in CI subunits. OBJECTIVES The present work evaluates the impact of mutations in the NDUFA1 and NDUFV1 genes of CI on mitochondrial bioenergetics and dynamics, in fibroblasts from patients suffering isolated CI deficiency. RESULTS Decreased oxygen consumption rate and slow growth rate were found in patients with severe CI deficiency. Mitochondrial diameter was slightly increased in patients' cells cultured in galactose or treated with 2'-deoxyglucose without evidence of mitochondrial fragmentation. Expression levels of the main proteins involved in mitochondrial dynamics, OPA1, MFN2, and DRP1, were slightly augmented in all patients' cells lines. The study of mitochondrial dynamics showed delayed recovery of the mitochondrial network after treatment with the uncoupler carbonyl cyanide m-chlorophenyl hydrazone (cccp) in patients with severe CI deficiency. Intracellular ROS levels were not increased neither in glucose nor galactose medium in patients' fibroblasts. CONCLUSION Our main finding was that severe CI deficiency in patients harbouring mutations in the NDUFA1 and NDUFV1 genes is linked to a delayed mitochondrial network recovery after cccp treatment. However, the CI deficiency is neither associated with massive mitochondrial fragmentation nor with increased ROS levels. The different genetic backgrounds of patients with OXPHOS disorders would explain, at least partially, differences in the pathophysiological manifestations of CI deficiency.
Collapse
Affiliation(s)
- M Morán
- Centro de Investigación, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Sun Q, Chen T, Wang X, Wei X. Taxol induces paraptosis independent of both protein synthesis and MAPK pathway. J Cell Physiol 2010; 222:421-32. [PMID: 19918793 DOI: 10.1002/jcp.21982] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our recent studies have shown that high concentration of taxol induced a caspase-independent paraptosis-like cell death and cytoplasmic vacuolization derived predominantly from endoplasmic reticulum (ER) swelling in human lung carcinoma cell lines (ASTC-a-1). In this report, we further explored the relationship between taxol-induced cell death and vacuolization, and the roles of protein synthesis, mitogen-activated protein kinase kinases (MEK), c-jun N-terminal kinase (JNK) and P38 in taxol-induced paraptosis. Enhanced green fluorescent protein (EGFP) was used to probe the cell morphological change, while ER-targeted red fluorescent protein (er-RFP) was used to probe ER spatial distribution. Real-time monitoring of the ER swelling dynamics during the formation of vacuolization inside single living cells co-expressing EGFP and er-RFP further demonstrated that taxol-induced cytoplasmic vacuolization was from the ER restructuring due to fusion and swelling. PI staining showed that taxol-induced vacuolization was not necrosis. These results further demonstrated that the taxol-induced cell death was neither apoptosis nor necrosis, and fitted the criteria of paraptosis characterized by cytoplasmic vacuolization, caspase-independence, lack of apoptotic morphology and insensitivity to broad caspase inhibitor. Our data further indicated that taxol-induced paraptosis required neither protein synthesis nor the participation of MEK, JNK, and P38, which was different from the insulin-like growth factor I receptor (IGFIR)-induced paraptosis. These results suggest that high concentration of taxol activates an alternative paraptotic cell death pathway.
Collapse
Affiliation(s)
- Qingrui Sun
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, South China Normal University, Guangzhou, China
| | | | | | | |
Collapse
|
59
|
Furuta E, Okuda H, Kobayashi A, Watabe K. Metabolic genes in cancer: their roles in tumor progression and clinical implications. Biochim Biophys Acta Rev Cancer 2010; 1805:141-52. [PMID: 20122995 DOI: 10.1016/j.bbcan.2010.01.005] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2009] [Revised: 01/11/2010] [Accepted: 01/24/2010] [Indexed: 12/12/2022]
Abstract
Re-programming of metabolic pathways is a hallmark of physiological changes in cancer cells. The expression of certain genes that directly control the rate of key metabolic pathways including glycolysis, lipogenesis and nucleotide synthesis are drastically altered at different stages of tumor progression. These alterations are generally considered as an adaptation of tumor cells; however, they also contribute to the progression of tumor cells to become more aggressive phenotypes. This review summarizes the recent information about the mechanistic link of these genes to oncogenesis and their potential utility as diagnostic markers as well as for therapeutic targets. We particularly focus on three groups of genes; GLUT1, G6PD, TKTL1 and PGI/AMF in glycolytic pathway, ACLY, ACC1 and FAS in lipogenesis and RRM2, p53R2 and TYMS for nucleotide synthesis. All these genes are highly up-regulated in a variety of tumor cells in cancer patients, and they play active roles in tumor progression rather than expressing merely as a consequence of phenotypic change of the cancer cells. Molecular dissection of their orchestrated networks and understanding the exact mechanism of their expression will provide a window of opportunity to target these genes for specific cancer therapy. We also reviewed existing database of gene microarray to validate the utility of these genes for cancer diagnosis.
Collapse
Affiliation(s)
- Eiji Furuta
- Department of Medical Microbiology and Immunology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | | | | | | |
Collapse
|
60
|
Sánchez-Aragó M, Chamorro M, Cuezva JM. Selection of cancer cells with repressed mitochondria triggers colon cancer progression. Carcinogenesis 2010; 31:567-76. [PMID: 20080835 DOI: 10.1093/carcin/bgq012] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The contribution that mitochondrial bioenergetics could have in cancer development is debated. Here, we have generated HCT116-derived colocarcinoma cell lines expressing different levels of the beta catalytic subunit of the mitochondrial H+-adenosine triphosphate synthase to assess the contribution of mitochondrial bioenergetics in colon cancer progression. The generated cells exhibit large ultrastructural, transcriptomic, proteomic and functional differences in their mitochondria and in their in vivo tumor forming capacity. We show that the activity of oxidative phosphorylation defines the rate of glucose utilization by aerobic glycolysis. The aggressive cellular phenotype, which is highly glycolytic, is bound to the deregulated expression of genes involved in metabolic processes, the regulation of the cell cycle, apoptosis, angiogenesis and cell adhesion. Remarkably, the molecular and ultrastructural analysis of the tumors derived from the three HCT116 cell lines under study highlight that tumor promotion inevitably requires the selection of cancer cells with a repressed biogenesis and functional activity of mitochondria, i.e. the highly glycolytic phenotype is selected for tumor development. The tumor forming potential of the cells is a non-genetically acquired condition that provides the cancer cell with a cell-death resistant phenotype. An abrogated mitochondrial respiration contributes to a diminished potential for reactive oxygen species signaling in response to 5-fluorouracil treatment. Treatment of cancer cells with dichloroacetate partially restores the functional differentiation of mitochondria and promotes tumor regression, emphasizing the reversible nature of the metabolic trait of cancer.
