1
|
Guo L. F-ATP synthase inhibitory factor 1 and mitochondria-organelle interactions: New insight and implications. Pharmacol Res 2024; 208:107393. [PMID: 39233058 DOI: 10.1016/j.phrs.2024.107393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/08/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitochondria are metabolic hub, and act as primary sites for reactive oxygen species (ROS) and metabolites generation. Mitochondrial Ca2+ uptake contributes to Ca2+ storage. Mitochondria-organelle interactions are important for cellular metabolic adaptation, biosynthesis, redox balance, cell fate. Organelle communications are mediated by Ca2+/ROS signals, vesicle transport and membrane contact sites. The permeability transition pore (PTP) is an unselective channel that provides a release pathway for Ca2+/ROS, mtDNA and metabolites. F-ATP synthase inhibitory factor 1 (IF1) participates in regulation of PTP opening and is required for the translocation of transcriptional factors c-Myc/PGC1α to mitochondria to stimulate metabolic switch. IF1, a mitochondrial specific protein, has been suggested to regulate other organelles including nucleus, endoplasmic reticulum and lysosomes. IF1 may be able to mediate mitochondria-organelle interactions and cellular physiology through regulation of PTP activity.
Collapse
Affiliation(s)
- Lishu Guo
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Department of Anesthesiology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
2
|
Alfaro GF, Palombo V, D’Andrea M, Cao W, Zhang Y, Beever JE, Muntifering RB, Pacheco WJ, Rodning SP, Wang X, Moisá SJ. Hepatic transcript profiling in beef cattle: Effects of feeding endophyte-infected tall fescue seeds. PLoS One 2024; 19:e0306431. [PMID: 39058685 PMCID: PMC11280227 DOI: 10.1371/journal.pone.0306431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
The objective of our study was to evaluate the effect of endophyte-infected tall fescue (E+) seeds intake on liver tissue transcriptome in growing Angus × Simmental steers and heifers through RNA-seq analysis. Normal weaned calves (~8 months old) received either endophyte-free tall fescue (E-; n = 3) or infected tall fescue (E+; n = 6) seeds for a 30-d period. The diet offered was ad libitum bermudagrass (Cynodon dactylon) hay combined with a nutritional supplement of 1.61 kg (DM basis) of E+ or E- tall fescue seeds, and 1.61 kg (DM basis) of energy/protein supplement pellets for a 30-d period. Dietary E+ tall fescue seeds were included in a rate of 20 μg of ergovaline/kg BW/day. Liver tissue was individually obtained through biopsy at d 30. After preparation and processing of the liver samples for RNA sequencing, we detected that several metabolic pathways were activated (i.e., upregulated) by the consumption of E+ tall fescue. Among them, oxidative phosphorylation, ribosome biogenesis, protein processing in endoplasmic reticulum and apoptosis, suggesting an active mechanism to cope against impairment in normal liver function. Interestingly, hepatic protein synthesis might increase due to E+ consumption. In addition, there was upregulation of "thermogenesis" KEGG pathway, showing a possible increase in energy expenditure in liver tissue due to consumption of E+ diet. Therefore, results from our study expand the current knowledge related to liver metabolism of growing beef cattle under tall fescue toxicosis.
Collapse
Affiliation(s)
- Gastón F. Alfaro
- Department of Animal Sciences, Auburn University, Auburn, AL, United States of America
| | - Valentino Palombo
- Department of Agricultural, Environmental and Food Sciences, Università degli Studi del Molise, Campobasso, Italy
| | - MariaSilvia D’Andrea
- Department of Agricultural, Environmental and Food Sciences, Università degli Studi del Molise, Campobasso, Italy
| | - Wenqi Cao
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Yue Zhang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Jonathan E. Beever
- Department of Animal Sciences, University of Tennessee, Knoxville, TN, United States of America
| | | | - Wilmer J. Pacheco
- Department of Poultry Sciences, Auburn University, Auburn, AL, United States of America
| | - Soren P. Rodning
- Department of Animal Sciences, Auburn University, Auburn, AL, United States of America
| | - Xu Wang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
| | - Sonia J. Moisá
- Department of Animal Sciences, University of Tennessee, Knoxville, TN, United States of America
| |
Collapse
|
3
|
Nan K, Zhong Z, Yue Y, Zhou W, Sun X, Shen Y, Qu M, Chen Z, Gu J, Sun C, Sun X, Lu L, Zhang J, Miao C, Sun M. HSK3486 Inhibits Colorectal Cancer Growth by Promoting Oxidative Stress and ATPase Inhibitory Factor 1 Activation. Dig Dis Sci 2024; 69:1214-1227. [PMID: 38376789 DOI: 10.1007/s10620-023-08213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/24/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND HSK3486 (ciprofol), a new candidate drug similar to propofol, exerts sedative and hypnotic effects through gamma-aminobutyric acid type A receptors; however, its potential role in colorectal cancer is currently unknown. AIMS This study aimed to evaluate the effects of HSK3486 on colorectal cancer cell proliferation. METHODS Imaging was performed to detect reactive oxygen species and mitochondrial membrane potential. Western blotting was used to determine the expression of target signals. The HSK3486 molecular mechanism was investigated through ATPase inhibitory factor 1 knockdown and xenograft model experiments to assess mitochondrial function in colorectal cancer cells. RESULTS Cell Counting Kit-8 and Annexin V/propidium iodide double staining assays showed that HSK3486 inhibited colorectal cancer cell proliferation in a concentration-dependent manner. In addition, HSK3486 treatment increased the expression of B-cell lymphoma-2-associated X, cleaved caspase 3, and cleaved poly (ADP-ribose) polymerase, whereas myeloid cell leukemia-1 and B-cell lymphoma 2 expression decreased. HSK3486 promoted mitochondrial dysfunction by inducing ATPase inhibitor factor 1 expression. Furthermore, HSK3486 promoted oxidative stress, as shown by the increase in reactive oxygen species and lactate dehydrogenase levels, along with a decrease in mitochondrial membrane potential and ATP levels. ATPase inhibitor factor 1 small interfering RNA pretreatment dramatically increased the mitochondrial membrane potential and tumor size in a xenograft model following exposure to HSK3486. CONCLUSION Collectively, our findings revealed that HSK3486 induces oxidative stress, resulting in colorectal cancer cell apoptosis, making it a potential candidate therapeutic strategy for colorectal cancer.