Collapse
Affiliation(s)
- María Sánchez-Aragó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM) and Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | | | | |
Collapse
|
61
|
Behera MA, Dai Q, Garde R, Saner C, Jungheim E, Price TM. Progesterone stimulates mitochondrial activity with subsequent inhibition of apoptosis in MCF-10A benign breast epithelial cells. Am J Physiol Endocrinol Metab 2009; 297:E1089-96. [PMID: 19690070 PMCID: PMC2781356 DOI: 10.1152/ajpendo.00209.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of progesterone on breast epithelial cells remain poorly defined with observations showing both proliferative and antiproliferative effects. As an example, progesterone levels correlate with increased epithelial cell proliferation, but there is discordance between the dividing cells and the cells with nuclear progesterone receptor expression. The release of paracrine growth factors from nuclear receptor-positive cells has been postulated as a mechanism, since in vitro studies show a lack of growth effect by progesterone in breast epithelial cells lacking nuclear receptors. This study examined possible nongenomic effects of progesterone in breast epithelia by using MCF-10A cells known to lack nuclear progesterone receptor expression. Treatment for 30-60 min with progesterone or the progestin, R5020, increased mitochondrial activity as shown by an increase in mitochondrial membrane potential (hyperpolarization) with a concordant increase in total cellular ATP. The reaction was inhibited by a specific progesterone receptor antagonist and not affected by the translation inhibitor cycloheximide. Progestin treatment inhibited apoptosis induced by activation of the FasL pathway, as shown by a decrease in sub-G(1) cell fraction during fluorescence-activated cell sorting and a decrease in caspase 3/7 levels. Progestin treatment did not alter the cell cycle over 48 h. Our study demonstrates a nongenomic action of progesterone on benign breast epithelial cells, resulting in enhanced cellular respiration and protection from apoptosis.
Collapse
|
62
|
Acebo P, Giner D, Calvo P, Blanco-Rivero A, Ortega AD, Fernández PL, Roncador G, Fernández-Malavé E, Chamorro M, Cuezva JM. Cancer abolishes the tissue type-specific differences in the phenotype of energetic metabolism. Transl Oncol 2009; 2:138-45. [PMID: 19701498 PMCID: PMC2730139 DOI: 10.1593/tlo.09106] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 02/27/2009] [Accepted: 03/02/2009] [Indexed: 12/31/2022] Open
Abstract
Nowadays, cellular bioenergetics has become a central issue of investigation in cancer biology. Recently, the metabolic activity of the cancer cell has been shown to correlate with a proteomic index that informs of the relative mitochondrial activity of the cell. Within this new field of investigation, we report herein the production and characterization of high-affinity monoclonal antibodies against proteins of the "bioenergetic signature" of the cell. The use of recombinant proteins and antibodies against the mitochondrial beta-F1-ATPase and Hsp60 proteins and the enzymes of the glycolytic pathway glyceraldehyde-3-phosphate dehydrogenase and pyruvate kinase M2 in quantitative assays provide, for the first time, the actual amount of these proteins in normal and tumor surgical specimens of breast, lung, and esophagus. The application of this methodology affords a straightforward proteomic signature that quantifies the variable energetic demand of human tissues. Furthermore, the results show an unanticipated finding: tumors from different tissues and/or histological types have the same proteomic signature of energetic metabolism. Therefore, the results indicate that cancer abolishes the tissue-specific differences in the bioenergetic phenotype of mitochondria. Overall, the results support that energetic metabolism represents an additional hallmark of the phenotype of the cancer cell and a promising target for the treatment of diverse neoplasias.
Collapse
Affiliation(s)
- Paloma Acebo
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, C.S.I.C.-U.A.M., Universidad Autónoma de Madrid, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28049 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Chen JQ, Cammarata PR, Baines CP, Yager JD. Regulation of mitochondrial respiratory chain biogenesis by estrogens/estrogen receptors and physiological, pathological and pharmacological implications. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1540-70. [PMID: 19559056 DOI: 10.1016/j.bbamcr.2009.06.001] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 12/21/2022]
Abstract
There has been increasing evidence pointing to the mitochondrial respiratory chain (MRC) as a novel and important target for the actions of 17beta-estradiol (E(2)) and estrogen receptors (ER) in a number of cell types and tissues that have high demands for mitochondrial energy metabolism. This novel E(2)-mediated mitochondrial pathway involves the cooperation of both nuclear and mitochondrial ERalpha and ERbeta and their co-activators on the coordinate regulation of both nuclear DNA- and mitochondrial DNA-encoded genes for MRC proteins. In this paper, we have: 1) comprehensively reviewed studies that reveal a novel role of estrogens and ERs in the regulation of MRC biogenesis; 2) discussed their physiological, pathological and pharmacological implications in the control of cell proliferation and apoptosis in relation to estrogen-mediated carcinogenesis, anti-cancer drug resistance in human breast cancer cells, neuroprotection for Alzheimer's disease and Parkinson's disease in brain, cardiovascular protection in human heart and their beneficial effects in lens physiology related to cataract in the eye; and 3) pointed out new research directions to address the key questions in this important and newly emerging area. We also suggest a novel conceptual approach that will contribute to innovative regimens for the prevention or treatment of a wide variety of medical complications based on E(2)/ER-mediated MRC biogenesis pathway.
Collapse
Affiliation(s)
- Jin-Qiang Chen
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | |
Collapse
|
64
|
Seppet E, Gruno M, Peetsalu A, Gizatullina Z, Nguyen HP, Vielhaber S, Wussling MH, Trumbeckaite S, Arandarcikaite O, Jerzembeck D, Sonnabend M, Jegorov K, Zierz S, Striggow F, Gellerich FN. Mitochondria and energetic depression in cell pathophysiology. Int J Mol Sci 2009; 10:2252-2303. [PMID: 19564950 PMCID: PMC2695278 DOI: 10.3390/ijms10052252] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 04/25/2009] [Accepted: 05/14/2009] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial dysfunction is a hallmark of almost all diseases. Acquired or inherited mutations of the mitochondrial genome DNA may give rise to mitochondrial diseases. Another class of disorders, in which mitochondrial impairments are initiated by extramitochondrial factors, includes neurodegenerative diseases and syndromes resulting from typical pathological processes, such as hypoxia/ischemia, inflammation, intoxications, and carcinogenesis. Both classes of diseases lead to cellular energetic depression (CED), which is characterized by decreased cytosolic phosphorylation potential that suppresses the cell's ability to do work and control the intracellular Ca(2+) homeostasis and its redox state. If progressing, CED leads to cell death, whose type is linked to the functional status of the mitochondria. In the case of limited deterioration, when some amounts of ATP can still be generated due to oxidative phosphorylation (OXPHOS), mitochondria launch the apoptotic cell death program by release of cytochrome c. Following pronounced CED, cytoplasmic ATP levels fall below the thresholds required for processing the ATP-dependent apoptotic cascade and the cell dies from necrosis. Both types of death can be grouped together as a mitochondrial cell death (MCD). However, there exist multiple adaptive reactions aimed at protecting cells against CED. In this context, a metabolic shift characterized by suppression of OXPHOS combined with activation of aerobic glycolysis as the main pathway for ATP synthesis (Warburg effect) is of central importance. Whereas this type of adaptation is sufficiently effective to avoid CED and to control the cellular redox state, thereby ensuring the cell survival, it also favors the avoidance of apoptotic cell death. This scenario may underlie uncontrolled cellular proliferation and growth, eventually resulting in carcinogenesis.
Collapse
Affiliation(s)
- Enn Seppet
- Department of Pathophysiology, University of Tartu, Tartu, Estonia; E-Mail:
(M.G.)
| | - Marju Gruno
- Department of Pathophysiology, University of Tartu, Tartu, Estonia; E-Mail:
(M.G.)
| | - Ants Peetsalu
- Department of Surgery, University of Tartu, Tartu, Estonia; E-Mail:
(A.P.)
| | - Zemfira Gizatullina
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Huu Phuc Nguyen
- Department of Medical Genetics, University of Tübingen, Tübingen, Germany; E-Mail:
(H.P.N.)
| | - Stefan Vielhaber
- Department of Neurology, Otto von Guericke University, Magdeburg, Germany; E-Mail:
(S.V.)
| | - Manfred H.P. Wussling
- Bernstein Institute for Physiology, Martin-Luther-University Halle-Wittenberg, Germany; E-Mail:
(M.H.P.W.)
| | - Sonata Trumbeckaite
- Institute for Biomedical Research, Kaunas University of Medicine, Kaunas, Lithuania; E-Mails:
(S.T.);
(O.A.)
| | - Odeta Arandarcikaite
- Institute for Biomedical Research, Kaunas University of Medicine, Kaunas, Lithuania; E-Mails:
(S.T.);
(O.A.)
| | - Doreen Jerzembeck
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Maria Sonnabend
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Katharina Jegorov
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Stephan Zierz
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Germany; E-Mail:
(S.Z.)
| | - Frank Striggow
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| | - Frank N. Gellerich
- KeyNeurotek AG, ZENIT-Technology Park Magdeburg, Magdeburg, Germany; E-Mails:
(Z.G.);
(D.J.);
(M.S.);
(K.J.);
(F.S.);
(F.N.G.)
| |
Collapse
|
65
|
Codarin E, Renzone G, Poz A, Avellini C, Baccarani U, Lupo F, di Maso V, Crocè SL, Tiribelli C, Arena S, Quadrifoglio F, Scaloni A, Tell G. Differential Proteomic Analysis of Subfractioned Human Hepatocellular Carcinoma Tissues. J Proteome Res 2009; 8:2273-84. [DOI: 10.1021/pr8009275] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Erika Codarin
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Giovanni Renzone
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Alessandra Poz
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Claudio Avellini
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Umberto Baccarani
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Francesco Lupo
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Vittorio di Maso
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Saveria Lory Crocè
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Claudio Tiribelli
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Simona Arena
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Franco Quadrifoglio
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Andrea Scaloni
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| | - Gianluca Tell
- Department of Biomedical Sciences and Technologies, University of Udine, 33100 Udine, Italy, Proteomics & Mass Spectrometry Laboratory, ISPAAM, National Research Council, 80147 Naples, Italy, Department of Clinical Pathology, University of Udine, 33100 Udine, Italy, Department of Surgery & Transplantation, University of Udine, 33100 Udine, Italy, Azienda Ospedaliero Universitaria, Molinette, 10100 Torino, Italy, and Centro Studi Fegato, AREA Science Park, 34012 Trieste, Italy
| |
Collapse
|
66
|
Xie H, Valera VA, Merino MJ, Amato AM, Signoretti S, Linehan WM, Sukhatme VP, Seth P. LDH-A inhibition, a therapeutic strategy for treatment of hereditary leiomyomatosis and renal cell cancer. Mol Cancer Ther 2009; 8:626-35. [PMID: 19276158 DOI: 10.1158/1535-7163.mct-08-1049] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The genetic basis for the hereditary leiomyomatosis and renal cell cancer syndrome is germ-line inactivating mutation in the gene for the Krebs/tricarboxylic acid cycle enzyme, fumarate hydratase (FH), the enzyme that converts fumarate to malate. These individuals are predisposed to development of leiomyomas of the skin and uterus as well as highly aggressive kidney cancers. Inhibition of FH should result in significant decrease in oxidative phosphorylation necessitating that glycolysis followed by fermentation of pyruvate to lactate will be required to provide adequate ATP as well as to regenerate NAD+. Moreover, FH deficiency is known to up-regulate expression of hypoxia-inducible factor (HIF)-1alpha by enhancing the stability of HIF transcript. This leads to activation of various HIF-regulated genes including vascular endothelial growth factor and glucose transporter GLUT1 and increased expression of several glycolytic enzymes. Because lactate dehydrogenase-A (LDH-A), also a HIF-1alpha target, promotes fermentative glycolysis (conversion of pyruvate to lactate), a step essential for regenerating NAD+, we asked whether FH-deficient cells would be exquisitely sensitive to LDH-A blockade. Here, we report that hereditary leiomyomatosis and renal cell cancer tumors indeed overexpress LDH-A, that LDH-A inhibition results in increased apoptosis in a cell with FH deficiency and that this effect is reactive oxygen species mediated, and that LDH-A knockdown in the background of FH knockdown results in significant reduction in tumor growth in a xenograft mouse model.