Collapse
Affiliation(s)
- Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ziwen Zhong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wenchang Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xingfeng Sun
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200438, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhaoyuan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jiahui Gu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Caihong Sun
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xun Sun
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lihong Lu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jie Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Minli Sun
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, No.180 Feng-Lin Road, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
4
|
Matsuyama M, Ortega JT, Fedorov Y, Scott-McKean J, Muller-Greven J, Buck M, Adams D, Jastrzebska B, Greenlee W, Matsuyama S. Development of novel cytoprotective small compounds inhibiting mitochondria-dependent cell death. iScience 2023; 26:107916. [PMID: 37841588 PMCID: PMC10568349 DOI: 10.1016/j.isci.2023.107916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/27/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
We identified cytoprotective small molecules (CSMs) by a cell-based high-throughput screening of Bax inhibitors. Through a medicinal chemistry program, M109S was developed, which is orally bioactive and penetrates the blood-brain/retina barriers. M109S protected retinal cells in ocular disease mouse models. M109S directly interacted with Bax and inhibited the conformational change and mitochondrial translocation of Bax. M109S inhibited ABT-737-induced apoptosis both in Bax-only and Bak-only mouse embryonic fibroblasts. M109S also inhibited apoptosis induced by staurosporine, etoposide, and obatoclax. M109S decreased maximal mitochondrial oxygen consumption rate and reactive oxygen species production, whereas it increased glycolysis. These effects on cellular metabolism may contribute to the cytoprotective activity of M109S. M109S is a novel small molecule protecting cells from mitochondria-dependent apoptosis both in vitro and in vivo. M109S has the potential to become a research tool for studying cell death mechanisms and to develop therapeutics targeting mitochondria-dependent cell death pathway.
Collapse
Affiliation(s)
- Mieko Matsuyama
- Department of Ophthalmology and Visual Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Joseph T. Ortega
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yuri Fedorov
- Department of Genetics and Genome Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jonah Scott-McKean
- Department of Ophthalmology and Visual Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Macromolecular Science and Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jeannie Muller-Greven
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Matthias Buck
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Drew Adams
- Department of Genetics and Genome Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Shigemi Matsuyama
- Department of Ophthalmology and Visual Science, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
5
|
Domínguez-Zorita S, Cuezva JM. The Mitochondrial ATP Synthase/IF1 Axis in Cancer Progression: Targets for Therapeutic Intervention. Cancers (Basel) 2023; 15:3775. [PMID: 37568591 PMCID: PMC10417293 DOI: 10.3390/cancers15153775] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer poses a significant global health problem with profound personal and economic implications on National Health Care Systems. The reprograming of metabolism is a major trait of the cancer phenotype with a clear potential for developing effective therapeutic strategies to combat the disease. Herein, we summarize the relevant role that the mitochondrial ATP synthase and its physiological inhibitor, ATPase Inhibitory Factor 1 (IF1), play in metabolic reprogramming to an enhanced glycolytic phenotype. We stress that the interplay in the ATP synthase/IF1 axis has additional functional roles in signaling mitohormetic programs, pro-oncogenic or anti-metastatic phenotypes depending on the cell type. Moreover, the same axis also participates in cell death resistance of cancer cells by restrained mitochondrial permeability transition pore opening. We emphasize the relevance of the different post-transcriptional mechanisms that regulate the specific expression and activity of ATP synthase/IF1, to stimulate further investigations in the field because of their potential as future targets to treat cancer. In addition, we review recent findings stressing that mitochondria metabolism is the primary altered target in lung adenocarcinomas and that the ATP synthase/IF1 axis of OXPHOS is included in the most significant signature of metastatic disease. Finally, we stress that targeting mitochondrial OXPHOS in pre-clinical mouse models affords a most effective therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28041 Madrid, Spain
| | - José M. Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28041 Madrid, Spain
| |
Collapse
|
6
|
Edwards-Hicks J, Su H, Mangolini M, Yoneten KK, Wills J, Rodriguez-Blanco G, Young C, Cho K, Barker H, Muir M, Guerrieri AN, Li XF, White R, Manasterski P, Mandrou E, Wills K, Chen J, Abraham E, Sateri K, Qian BZ, Bankhead P, Arends M, Gammoh N, von Kriegsheim A, Patti GJ, Sims AH, Acosta JC, Brunton V, Kranc KR, Christophorou M, Pearce EL, Ringshausen I, Finch AJ. MYC sensitises cells to apoptosis by driving energetic demand. Nat Commun 2022; 13:4674. [PMID: 35945217 PMCID: PMC9363429 DOI: 10.1038/s41467-022-32368-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/27/2022] [Indexed: 12/27/2022] Open
Abstract
The MYC oncogene is a potent driver of growth and proliferation but also sensitises cells to apoptosis, which limits its oncogenic potential. MYC induces several biosynthetic programmes and primary cells overexpressing MYC are highly sensitive to glutamine withdrawal suggesting that MYC-induced sensitisation to apoptosis may be due to imbalance of metabolic/energetic supply and demand. Here we show that MYC elevates global transcription and translation, even in the absence of glutamine, revealing metabolic demand without corresponding supply. Glutamine withdrawal from MRC-5 fibroblasts depletes key tricarboxylic acid (TCA) cycle metabolites and, in combination with MYC activation, leads to AMP accumulation and nucleotide catabolism indicative of energetic stress. Further analyses reveal that glutamine supports viability through TCA cycle energetics rather than asparagine biosynthesis and that TCA cycle inhibition confers tumour suppression on MYC-driven lymphoma in vivo. In summary, glutamine supports the viability of MYC-overexpressing cells through an energetic rather than a biosynthetic mechanism.
Collapse
Affiliation(s)
- Joy Edwards-Hicks
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, D-79108, Freiburg, Germany
| | - Huizhong Su
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Maurizio Mangolini
- Wellcome Trust/MRC Cambridge Stem Cell Institute & Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Kubra K Yoneten
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jimi Wills
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Giovanny Rodriguez-Blanco
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Christine Young
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Kevin Cho
- Department of Chemistry and Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Heather Barker
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Morwenna Muir
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Ania Naila Guerrieri
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Xue-Feng Li
- MRC University of Edinburgh Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Rachel White
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Piotr Manasterski
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Elena Mandrou
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Karen Wills
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Jingyu Chen
- Wellcome Trust/MRC Cambridge Stem Cell Institute & Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Emily Abraham
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Kianoosh Sateri
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Bin-Zhi Qian
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
- MRC University of Edinburgh Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Peter Bankhead
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Mark Arends
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Noor Gammoh
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Alex von Kriegsheim
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Gary J Patti
- Department of Chemistry and Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrew H Sims
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Juan Carlos Acosta
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC, Universidad de Cantabria). C/ Albert Einstein 22, Santander, 39011, Spain
| | - Valerie Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Kamil R Kranc
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, EH8 9YL, UK
| | - Maria Christophorou
- Wellcome Trust/MRC Cambridge Stem Cell Institute & Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, D-79108, Freiburg, Germany
- Department of Oncology, The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA
| | - Ingo Ringshausen
- Wellcome Trust/MRC Cambridge Stem Cell Institute & Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, UK
| | - Andrew J Finch
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK.