Collapse
Affiliation(s)
- Han Xie
- Department of Medicine, Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Lu YY, Chen TS, Qu JL, Pan WL, Sun L, Wei XB. Dihydroartemisinin (DHA) induces caspase-3-dependent apoptosis in human lung adenocarcinoma ASTC-a-1 cells. J Biomed Sci 2009; 16:16. [PMID: 19272183 PMCID: PMC2653522 DOI: 10.1186/1423-0127-16-16] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Accepted: 02/02/2009] [Indexed: 01/15/2023] Open
Abstract
Background Dihydroartemisinin (DHA), a semi-synthetic derivative of artemisinin, isolated from the traditional Chinese herb Artemisia annua, is recommended as the first-line anti-malarial drug with low toxicity. DHA has been shown to possess promising anticancer activities and induce cancer cell death through apoptotic pathways, although the molecular mechanisms are not well understood. Methods In this study, cell counting kit (CCK-8) assay was employed to evaluate the survival of DHA-treated ASTC-a-1 cells. The induction of apoptosis was detected by Hoechst 33258 and PI staining as well as flow cytometry analysis. Collapse of mitochondrial transmembrane potential (ΔΨm) was measured by dynamic detection under a laser scanning confocal microscope and flow cytometry analysis using Rhodamine123. Caspase-3 activities measured with or without Z-VAD-fmk (a broad spectrum caspase inhibitor) pretreatment by FRET techniques, caspase-3 activity measurement, and western blotting analysis. Results Our results indicated that DHA induced apoptotic cell death in a dose- and time-dependent manner, which was accompanied by mitochondrial morphology changes, the loss of ΔΨm and the activation of caspase-3. Conclusion These results show for the first time that DHA can inhibit proliferation and induce apoptosis via caspase-3-dependent mitochondrial death pathway in ASTC-a-1 cells. Our work may provide evidence for further studies of DHA as a possible anticancer drug in the clinical treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Ying-Ying Lu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, South China Normal University, Guangzhou 510631, PR China.
| | | | | | | | | | | |
Collapse
|
68
|
Cuezva JM, Ortega AD, Willers I, Sánchez-Cenizo L, Aldea M, Sánchez-Aragó M. The tumor suppressor function of mitochondria: translation into the clinics. Biochim Biophys Acta Mol Basis Dis 2009; 1792:1145-58. [PMID: 19419707 DOI: 10.1016/j.bbadis.2009.01.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 12/15/2008] [Accepted: 01/16/2009] [Indexed: 01/30/2023]
Abstract
Recently, the inevitable metabolic reprogramming experienced by cancer cells as a result of the onset of cellular proliferation has been added to the list of hallmarks of the cancer cell phenotype. Proliferation is bound to the synchronous fluctuation of cycles of an increased glycolysis concurrent with a restrained oxidative phosphorylation. Mitochondria are key players in the metabolic cycling experienced during proliferation because of their essential roles in the transduction of biological energy and in defining the life-death fate of the cell. These two activities are molecularly and functionally integrated and are both targets of commonly altered cancer genes. Moreover, energetic metabolism of the cancer cell also affords a target to develop new therapies because the activity of mitochondria has an unquestionable tumor suppressor function. In this review, we summarize most of these findings paying special attention to the opportunity that translation of energetic metabolism into the clinics could afford for the management of cancer patients. More specifically, we emphasize the role that mitochondrial beta-F1-ATPase has as a marker for the prognosis of different cancer patients as well as in predicting the tumor response to therapy.
Collapse
Affiliation(s)
- José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
69
|
Susnow N, Zeng L, Margineantu D, Hockenbery DM. Bcl-2 family proteins as regulators of oxidative stress. Semin Cancer Biol 2008. [PMID: 19138742 DOI: 10.1016/j.semcancer.2008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Bcl-2 family of proteins includes pro- and anti-apoptotic factors acting at mitochondrial and microsomal membranes. An impressive body of published studies, using genetic and physical reconstitution experiments in model organisms and cell lines, supports a view of Bcl-2 proteins as the critical arbiters of apoptotic cell death decisions in most circumstances (excepting CD95 death receptor signaling in Type I cells). Evasion of apoptosis is one of the hallmarks of cancer [Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70], relevant to tumorigenesis as well as resistance to cytotoxic drugs, and deregulation of Bcl-2 proteins is observed in many cancers [Manion MK, Hockenbery DM. Targeting BCL-2-related proteins in cancer therapy. Cancer Biol Ther. 2003;2:S105-14; Olejniczak ET, Van Sant C, Anderson MG, Wang G, Tahir SK, Sauter G, et al. Integrative genomic analysis of small-cell lung carcinoma reveals correlates of sensitivity to bcl-2 antagonists and uncovers novel chromosomal gains. Mol Cancer Res. 2007;5:331-9]. The rekindled interest in aerobic glycolysis as a cancer trait raises interesting questions as to how metabolic changes in cancer cells are integrated with other essential alterations in cancer, e.g. promotion of angiogenesis and unbridled growth signals. Apoptosis induced by multiple different signals involves loss of mitochondrial homeostasis, in particular, outer mitochondrial membrane integrity, releasing cytochrome c and other proteins from the intermembrane space. This integrative process, controlled by Bcl-2 family proteins, is also influenced by the metabolic state of the cell. In this review, we consider the role of reactive oxygen species, a metabolic by-product, in the mitochondrial pathway of apoptosis, and the relationships between Bcl-2 functions and oxidative stress.
Collapse
Affiliation(s)
- Nathan Susnow
- Department of Medicine, University of Washington, Seattle, 98195-6424, United States
| | | | | | | |
Collapse
|
70
|
Bcl-2 family proteins as regulators of oxidative stress. Semin Cancer Biol 2008; 19:42-9. [PMID: 19138742 DOI: 10.1016/j.semcancer.2008.12.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 12/13/2008] [Indexed: 12/22/2022]
Abstract
The Bcl-2 family of proteins includes pro- and anti-apoptotic factors acting at mitochondrial and microsomal membranes. An impressive body of published studies, using genetic and physical reconstitution experiments in model organisms and cell lines, supports a view of Bcl-2 proteins as the critical arbiters of apoptotic cell death decisions in most circumstances (excepting CD95 death receptor signaling in Type I cells). Evasion of apoptosis is one of the hallmarks of cancer [Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70], relevant to tumorigenesis as well as resistance to cytotoxic drugs, and deregulation of Bcl-2 proteins is observed in many cancers [Manion MK, Hockenbery DM. Targeting BCL-2-related proteins in cancer therapy. Cancer Biol Ther. 2003;2:S105-14; Olejniczak ET, Van Sant C, Anderson MG, Wang G, Tahir SK, Sauter G, et al. Integrative genomic analysis of small-cell lung carcinoma reveals correlates of sensitivity to bcl-2 antagonists and uncovers novel chromosomal gains. Mol Cancer Res. 2007;5:331-9]. The rekindled interest in aerobic glycolysis as a cancer trait raises interesting questions as to how metabolic changes in cancer cells are integrated with other essential alterations in cancer, e.g. promotion of angiogenesis and unbridled growth signals. Apoptosis induced by multiple different signals involves loss of mitochondrial homeostasis, in particular, outer mitochondrial membrane integrity, releasing cytochrome c and other proteins from the intermembrane space. This integrative process, controlled by Bcl-2 family proteins, is also influenced by the metabolic state of the cell. In this review, we consider the role of reactive oxygen species, a metabolic by-product, in the mitochondrial pathway of apoptosis, and the relationships between Bcl-2 functions and oxidative stress.
Collapse
|
71
|
Expression of beta-F1-ATPase and mitochondrial transcription factor A and the change in mitochondrial DNA content in colorectal cancer: clinical data analysis and evidence from an in vitro study. Int J Colorectal Dis 2008; 23:1223-32. [PMID: 18769884 DOI: 10.1007/s00384-008-0539-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2008] [Indexed: 02/04/2023]
Abstract
PURPOSE Mitochondria play an important role in regulating apoptosis and thus may be involved in tumor progression. This study was conducted to elucidate the role of mitochondrial dysfunction in colorectal cancer (CRC). METHODS Mitochondrial DNA (mtDNA) content was analyzed with real-time polymerase chain reaction in 153 CRC patients who had received surgery at the Taipei Veterans General Hospital from January 1999 to December 2000. The expression of mitochondrial transcription factor A (TFAM) and beta-F1-ATPase were analyzed using immunohistochemistry. HCT116 cells were cultured in 1% O(2) for at least 20 passages. Mitochondrial biogenesis, ATP production, and the apoptotic response to 5-fluorouracil were analyzed in the derived cells. RESULTS Disease stage was associated with changes in mtDNA content (p < 0.001), expression of TFAM (p = 0.004), and/or beta-F1-ATPase (p < 0.001). CRCs with low expression of TFAM or beta-F1-ATPase had a lower mtDNA content. In the multivariate analysis, disease stage was the most significant prognostic factor [95% confidence interval (CI), 2.82-6.23], followed by beta-F1-ATPase [95% CI, 1.10-4.10]. In patients receiving 5-FU based chemotherapy, the 5-year disease-free survival rate was only 27% in CRC patients with a low beta-F1-ATPase tumor and was significantly lower than that in those with a high beta-F1-ATPase tumor (60%; p = 0.042). In the hypoxia-treated cells, mitochondrial mass increased, mtDNA content decreased, sensitivity to 5-fluorouracil decreased, and beta-F1-ATPase expression decreased. CONCLUSION Mitochondrial dysfunction may be associated with poor outcomes in CRC patients.
Collapse
|
72
|
Ortega AD, Sánchez-Aragó M, Giner-Sánchez D, Sánchez-Cenizo L, Willers I, Cuezva JM. Glucose avidity of carcinomas. Cancer Lett 2008; 276:125-35. [PMID: 18790562 DOI: 10.1016/j.canlet.2008.08.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 07/21/2008] [Accepted: 08/04/2008] [Indexed: 01/12/2023]
Abstract
The cancer cell phenotype has been summarized in six hallmarks [D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (1) (2000) 57-70]. Following the conceptual trait established in that review towards the comprehension of cancer, herein we summarize the basis of an underlying principle that is fulfilled by cancer cells and tumors: its avidity for glucose. Our purpose is to push forward that the metabolic reprogramming that operates in the cancer cell represents a seventh hallmark of the phenotype that offers a vast array of possibilities for the future treatment of the disease. We summarize the metabolic pathways that extract matter and energy from glucose, paying special attention to the concerted regulation of these pathways by the ATP mass-action ratio. The molecular and functional evidences that support the high glucose uptake and the "abnormal" aerobic glycolysis of the carcinomas are detailed discussing also the role that some oncogenes and tumor suppressors have in these pathways. We overview past and present evidences that sustain that mitochondria of the cancer cell are impaired, supporting the original Warburg's formulation that ascribed the high glucose uptake of cancer cells to a defective mitochondria. A simple proteomic approach designed to assess the metabolic phenotype of cancer, i.e., its bioenergetic signature, molecularly and functionally supports Warburg's hypothesis. Furthermore, we discuss the clinical utility that the bioenergetic signature might provide. Glycolysis is presented as the "selfish" pathway used for cellular proliferation, providing both the metabolic precursors and the energy required for biosynthetic purposes, in the context of a plethora of substrates. The glucose avidity of carcinomas is thus presented as the result of both the installment of glycolysis for cellular proliferation and of the impairment of mitochondrial activity in the cancer cell. At the end, the repression of mitochondrial activity affords the cancer cell with a cell-death resistant phenotype making them prone to malignant growth.