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
7
|
Killackey SA, Bi Y, Soares F, Hammi I, Winsor NJ, Abdul-Sater AA, Philpott DJ, Arnoult D, Girardin SE. Mitochondrial protein import stress regulates the LC3 lipidation step of mitophagy through NLRX1 and RRBP1. Mol Cell 2022; 82:2815-2831.e5. [PMID: 35752171 DOI: 10.1016/j.molcel.2022.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 12/26/2022]
Abstract
Protein import into mitochondria is a highly regulated process, yet how cells clear mitochondria undergoing dysfunctional protein import remains poorly characterized. Here we showed that mitochondrial protein import stress (MPIS) triggers localized LC3 lipidation. This arm of the mitophagy pathway occurs through the Nod-like receptor (NLR) protein NLRX1 while, surprisingly, without the engagement of the canonical mitophagy protein PINK1. Mitochondrial depolarization, which itself induces MPIS, also required NLRX1 for LC3 lipidation. While normally targeted to the mitochondrial matrix, cytosol-retained NLRX1 recruited RRBP1, a ribosome-binding transmembrane protein of the endoplasmic reticulum, which relocated to the mitochondrial vicinity during MPIS, and the NLRX1/RRBP1 complex in turn controlled the recruitment and lipidation of LC3. Furthermore, NLRX1 controlled skeletal muscle mitophagy in vivo and regulated endurance capacity during exercise. Thus, localization and lipidation of LC3 at the site of mitophagosome formation is a regulated step of mitophagy controlled by NLRX1/RRBP1 in response to MPIS.
Collapse
Affiliation(s)
- Samuel A Killackey
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yuntian Bi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Fraser Soares
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Ikram Hammi
- INSERM U1197, Hôpital Paul Brousse, Bâtiment Lavoisier, 94807 Villejuif Cedex, France
| | - Nathaniel J Winsor
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ali A Abdul-Sater
- School of Kinesiology and Health Science, Muscle Health Research Centre (MHRC), Faculty of Health, York University, Toronto, ON M3J 1P3, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Damien Arnoult
- INSERM U1197, Hôpital Paul Brousse, Bâtiment Lavoisier, 94807 Villejuif Cedex, France
| | - Stephen E Girardin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
8
|
Guo L. Mitochondrial ATP synthase inhibitory factor 1 interacts with the p53-cyclophilin D complex and promotes opening of the permeability transition pore. J Biol Chem 2022; 298:101858. [PMID: 35337801 PMCID: PMC9043413 DOI: 10.1016/j.jbc.2022.101858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 01/17/2023] Open
Abstract
The mitochondrial permeability transition pore (PTP) is a Ca2+-dependent megachannel that plays an important role in mitochondrial physiology and cell fate. Cyclophilin D (CyPD) is a well-characterized PTP regulator, and its binding to the PTP favors pore opening. It has previously been shown that p53 physically interacts with CyPD and opens the PTP during necrosis. Accumulating studies also suggest that the F-ATP synthase contributes to the regulation and formation of the PTP. F-ATP synthase IF1 (mitochondrial ATP synthase inhibitory factor 1) is a natural inhibitor of F-ATP synthase activity; however, whether IF1 participates in the modulation of PTP opening is basically unknown. Here, we demonstrate using calcium retention capacity assay that IF1 overexpression promotes mitochondrial permeability transition via opening of the PTP. Intriguingly, we show that IF1 can interact with the p53-CyPD complex and facilitate cell death. We also demonstrate that the presence of IF1 is necessary for the formation of p53-CyPD complex. Therefore, we suggest that IF1 regulates the PTP via interaction with the p53-CyPD complex, and that IF1 is necessary for the inducing effect of p53-CyPD complex on PTP opening.
Collapse
Affiliation(s)
- Lishu Guo
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
9
|
Carrer A, Laquatra C, Tommasin L, Carraro M. Modulation and Pharmacology of the Mitochondrial Permeability Transition: A Journey from F-ATP Synthase to ANT. Molecules 2021; 26:molecules26216463. [PMID: 34770872 PMCID: PMC8587538 DOI: 10.3390/molecules26216463] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/22/2022] Open
Abstract
The permeability transition (PT) is an increased permeation of the inner mitochondrial membrane due to the opening of the PT pore (PTP), a Ca2+-activated high conductance channel involved in Ca2+ homeostasis and cell death. Alterations of the PTP have been associated with many pathological conditions and its targeting represents an incessant challenge in the field. Although the modulation of the PTP has been extensively explored, the lack of a clear picture of its molecular nature increases the degree of complexity for any target-based approach. Recent advances suggest the existence of at least two mitochondrial permeability pathways mediated by the F-ATP synthase and the ANT, although the exact molecular mechanism leading to channel formation remains elusive for both. A full comprehension of this to-pore conversion will help to assist in drug design and to develop pharmacological treatments for a fine-tuned PT regulation. Here, we will focus on regulatory mechanisms that impinge on the PTP and discuss the relevant literature of PTP targeting compounds with particular attention to F-ATP synthase and ANT.
Collapse
|
10
|
Umbrasas D, Arandarcikaite O, Grigaleviciute R, Stakauskas R, Borutaite V. Neuroprotective Effect of a Novel ATP-Synthase Inhibitor Bedaquiline in Cerebral Ischemia-Reperfusion Injury. Int J Mol Sci 2021; 22:9717. [PMID: 34575875 PMCID: PMC8472139 DOI: 10.3390/ijms22189717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dysfunction during ischemic stroke ultimately manifests as ATP depletion. Mitochondrial ATP synthase upon loss of mitochondrial membrane potential during ischemia rapidly hydrolyses ATP and thus contributes to ATP depletion. Increasing evidence suggests that inhibition of ATP synthase limits ATP depletion and is protective against ischemic tissue damage. Bedaquiline (BDQ) is an anti-microbial agent, approved for clinical use, that inhibits ATP synthase of Mycobacteria; however recently it has been shown to act on mitochondrial ATP synthase, inhibiting both ATP synthesis and hydrolysis in low micromolar concentrations. In this study, we investigated whether preconditioning with BDQ can alleviate ischemia/reperfusion-induced brain injury in Wistar rats after middle cerebral artery occlusion-reperfusion and whether it affects mitochondrial functions. We found that BDQ was effective in limiting necrosis and neurological dysfunction during ischemia-reperfusion. BDQ also caused inhibition of ATPase activity, mild uncoupling of respiration, and stimulated mitochondrial respiration both in healthy and ischemic mitochondria. Mitochondrial calcium retention capacity was unaffected by BDQ preconditioning. We concluded that BDQ has neuroprotective properties associated with its action on mitochondrial respiration and ATPase activity.
Collapse
Affiliation(s)
- Danielius Umbrasas
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (O.A.); (V.B.)
| | - Odeta Arandarcikaite
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (O.A.); (V.B.)
| | - Ramune Grigaleviciute
- Biological Research Center, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (R.G.); (R.S.)
| | - Rimantas Stakauskas
- Biological Research Center, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (R.G.); (R.S.)
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania; (O.A.); (V.B.)
| |
Collapse
|
11
|
Liu X, Chu W, Shang S, Ma L, Jiang C, Ding Y, Wang J, Zhang S, Shao B. Preliminary study on the anti-apoptotic mechanism of Astragaloside IV on radiation-induced brain cells. Int J Immunopathol Pharmacol 2021; 34:2058738420954594. [PMID: 32902354 PMCID: PMC7485151 DOI: 10.1177/2058738420954594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
With multiple targets and low cytotoxicity, natural medicines can be used as potential neuroprotective agents. The increase in oxidative stress levels and inflammatory responses in the brain caused by radiation affects cognitive function and neuronal structure, and ultimately leads to abnormal changes in neurogenesis, differentiation, and apoptosis. Astragaloside Ⅳ (AS-Ⅳ), one of the main active constituents of astragalus, is known for its antioxidant, antihypertensive, antidiabetic, anti-infarction, anti-inflammatory, anti-apoptotic and wound healing, angiogenesis, and other protective effects. In this study, the mechanism of AS-IV against radiation-induced apoptosis of brain cells in vitro and in vivo was explored by radiation modeling, which provided a theoretical basis for the development of anti-radiation Chinese herbal active molecules and brain health products. In order to study the protective mechanism of AS-IV on radiation-induced brain cell apoptosis in mice, the paper constructed a radiation-induced brain cell apoptosis model, using TUNEL staining, flow cytometry, Western blotting to analyze AS-IV resistance mechanism to radiation-induced brain cell apoptosis. The results of TUNEL staining and flow cytometry showed that the apoptosis rate of radiation group was significantly increased. The results of Western blotting indicated that the expression levels of p-JNK, p-p38, p53, Caspase-9 and Caspase-3 protein, and the ratio of Bax to Bcl-2 in radiation group were significantly increased. There was no significant difference in the expression levels of JNK and p38. After AS-IV treatment, the apoptosis was reduced and the expression of apoptosis related proteins was changed. These data suggested that AS-IV can effectively reduce radiation-induced apoptosis of brain cells, and its mechanism may be related to the phosphorylation regulation of JNK-p38.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Weiwei Chu
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Shuying Shang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Liang Ma
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Chenxin Jiang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yanping Ding
- School wof Life Sciences, Northwest Normal University, Lanzhou, Gansu Province, China
| | - Jianlin Wang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Shengxiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| | - Baoping Shao
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
12
|
Mollinedo F, Gajate C. Mitochondrial Targeting Involving Cholesterol-Rich Lipid Rafts in the Mechanism of Action of the Antitumor Ether Lipid and Alkylphospholipid Analog Edelfosine. Pharmaceutics 2021; 13:763. [PMID: 34065546 PMCID: PMC8161315 DOI: 10.3390/pharmaceutics13050763] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
The ether lipid edelfosine induces apoptosis selectively in tumor cells and is the prototypic molecule of a family of synthetic antitumor compounds collectively known as alkylphospholipid analogs. Cumulative evidence shows that edelfosine interacts with cholesterol-rich lipid rafts, endoplasmic reticulum (ER) and mitochondria. Edelfosine induces apoptosis in a number of hematological cancer cells by recruiting death receptors and downstream apoptotic signaling into lipid rafts, whereas it promotes apoptosis in solid tumor cells through an ER stress response. Edelfosine-induced apoptosis, mediated by lipid rafts and/or ER, requires the involvement of a mitochondrial-dependent step to eventually elicit cell death, leading to the loss of mitochondrial membrane potential, cytochrome c release and the triggering of cell death. The overexpression of Bcl-2 or Bcl-xL blocks edelfosine-induced apoptosis. Edelfosine induces the redistribution of lipid rafts from the plasma membrane to the mitochondria. The pro-apoptotic action of edelfosine on cancer cells is associated with the recruitment of F1FO-ATP synthase into cholesterol-rich lipid rafts. Specific inhibition of the FO sector of the F1FO-ATP synthase, which contains the membrane-embedded c-subunit ring that constitutes the mitochondrial permeability transcription pore, hinders edelfosine-induced cell death. Taking together, the evidence shown here suggests that the ether lipid edelfosine could modulate cell death in cancer cells by direct interaction with mitochondria, and the reorganization of raft-located mitochondrial proteins that critically modulate cell death or survival. Here, we summarize and discuss the involvement of mitochondria in the antitumor action of the ether lipid edelfosine, pointing out the mitochondrial targeting of this drug as a major therapeutic approach, which can be extrapolated to other alkylphospholipid analogs. We also discuss the involvement of cholesterol transport and cholesterol-rich lipid rafts in the interactions between the organelles as well as in the role of mitochondria in the regulation of apoptosis in cancer cells and cancer therapy.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, C/Ramiro de Maeztu 9, E-28040 Madrid, Spain;
| | | |
Collapse
|
13
|
Esparza-Moltó PB, Romero-Carramiñana I, Núñez de Arenas C, Pereira MP, Blanco N, Pardo B, Bates GR, Sánchez-Castillo C, Artuch R, Murphy MP, Esteban JA, Cuezva JM. Generation of mitochondrial reactive oxygen species is controlled by ATPase inhibitory factor 1 and regulates cognition. PLoS Biol 2021; 19:e3001252. [PMID: 33983919 PMCID: PMC8148373 DOI: 10.1371/journal.pbio.3001252] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/25/2021] [Accepted: 04/29/2021] [Indexed: 11/18/2022] Open
Abstract
The mitochondrial ATP synthase emerges as key hub of cellular functions controlling the production of ATP, cellular signaling, and fate. It is regulated by the ATPase inhibitory factor 1 (IF1), which is highly abundant in neurons. Herein, we ablated or overexpressed IF1 in mouse neurons to show that IF1 dose defines the fraction of active/inactive enzyme in vivo, thereby controlling mitochondrial function and the production of mitochondrial reactive oxygen species (mtROS). Transcriptomic, proteomic, and metabolomic analyses indicate that IF1 dose regulates mitochondrial metabolism, synaptic function, and cognition. Ablation of IF1 impairs memory, whereas synaptic transmission and learning are enhanced by IF1 overexpression. Mechanistically, quenching the IF1-mediated increase in mtROS production in mice overexpressing IF1 reduces the increased synaptic transmission and obliterates the learning advantage afforded by the higher IF1 content. Overall, IF1 plays a key role in neuronal function by regulating the fraction of ATP synthase responsible for mitohormetic mtROS signaling.