Collapse
Affiliation(s)
- Alvaro D Ortega
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Universidad Autónoma de Madrid and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
73
|
Ortega AD, Sala S, Espinosa E, González-Barón M, Cuezva JM. HuR and the bioenergetic signature of breast cancer: a low tumor expression of the RNA-binding protein predicts a higher risk of disease recurrence. Carcinogenesis 2008; 29:2053-61. [PMID: 18687667 DOI: 10.1093/carcin/bgn185] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Downregulation of the catalytic subunit of the mitochondrial H(+)-ATP synthase (beta-F1-ATPase) is a hallmark of many types of cancer. The expression of beta-F1-ATPase is stringently controlled by posttranscriptional mechanisms. Herein, we pursue the identification of beta-F1-ATPase messenger RNA-binding proteins (beta-mRNABPs) that interact and could define the bioenergetic phenotype of the cancer cell in order to establish its relevance as markers of breast cancer progression. RNA immunoprecipitation and RNA affinity chromatography identify HuR as a beta-mRNABP that interacts with the 3'-untranslated region of the transcript. Subcellular fractionation and high-resolution immunoelectron microscopy revealed the cofractionation and presence of HuR in subcellular structures associated to liver mitochondria. Analysis of the expression level of HuR in a cohort of breast carcinomas shows its association with the degree of alteration of the bioenergetic phenotype of the tumor. Moreover, HuR expression is shown to be an independent marker of breast cancer prognosis. A low tumor expression of HuR predicts a higher risk of disease recurrence in early stage breast cancer patients as assessed by clinical and bioenergetic markers of prognosis, strongly supporting the incorporation of HuR as an additional marker for the follow-up of these patients. Mechanistically, overexpression experiments and short hairpin RNA-mediated silencing of HuR in human embryonic kidney and HeLa cells indicate that HuR is not regulating beta-F1-ATPase expression. Overall, the participation of additional RNA-binding proteins in controlling beta-F1-ATPase expression and therefore in defining the bioenergetic signature of the cancer cell is expected.
Collapse
Affiliation(s)
- Alvaro D Ortega
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Spain
| | | | | | | | | |
Collapse
|
74
|
Gasparre G, Hervouet E, de Laplanche E, Demont J, Pennisi LF, Colombel M, Mège-Lechevallier F, Scoazec JY, Bonora E, Smeets R, Smeitink J, Lazar V, Lespinasse J, Giraud S, Godinot C, Romeo G, Simonnet H. Clonal expansion of mutated mitochondrial DNA is associated with tumor formation and complex I deficiency in the benign renal oncocytoma. Hum Mol Genet 2007; 17:986-95. [PMID: 18156159 DOI: 10.1093/hmg/ddm371] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mutations in mitochondrial DNA (mtDNA) are frequent in cancers but it is not yet clearly established whether they are modifier events involved in cancer progression or whether they are a consequence of tumorigenesis. Here we show a benign tumor type in which mtDNA mutations that lead to complex I (CI) enzyme deficiency are found in all tumors and are the only genetic alteration detected. Actually renal oncocytomas are homogeneous tumors characterized by dense accumulation of mitochondria and we had found that they are deficient in electron transport chain complex I (CI, NADH-ubiquinone oxidoreductase). In this work total sequencing of mtDNA showed that 9/9 tumors harbored point mutations in mtDNA, seven in CI genes, one in complex III, and one in the control region. 7/8 mutations were somatic. All tumors were somatically deficient for CI. The clonal amplification of mutated mtDNA in 8/9 tumors demonstrates that these alterations are selected and therefore favor or trigger growth. No nuclear DNA rearrangement was detected beside mtDNA defects. We hypothesize that functional deficiency of the oxidative phosphorylation CI could create a loop of amplification of mitochondria during cell division, impair substrates oxidation and increase intermediary metabolites availability.
Collapse
Affiliation(s)
- Giuseppe Gasparre
- Unità di Genetica Medica, Policlinico Universitario S. Orsola-Malpighi, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Cuezva JM, Sánchez-Aragó M, Sala S, Blanco-Rivero A, Ortega AD. A message emerging from development: the repression of mitochondrial β-F1-ATPase expression in cancer. J Bioenerg Biomembr 2007; 39:259-65. [PMID: 17712532 DOI: 10.1007/s10863-007-9087-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mitochondrial research has experienced a considerable boost during the last decade because organelle malfunctioning is in the genesis and/or progression of a vast array of human pathologies including cancer. The renaissance of mitochondria in the cancer field has been promoted by two main facts: (1) the molecular and functional integration of mitochondrial bioenergetics with the execution of cell death and (2) the implementation of (18)FDG-PET for imaging and staging of tumors in clinical practice. The latter, represents the bed-side translational development of the metabolic hallmark that describes the bioenergetic phenotype of most cancer cells as originally predicted at the beginning of previous century by Otto Warburg. In this minireview we will briefly summarize how the study of energy metabolism during liver development forced our encounter with Warburg's postulates and prompted us to study the mechanisms that regulate the biogenesis of mitochondria in the cancer cell.