Collapse
Affiliation(s)
- Pau B. Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Inés Romero-Carramiñana
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Cristina Núñez de Arenas
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Marta P. Pereira
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Noelia Blanco
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Beatriz Pardo
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Georgina R. Bates
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Carla Sánchez-Castillo
- Unidad de Neuropatología Molecular, Centro de Biología Molecular Severo Ochoa, Madrid, Spain
| | - Rafael Artuch
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Departamento de Bioquímica Clínica, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - José A. Esteban
- Unidad de Neuropatología Molecular, Centro de Biología Molecular Severo Ochoa, Madrid, Spain
| | - José M. Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
14
|
Guo L. Mitochondria and the permeability transition pore in cancer metabolic reprogramming. Biochem Pharmacol 2021; 188:114537. [PMID: 33811907 DOI: 10.1016/j.bcp.2021.114537] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Mitochondria are a major source of ATP provision as well as cellular suicidal weapon store. Accumulating evidences demonstrate that mitochondrial bioenergetics, biosynthesis and signaling are important mediators of tumorigenesis. Metabolic plasticity enables cancer cell reprogramming to cope with cellular and environmental alterations, a process requires mitochondria biology. Mitochondrial metabolism emerges to be a promising arena for cancer therapeutic targets. The permeability transition pore (PTP) participates in physiological Ca2+ and ROS homeostasis as well as cell death depending on the open state. The hypothesis that PTP forms from F-ATP synthase provides clues to the potential collaborative role of mitochondrial respiration and PTP in regulating cancer cell fate and metabolic reprogramming.
Collapse
Affiliation(s)
- Lishu Guo
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
15
|
Shaw RL, Norton CE, Segal SS. Apoptosis in resistance arteries induced by hydrogen peroxide: greater resilience of endothelium versus smooth muscle. Am J Physiol Heart Circ Physiol 2021; 320:H1625-H1633. [PMID: 33606587 DOI: 10.1152/ajpheart.00956.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Reactive oxygen species (ROS) are implicated in cardiovascular and neurologic disorders including atherosclerosis, heart attack, stroke, and traumatic brain injury. Although oxidative stress can lead to apoptosis of vascular cells, such findings are largely based upon isolated vascular smooth muscle cells (SMCs) and endothelial cells (ECs) studied in culture. Studying intact resistance arteries, we have focused on understanding how SMCs and ECs in the blood vessel wall respond to acute oxidative stress induced by hydrogen peroxide, a ubiquitous, membrane-permeant ROS. We find that apoptosis induced by H2O2 is far greater in SMCs compared to ECs. For both cell types, apoptosis is associated with a rise in intracellular calcium concentration ([Ca2+]i) during H2O2 exposure. Consistent with their greater death, the rise in [Ca2+]i for SMCs exceeds that in ECs. Finding that disruption of the endothelium increases SMC death, we address how myoendothelial coupling and paracrine signaling attenuate apoptosis. Remarkably, conditions associated with chronic oxidative stress (advanced age, Western-style diet) protect SMCs during H2O2 exposure, as does female sex. In light of intracellular Ca2+ handling, we consider how glycolytic versus oxidative pathways for ATP production and changes in mitochondrial structure and function impact cellular resilience to H2O2-induced apoptosis. Gaining new insight into protective signaling within and between SMCs and ECs of the arterial wall can be applied to promote vascular cell survival (and recovery of blood flow) in tissues subjected to acute oxidative stress as occurs during reperfusion following myocardial infarction and thrombotic stroke.
Collapse
Affiliation(s)
- Rebecca L Shaw
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Charles E Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
16
|
Esparza-Moltó PB, Cuezva JM. Reprogramming Oxidative Phosphorylation in Cancer: A Role for RNA-Binding Proteins. Antioxid Redox Signal 2020; 33:927-945. [PMID: 31910046 DOI: 10.1089/ars.2019.7988] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Cancer is a major disease imposing high personal and economic burden draining large part of National Health Care and Research budgets worldwide. In the last decade, research in cancer has underscored the reprogramming of metabolism to an enhanced aerobic glycolysis as a major trait of the cancer phenotype with great potential for targeted therapy. Recent Advances: Mitochondria are essential organelles in metabolic reprogramming for controlling the production of biological energy through oxidative phosphorylation (OXPHOS) and the supply of metabolic precursors that sustain proliferation. In addition, mitochondria are critical hubs that integrate different signaling pathways that control cellular metabolism and cell fate. The mitochondrial ATP synthase plays a fundamental role in OXPHOS and cellular signaling. Critical Issues: This review overviews mitochondrial metabolism and OXPHOS, and the major changes reported in the expression and function of mitochondrial proteins of OXPHOS in oncogenesis and in cellular differentiation. We summarize the prominent role that RNA-binding proteins (RNABPs) play in the sorting and localized translation of nuclear-encoded mRNAs that help define the mitochondrial cell-type-specific phenotype. Moreover, we emphasize the mechanisms that contribute to restrain the activity and expression of the mitochondrial ATP synthase in carcinomas, and illustrate that the dysregulation of proteins that control energy metabolism correlates with patients' survival. Future Directions: Future research should elucidate the mechanisms and RNABPs that promote the specific alterations of the mitochondrial phenotype in carcinomas arising from different tissues with the final aim of developing new therapeutic strategies to treat cancer.
Collapse
Affiliation(s)
- Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
17
|
Kim MS, Gernapudi R, Cedeño YC, Polster BM, Martinez R, Shapiro P, Kesari S, Nurmemmedov E, Passaniti A. Targeting breast cancer metabolism with a novel inhibitor of mitochondrial ATP synthesis. Oncotarget 2020; 11:3863-3885. [PMID: 33196708 PMCID: PMC7597410 DOI: 10.18632/oncotarget.27743] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 01/17/2023] Open
Abstract
Inhibitors of mitochondrial respiration and ATP synthesis may promote the selective killing of respiration-competent cancer cells that are critical for tumor progression. We previously reported that CADD522, a small molecule inhibitor of the RUNX2 transcription factor, has potential for breast cancer treatment. In the current study, we show that CADD522 inhibits mitochondrial oxidative phosphorylation by decreasing the mitochondrial oxygen consumption rate (OCR) and ATP production in human breast cancer cells in a RUNX2-independent manner. The enzyme activity of mitochondrial ATP synthase was inhibited by CADD522 treatment. Importantly, results from cellular thermal shift assays that detect drug-induced protein stabilization revealed that CADD522 interacts with both α and β subunits of the F1-ATP synthase complex. Differential scanning fluorimetry also demonstrated interaction of α subunits of the F1-ATP synthase to CADD522. These results suggest that CADD522 might target the enzymatic F1 subunits in the ATP synthase complex. CADD522 increased the levels of intracellular reactive oxygen species (ROS), which was prevented by MitoQ, a mitochondria-targeted antioxidant, suggesting that cancer cells exposed to CADD522 may elevate ROS from mitochondria. CADD522-increased mitochondrial ROS levels were enhanced by exogenously added pro-oxidants such as hydrogen peroxide or tert-butyl hydroperoxide. Conversely, CADD522-mediated cell growth inhibition was blocked by N-acetyl-l-cysteine, a general ROS scavenger. Therefore, CADD522 may exert its antitumor activity by increasing mitochondrial driven cellular ROS levels. Collectively, our data suggest in vitro proof-of-concept that supports inhibition of mitochondrial ATP synthase and ROS generation as contributors to the effectiveness of CADD522 in suppression of tumor growth.