Collapse
Affiliation(s)
- José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa", Centro de Investigación Biomédica en Red de Enfermedades Raras, Universidad Autónoma de Madrid, 28049. Madrid, Spain.
| | | | | | | | | |
Collapse
|
76
|
Cannino G, Di Liegro CM, Rinaldi AM. Nuclear-mitochondrial interaction. Mitochondrion 2007; 7:359-66. [PMID: 17822963 DOI: 10.1016/j.mito.2007.07.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 07/24/2007] [Accepted: 07/24/2007] [Indexed: 12/16/2022]
Abstract
The biogenesis of mitochondria depends on the coordinated expression of nuclear and mitochondrial genomes. Consequently, the control of mitochondrial biogenesis and function depends on extremely complex processes requiring a variety of well orchestrated regulatory mechanisms. It is clear that the interplay of transcription factors and coactivators contributes to the expression of both nuclear and mitochondrial respiratory genes. In addition, the regulation of mitochondria biogenesis depends on proteins that, interacting with messenger RNAs for mitochondrial proteins, influence their metabolism and expression. Moreover, a tight regulation of the import and final assembly of mitochondrial protein is essential to endow mitochondria with functional complexes. These studies represent the basis for understanding the mechanisms involved in the nucleus-mitochondrion communication, a cross-talk essential for the cell.
Collapse
Affiliation(s)
- G Cannino
- Dipartimento di Biologia Cellulare e dello Sviluppo A.Monroy, University of Palermo, Italy
| | | | | |
Collapse
|
77
|
Mizumachi T, Suzuki S, Naito A, Carcel-Trullols J, Evans TT, Spring PM, Oridate N, Furuta Y, Fukuda S, Higuchi M. Increased mitochondrial DNA induces acquired docetaxel resistance in head and neck cancer cells. Oncogene 2007; 27:831-8. [PMID: 17637738 PMCID: PMC2268644 DOI: 10.1038/sj.onc.1210681] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Docetaxel is one of the most effective chemotherapeutic agents against cancer; nevertheless, some patients develop resistance. Unfortunately, their causes and mechanisms remain unknown. We created docetaxel-resistant DRHEp2 from human laryngeal cancer HEp2 and investigated the roles of mitochondrial DNA (mtDNA) and reactive oxygen species (ROS) on docetaxel resistance. DRHEp2 had greatly increased mtDNA content. Reduction of mtDNA content in DRHEp2 by ethidium bromide treatment reduced the resistance. These results indicate the possible roles of mtDNA-coded enzymes in mitochondrial respiratory chain (MRC) in resistant mechanisms. Oligomycin A, an Fo-ATPase inhibitor, eliminated docetaxel resistance in DRHEp2; in contrast, inhibitors of other MRC did not. RNA interference targeted to Fo-ATPase d-subunit restored docetaxel-induced cytotoxicity to DRHEp2. These results indicate the roles of Fo-ATPase for resistant mechanisms. Docetaxel induced ROS generation in HEp2 but not in DRHEp2 and antioxidant pyrrolidine dithiocarbamate eliminated docetaxel-induced cytotoxicity, suggesting roles of ROS in docetaxel-induced cell death. Furthermore, inhibition of Fo-ATPase by Oligomycin A induced docetaxel-mediated ROS generation in DRHEp2. Taken together, DRHEp2 acquired docetaxel resistance through increasing Fo-ATPase, which led to diminish docetaxel-induced ROS generation and subsequently inhibited cell death. In conclusion, mtDNA plays an important role in developing docetaxel resistance through the reduction of ROS generation by regulating Fo-ATPase.
Collapse
Affiliation(s)
- T Mizumachi
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - S Suzuki
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Otolaryngology-Head and Neck Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - A Naito
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - J Carcel-Trullols
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - TT Evans
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - PM Spring
- Department of Otolaryngology-Head and Neck Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - N Oridate
- Department of Otolaryngology-Head and Neck Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Y Furuta
- Department of Otolaryngology-Head and Neck Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - S Fukuda
- Department of Otolaryngology-Head and Neck Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - M Higuchi
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
78
|
Zhang H, Gao P, Fukuda R, Kumar G, Krishnamachary B, Zeller KI, Dang CV, Semenza GL. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell 2007; 11:407-20. [PMID: 17482131 DOI: 10.1016/j.ccr.2007.04.001] [Citation(s) in RCA: 678] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Revised: 01/22/2007] [Accepted: 04/02/2007] [Indexed: 02/01/2023]
Abstract
Many cancer cells are characterized by increased glycolysis and decreased respiration, even under aerobic conditions. The molecular mechanisms underlying this metabolic reprogramming are unclear. Here we show that hypoxia-inducible factor 1 (HIF-1) negatively regulates mitochondrial biogenesis and O(2) consumption in renal carcinoma cells lacking the von Hippel-Lindau tumor suppressor (VHL). HIF-1 mediates these effects by inhibiting C-MYC activity via two mechanisms. First, HIF-1 binds to and activates transcription of the MXI1 gene, which encodes a repressor of C-MYC transcriptional activity. Second, HIF-1 promotes MXI-1-independent, proteasome-dependent degradation of C-MYC. We demonstrate that transcription of the gene encoding the coactivator PGC-1beta is C-MYC dependent and that loss of PGC-1beta expression is a major factor contributing to reduced respiration in VHL-deficient renal carcinoma cells.
Collapse
Affiliation(s)
- Huafeng Zhang
- Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Martínez-Diez M, Santamaría G, Ortega ÁD, Cuezva JM. Biogenesis and dynamics of mitochondria during the cell cycle: significance of 3'UTRs. PLoS One 2006; 1:e107. [PMID: 17205111 PMCID: PMC1762426 DOI: 10.1371/journal.pone.0000107] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Accepted: 11/24/2006] [Indexed: 11/18/2022] Open
Abstract
Nowadays, we are facing a renaissance of mitochondria in cancer biology. However, our knowledge of the basic cell biology and on the timing and mechanisms that control the biosynthesis of mitochondrial constituents during progression through the cell cycle of mammalian cells remain largely unknown. Herein, we document the in vivo changes on mitochondrial morphology and dynamics that accompany cellular mitosis, and illustrate the following key points of the biogenesis of mitochondria during progression of liver cells through the cycle: (i) the replication of nuclear and mitochondrial genomes is synchronized during cellular proliferation, (ii) the accretion of OXPHOS proteins is asynchronously regulated during proliferation being the synthesis of beta-F1-ATPase and Hsp60 carried out also at G2/M and, (iii) the biosynthesis of cardiolipin is achieved during the S phase, although full development of the mitochondrial membrane potential (DeltaPsim) is attained at G2/M. Furthermore, we demonstrate using reporter constructs that the mechanism regulating the accretion of beta-F1-ATPase during cellular proliferation is controlled at the level of mRNA translation by the 3'UTR of the transcript. The 3'UTR-driven synthesis of the protein at G2/M is essential for conferring to the daughter cells the original phenotype of the parental cell. Our findings suggest that alterations on this process may promote deregulated beta-F1-ATPase expression in human cancer.