Collapse
Affiliation(s)
- Myoung Sook Kim
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Ramkishore Gernapudi
- Department of Biochemistry & Molecular Biology and Program in Molecular Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | | | - Brian M. Polster
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Research Health Scientist, The Veteran's Health Administration Research & Development Service (VAMHCS), Baltimore, MD, USA
| | - Ramon Martinez
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Santosh Kesari
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Elmar Nurmemmedov
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Antonino Passaniti
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry & Molecular Biology and Program in Molecular Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
- Research Health Scientist, The Veteran's Health Administration Research & Development Service (VAMHCS), Baltimore, MD, USA
| |
Collapse
|
18
|
Galber C, Acosta MJ, Minervini G, Giorgio V. The role of mitochondrial ATP synthase in cancer. Biol Chem 2020; 401:1199-1214. [PMID: 32769215 DOI: 10.1515/hsz-2020-0157] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022]
Abstract
The mitochondrial ATP synthase is a multi-subunit enzyme complex located in the inner mitochondrial membrane which is essential for oxidative phosphorylation under physiological conditions. In this review, we analyse the enzyme functions involved in cancer progression by dissecting specific conditions in which ATP synthase contributes to cancer development or metastasis. Moreover, we propose the role of ATP synthase in the formation of the permeability transition pore (PTP) as an additional mechanism which controls tumour cell death. We further describe transcriptional and translational modifications of the enzyme subunits and of the inhibitor protein IF1 that may promote adaptations leading to cancer metabolism. Finally, we outline ATP synthase gene mutations and epigenetic modifications associated with cancer development or drug resistance, with the aim of highlighting this enzyme complex as a potential novel target for future anti-cancer therapy.
Collapse
Affiliation(s)
- Chiara Galber
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Manuel Jesus Acosta
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Giovanni Minervini
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Valentina Giorgio
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| |
Collapse
|
19
|
Reconstituting the Mammalian Apoptotic Switch in Yeast. Genes (Basel) 2020; 11:genes11020145. [PMID: 32013249 PMCID: PMC7073680 DOI: 10.3390/genes11020145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/22/2022] Open
Abstract
Proteins of the Bcl-2 family regulate the permeabilization of the mitochondrial outer membrane that represents a crucial irreversible step in the process of induction of apoptosis in mammalian cells. The family consists of both proapoptotic proteins that facilitate the membrane permeabilization and antiapoptotic proteins that prevent it in the absence of an apoptotic signal. The molecular mechanisms, by which these proteins interact with each other and with the mitochondrial membranes, however, remain under dispute. Although yeast do not have apparent homologues of these apoptotic regulators, yeast cells expressing mammalian members of the Bcl-2 family have proved to be a valuable model system, in which action of these proteins can be effectively studied. This review focuses on modeling the activity of proapoptotic as well as antiapoptotic proteins of the Bcl-2 family in yeast.
Collapse
|
20
|
Wang X, Niu J, Li J, Shen X, Shen S, Straubinger RM, Qu J. Temporal Effects of Combined Birinapant and Paclitaxel on Pancreatic Cancer Cells Investigated via Large-Scale, Ion-Current-Based Quantitative Proteomics (IonStar). Mol Cell Proteomics 2018; 17:655-671. [PMID: 29358341 PMCID: PMC5880105 DOI: 10.1074/mcp.ra117.000519] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Indexed: 01/05/2023] Open
Abstract
Despite decades of effort, pancreatic adenocarcinoma (PDAC) remains an intractable clinical challenge. An insufficient understanding of mechanisms underlying tumor cell responses to chemotherapy contributes significantly to the lack of effective treatment regimens. Here, paclitaxel, a first-line chemotherapeutic agent, was observed to interact synergistically with birinapant, a second mitochondrial-derived activator of caspases mimetic. Therefore, we investigated molecular-level drug interaction mechanisms using comprehensive, reproducible, and well-controlled ion-current-based MS1 quantification (IonStar). By analyzing 40 biological samples in a single batch, we compared temporal proteomic responses of PDAC cells treated with birinapant and paclitaxel, alone and combined. Using stringent criteria (e.g. strict false-discovery-rate (FDR) control, two peptides/protein), we quantified 4069 unique proteins confidently (99.8% without any missing data), and 541 proteins were significantly altered in the three treatment groups, with an FDR of <1%. Interestingly, most of these proteins were altered only by combined birinapant/paclitaxel, and these predominantly represented three biological processes: mitochondrial function, cell growth and apoptosis, and cell cycle arrest. Proteins responsible for activation of oxidative phosphorylation, fatty acid β-oxidation, and inactivation of aerobic glycolysis were altered largely by combined birinapant/paclitaxel compared with single drugs, suggesting the Warburg effect, which is critical for survival and proliferation of cancer cells, was alleviated by the combination treatment. Metabolic profiling was performed to confirm substantially greater suppression of the Warburg effect by the combined agents compared with either drug alone. Immunoassays confirmed proteomic data revealing changes in apoptosis/survival signaling pathways, such as inhibition of PI3K/AKT, JAK/STAT, and MAPK/ERK signal transduction, as well as induction of G2/M arrest, and showed the drug combination induced much more apoptosis than did single agents. Overall, this in-depth, large-scale proteomics study provided novel insights into molecular mechanisms underlying synergy of combined birinapant/paclitaxel and describes a proteomics/informatics pipeline that can be applied broadly to the development of cancer drug combination regimens.
Collapse
Affiliation(s)
- Xue Wang
- From the ‡Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
| | - Jin Niu
- ¶Department of Pharmaceutical Sciences
| | - Jun Li
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
| | - Xiaomeng Shen
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ‖Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York 14214
| | - Shichen Shen
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ‖Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York 14214
| | - Robert M Straubinger
- From the ‡Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263;
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ¶Department of Pharmaceutical Sciences
| | - Jun Qu
- From the ‡Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263;
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ¶Department of Pharmaceutical Sciences
| |
Collapse
|
21
|
Complete sequence of the ATP6 and ND3 mitochondrial genes in breast cancer tissue of postmenopausal women with different body mass indexes. Ann Diagn Pathol 2018; 32:23-27. [DOI: 10.1016/j.anndiagpath.2017.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/24/2017] [Accepted: 09/08/2017] [Indexed: 12/11/2022]
|
22
|
Villa-Pulgarín JA, Gajate C, Botet J, Jimenez A, Justies N, Varela-M RE, Cuesta-Marbán Á, Müller I, Modolell M, Revuelta JL, Mollinedo F. Mitochondria and lipid raft-located FOF1-ATP synthase as major therapeutic targets in the antileishmanial and anticancer activities of ether lipid edelfosine. PLoS Negl Trop Dis 2017; 11:e0005805. [PMID: 28829771 PMCID: PMC5568728 DOI: 10.1371/journal.pntd.0005805] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/13/2017] [Indexed: 11/18/2022] Open
Abstract
Background Leishmaniasis is the world’s second deadliest parasitic disease after malaria, and current treatment of the different forms of this disease is far from satisfactory. Alkylphospholipid analogs (APLs) are a family of anticancer drugs that show antileishmanial activity, including the first oral drug (miltefosine) for leishmaniasis and drugs in preclinical/clinical oncology trials, but their precise mechanism of action remains to be elucidated. Methodology/Principal findings Here we show that the tumor cell apoptosis-inducer edelfosine was the most effective APL, as compared to miltefosine, perifosine and erucylphosphocholine, in killing Leishmania spp. promastigotes and amastigotes as well as tumor cells, as assessed by DNA breakdown determined by flow cytometry. In studies using animal models, we found that orally-administered edelfosine showed a potent in vivo antileishmanial activity and diminished macrophage pro-inflammatory responses. Edelfosine was also able to kill Leishmania axenic amastigotes. Edelfosine was taken up by host macrophages and killed intracellular Leishmania amastigotes in infected macrophages. Edelfosine accumulated in tumor cell mitochondria and Leishmania kinetoplast-mitochondrion, and led to mitochondrial transmembrane potential disruption, and to the successive breakdown of parasite mitochondrial and nuclear DNA. Ectopic expression of Bcl-XL inhibited edelfosine-induced cell death in both Leishmania parasites and tumor cells. We found that the cytotoxic activity of edelfosine against Leishmania parasites and tumor cells was associated with a dramatic recruitment of FOF1-ATP synthase into lipid rafts following edelfosine treatment in both parasites and cancer cells. Raft disruption and specific FOF1-ATP synthase inhibition hindered edelfosine-induced cell death in both Leishmania parasites and tumor cells. Genetic deletion of FOF1-ATP synthase led to edelfosine drug resistance in Saccharomyces cerevisiae yeast. Conclusions/Significance The present study shows that the antileishmanial and anticancer actions of edelfosine share some common signaling processes, with mitochondria and raft-located FOF1-ATP synthase being critical in the killing process, thus identifying novel druggable targets for the treatment of leishmaniasis. Leishmaniasis is a major health problem worldwide, and can result in loss of human life or a lifelong stigma because of bodily scars. According to World Health Organization, leishmaniasis is considered as an emerging and uncontrolled disease, and its current treatment is far from ideal, with only a few drugs available that could lead to drug resistance or cause serious side-effects. Here, we have found that mitochondria and raft-located FOF1-ATPase synthase are efficient druggable targets, through which an ether lipid named edelfosine exerts its antileishmanial action. Edelfosine effectively kills Leishmania spp. promastigotes and amastigotes. Our experimental animal models demonstrate that oral administration of edelfosine exerts a potent antileishmanial activity, while inhibits macrophage pro-inflammatory responses. Our results show that both Leishmania and tumor cells share mitochondria and raft-located FOF1-ATPase synthase as major druggable targets in leishmaniasis and cancer therapy. These data, showing a potent antileishmanial activity of edelfosine and unveiling its mechanism of action, together with the inhibition of the inflammatory responses elicited by macrophages, suggest that the ether lipid edelfosine is a promising oral drug for leishmaniasis, and highlight mitochondria and lipid raft-located FOF1-ATP synthase as major therapeutic targets for the treatment of this disease.
Collapse
Affiliation(s)
- Janny A Villa-Pulgarín
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Consuelo Gajate
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Javier Botet
- Metabolic Engineering Group, Departamento de Microbiología y Genética, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Alberto Jimenez
- Metabolic Engineering Group, Departamento de Microbiología y Genética, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Nicole Justies
- Department of Cellular Immunology, Max-Planck-Institut für Immunbiologie und Epigenetik, Freiburg, Germany
| | - Rubén E Varela-M
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Álvaro Cuesta-Marbán
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Ingrid Müller
- Department of Medicine, Section of Immunology, St. Mary's Campus, Imperial College London, London, United Kingdom
| | - Manuel Modolell
- Department of Cellular Immunology, Max-Planck-Institut für Immunbiologie und Epigenetik, Freiburg, Germany
| | - José L Revuelta
- Metabolic Engineering Group, Departamento de Microbiología y Genética, Universidad de Salamanca, Edificio Departamental, Campus Miguel de Unamuno, Salamanca, Spain
| | - Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain.,Laboratory of Cell Death and Cancer Therapy, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
23
|
Han W, Xu Y, Feng X, Liang YX, Huang L, Shen Y, She Q. NQO-Induced DNA-Less Cell Formation Is Associated with Chromatin Protein Degradation and Dependent on A 0A 1-ATPase in Sulfolobus. Front Microbiol 2017; 8:1480. [PMID: 28855893 PMCID: PMC5557786 DOI: 10.3389/fmicb.2017.01480] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/24/2017] [Indexed: 12/29/2022] Open
Abstract
To investigate DNA damage response in the model crenarchaeon Sulfolobus islandicus, four different DNA damage agents were tested for their effects on cell death of this archaeon, including UV irradiation, methyl methanesulfonate, cisplatin, and 4-nitroquinoline 1-oxide (NQO). Cell death featured with DNA-less cell formation was revealed in DNA damage treatment with each agent. Cellular responses upon NQO treatment were characterized in details, and following sequential events were revealed, including: a modest accumulation of G1/S phase cells, membrane depolarization, proteolytic degradation of chromatin proteins, and chromosomal DNA degradation. Further insights into the process were gained from studying drugs that affect the archaeal ATP synthase, including a proton gradient uncoupler and an ATP synthase inhibitor. Whereas the proton uncoupler-mediated excess proton influx yielded cell death as observed for the NQO treatment, inhibition of ATP synthase attenuated NQO-induced membrane depolarization and DNA-less cell formation. In conclusion, the NQO-induced cell death in S. islandicus is characterized by proteolytic degradation of chromatin protein, and chromosomal DNA degradation, which probably represents a common feature for the cell death induced by different DNA damage agents.