Collapse
|
80
|
Wu M, Neilson A, Swift AL, Moran R, Tamagnine J, Parslow D, Armistead S, Lemire K, Orrell J, Teich J, Chomicz S, Ferrick DA. Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells. Am J Physiol Cell Physiol 2006; 292:C125-36. [PMID: 16971499 DOI: 10.1152/ajpcell.00247.2006] [Citation(s) in RCA: 719] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased conversion of glucose to lactic acid associated with decreased mitochondrial respiration is a unique feature of tumors first described by Otto Warburg in the 1920s. Recent evidence suggests that the Warburg effect is caused by oncogenes and is an underlying mechanism of malignant transformation. Using a novel approach to measure cellular metabolic rates in vitro, the bioenergetic basis of this increased glycolysis and reduced mitochondrial respiration was investigated in two human cancer cell lines, H460 and A549. The bioenergetic phenotype was analyzed by measuring cellular respiration, glycolysis rate, and ATP turnover of the cells in response to various pharmacological modulators. H460 and A549 cells displayed a dependency on glycolysis and an ability to significantly upregulate this pathway when their respiration was inhibited. The converse, however, was not true. The cell lines were attenuated in oxidative phosphorylation (OXPHOS) capacity and were unable to sufficiently upregulate mitochondrial OXPHOS when glycolysis was disabled. This observed mitochondrial impairment was intimately linked to the increased dependency on glycolysis. Furthermore, it was demonstrated that H460 cells were more glycolytic, having a greater impairment of mitochondrial respiration, compared with A549 cells. Finally, the upregulation of glycolysis in response to mitochondrial ATP synthesis inhibition was dependent on AMP-activated protein kinase activity. In summary, our results demonstrate a bioenergetic phenotype of these two cancer cell lines characterized by increased rate of glycolysis and a linked attenuation in their OXPHOS capacity. These metabolic alterations provide a mechanistic explanation for the growth advantage and apoptotic resistance of tumor cells.
Collapse
Affiliation(s)
- Min Wu
- Seahorse Bioscience, 16 Esquire Road, North Billerica, MA 01862, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Pardo J, Urban C, Galvez EM, Ekert PG, Müller U, Kwon-Chung J, Lobigs M, Müllbacher A, Wallich R, Borner C, Simon MM. The mitochondrial protein Bak is pivotal for gliotoxin-induced apoptosis and a critical host factor of Aspergillus fumigatus virulence in mice. ACTA ACUST UNITED AC 2006; 174:509-19. [PMID: 16893972 PMCID: PMC2064257 DOI: 10.1083/jcb.200604044] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aspergillus fumigatus infections cause high levels of morbidity and mortality in immunocompromised patients. Gliotoxin (GT), a secondary metabolite, is cytotoxic for mammalian cells, but the molecular basis and biological relevance of this toxicity remain speculative. We show that GT induces apoptotic cell death by activating the proapoptotic Bcl-2 family member Bak, but not Bax, to elicit the generation of reactive oxygen species, the mitochondrial release of apoptogenic factors, and caspase-3 activation. Activation of Bak by GT is direct, as GT triggers in vitro a dose-dependent release of cytochrome c from purified mitochondria isolated from wild-type and Bax- but not Bak-deficient cells. Resistance to A. fumigatus of mice lacking Bak compared to wild-type mice demonstrates the in vivo relevance of this GT-induced apoptotic pathway involving Bak and suggests a correlation between GT production and virulence. The elucidation of the molecular basis opens new strategies for the development of therapeutic regimens to combat A. fumigatus and related fungal infections.
Collapse
Affiliation(s)
- Julian Pardo
- Metschnikoff Laboratory, Max-Planck-Institut für Immunbiologie, D-79108 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
García-Aguilar A, Cuezva JM. Immunocytochemistry: its applications and drawbacks for the study of gut neuroendocrinology. Front Physiol 1980; 9:1322. [PMID: 30283362 PMCID: PMC6156145 DOI: 10.3389/fphys.2018.01322] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/31/2018] [Indexed: 01/10/2023] Open
Abstract
The ATPase Inhibitory Factor 1 (IF1) is the physiological inhibitor of the mitochondrial ATP synthase. Herein, we summarize the regulation of the expression and activity of IF1 as a main driver of the activity of oxidative phosphorylation (OXPHOS) in mammalian tissues. We emphasize that the expression of IF1, which is a mitochondrial protein with very short half-life, is tissue-specifically expressed and primarily controlled at posttranscriptional levels. Inhibition of the activity of IF1 as inhibitor of the ATP synthase under normal physiological conditions is exerted by phosphorylation of S39 by a cAMP-dependent PKA-like activity of mitochondria in response to different physiological cues. Conditional tissue-specific transgenic mice overexpressing IF1 in colon, or a mutant active version of IF1 (IF1-H49K) in liver or in neurons, revealed the inhibition of the ATP synthase and the reprograming of energy metabolism to an enhanced glycolysis. In the IF1-H49K models, the assembly/activity of complex IV and the superassembly of complex V are also affected. Moreover, the IF1-mediated inhibition of the ATP synthase generates a reactive oxygen species (mtROS) signal that switches on the expression of nuclear genes that facilitate adaptation to a restrained OXPHOS. In contrast to normal mice, metabolically preconditioned animals are partially protected from the action of cytotoxic agents by upgrading the activation of stress kinases and transcription factors involved in resolving metabolic adaptation, the antioxidant response, cell survival, and the immune response of the tissue microenvironment. Altogether, we stress a fundamental physiological function for the ATP synthase and its inhibitor in mitohormesis.
Collapse
|