Collapse
Affiliation(s)
- Wenyuan Han
- Archaea Centre, Department of Biology, University of CopenhagenCopenhagen, Denmark
| | - Yanqun Xu
- Archaea Centre, Department of Biology, University of CopenhagenCopenhagen, Denmark.,State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Huazhong Agricultural UniversityWuhan, China
| | - Xu Feng
- Archaea Centre, Department of Biology, University of CopenhagenCopenhagen, Denmark.,State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Huazhong Agricultural UniversityWuhan, China
| | - Yun X Liang
- State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Huazhong Agricultural UniversityWuhan, China
| | - Li Huang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijing, China
| | - Yulong Shen
- State Key Laboratory of Microbial Technology, Shandong UniversityJinan, China
| | - Qunxin She
- Archaea Centre, Department of Biology, University of CopenhagenCopenhagen, Denmark.,State Key Laboratory of Agricultural Microbiology and College of Life Science and Technology, Huazhong Agricultural UniversityWuhan, China
| |
Collapse
|
24
|
Canto P, Benítez Granados J, Martínez Ramírez MA, Reyes E, Feria-Bernal G, García-García E, Tejeda ME, Zavala E, Tapia A, Rojano-Mejía D, Méndez JP. Genetic variants in ATP6 and ND3 mitochondrial genes are not associated with aggressive prostate cancer in Mexican-Mestizo men with overweight or obesity. Aging Male 2016; 19:187-191. [PMID: 27187822 DOI: 10.1080/13685538.2016.1185409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Mitochondrial defects have been related to obesity and prostate cancer. We investigated if Mexican-Mestizo men presenting this type of cancer, exhibited somatic mutations of ATP6 and/or ND3.Body mass index (BMI) was determined; the degree of prostate cancer aggressiveness was demarcated by the Gleason score. DNA from tumor tissue and from blood leukocytes was amplified by the polymerase chain reaction and ATP6 and ND3 were sequenced. We included 77 men: 20 had normal BMI, 38 were overweight and 19 had obesity; ages ranged from 52 to 83. After sequencing ATP6 and ND3, from DNA obtained from leukocytes and tumor tissue, we did not find any somatic mutations. All changes observed, in both genes, were polymorphisms. In ATP6 we identified, in six patients, two non-synonymous nucleotide changes and in ND3 we observed that twelve patients presented non-synonymous polymorphisms. To our knowledge, this constitutes the first report where the complete sequences of the ATP6 and ND3 have been analyzed in Mexican-Mestizo men with prostate cancer and diverse BMI. Our results differ with those reported in Caucasian populations, possibly due to ethnic differences.
Collapse
Affiliation(s)
- Patricia Canto
- a Facultad de Medicina , Unidad de Investigación en Obesidad, Universidad Nacional Autónoma de México , México, D.F , México
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - Jesús Benítez Granados
- a Facultad de Medicina , Unidad de Investigación en Obesidad, Universidad Nacional Autónoma de México , México, D.F , México
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - Mónica Adriana Martínez Ramírez
- a Facultad de Medicina , Unidad de Investigación en Obesidad, Universidad Nacional Autónoma de México , México, D.F , México
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - Edgardo Reyes
- c Departamento de Patología , Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - Guillermo Feria-Bernal
- d Departamento de Urología , Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México , and
| | - Eduardo García-García
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - María Elena Tejeda
- a Facultad de Medicina , Unidad de Investigación en Obesidad, Universidad Nacional Autónoma de México , México, D.F , México
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - Esperanza Zavala
- a Facultad de Medicina , Unidad de Investigación en Obesidad, Universidad Nacional Autónoma de México , México, D.F , México
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - André Tapia
- a Facultad de Medicina , Unidad de Investigación en Obesidad, Universidad Nacional Autónoma de México , México, D.F , México
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| | - David Rojano-Mejía
- e Unidad de Medicina Física y Rehabilitación Centro, UMAE, Hospital de Traumatología y Ortopedia "Lomas Verdes", Instituto Mexicano del Seguro Social , México, D.F , México
| | - Juan Pablo Méndez
- a Facultad de Medicina , Unidad de Investigación en Obesidad, Universidad Nacional Autónoma de México , México, D.F , México
- b Clínica de Obesidad, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" , México, D.F , México
| |
Collapse
|
25
|
Avcilar T, Kirac D, Ergec D, Koc G, Ulucan K, Kaya Z, Kaspar EC, Turkeri L, Guney AI. Investigation of the association between mitochondrial DNA and p53 gene mutations in transitional cell carcinoma of the bladder. Oncol Lett 2016; 12:2872-2879. [PMID: 27698873 DOI: 10.3892/ol.2016.5000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 04/29/2016] [Indexed: 12/19/2022] Open
Abstract
Bladder carcinoma is the most common malignancy of the urinary tract. The major aim of the present study is to investigate the association between mitochondrial DNA (mtDNA) and p53 gene mutations in bladder carcinoma. A total of 30 patients with transitional cell carcinoma and 27 controls were recruited for the study. Bladder cancer tissues were obtained by radical cystectomy or transurethral resection. Genomic DNA was extracted from peripheral blood. mtDNA and p53 genes were amplified by polymerase chain reaction and sequenced directly. A total of 37 polymorphisms were identified, among which, 2 mutations were significant in the patient group, and 1 mutation was significant in the control group. Additionally, 5 different moderate positive correlations between mtDNA mutations and 3 different positive correlations between p53 gene and mtDNA mutations were detected. The high incidence of mtDNA and p53 gene mutations in bladder cancer suggests that these genes could be important in carcinogenesis.
Collapse
Affiliation(s)
- Tuba Avcilar
- Department of Medical Genetics, Faculty of Medicine, Marmara University, Istanbul 34890, Turkey
| | - Deniz Kirac
- Department of Medical Biology, Faculty of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Deniz Ergec
- Department of Medical Genetics, Faculty of Medicine, Marmara University, Istanbul 34890, Turkey
| | - Gulsah Koc
- Department of Medical Biology, Faculty of Medicine, Istanbul Aydın University, Istanbul 34295, Turkey
| | - Korkut Ulucan
- Department of Medical Biology and Genetics, Faculty of Dentistry, Marmara University, Istanbul 34854, Turkey
| | - Zehra Kaya
- Department of Medical Biology, Faculty of Medicine, Yuzuncu Yıl University, Van 65080, Turkey
| | - Elif Cigdem Kaspar
- Department of Biostatistics, Faculty of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Levent Turkeri
- Department of Urology, Faculty of Medicine, Marmara University, Istanbul 34890, Turkey
| | - Ahmet Ilter Guney
- Department of Medical Genetics, Faculty of Medicine, Marmara University, Istanbul 34890, Turkey
| |
Collapse
|
26
|
García-Bermúdez J, Cuezva JM. The ATPase Inhibitory Factor 1 (IF1): A master regulator of energy metabolism and of cell survival. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1857:1167-1182. [PMID: 26876430 DOI: 10.1016/j.bbabio.2016.02.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/28/2016] [Accepted: 02/07/2016] [Indexed: 12/19/2022]
Abstract
In this contribution we summarize most of the findings reported for the molecular and cellular biology of the physiological inhibitor of the mitochondrial H(+)-ATP synthase, the engine of oxidative phosphorylation (OXPHOS) and gate of cell death. We first describe the structure and major mechanisms and molecules that regulate the activity of the ATP synthase placing the ATPase Inhibitory Factor 1 (IF1) as a major determinant in the regulation of the activity of the ATP synthase and hence of OXPHOS. Next, we summarize the post-transcriptional mechanisms that regulate the expression of IF1 and emphasize, in addition to the regulation afforded by the protonation state of histidine residues, that the activity of IF1 as an inhibitor of the ATP synthase is also regulated by phosphorylation of a serine residue. Phosphorylation of S39 in IF1 by the action of a mitochondrial cAMP-dependent protein kinase A hampers its interaction with the ATP synthase, i.e., only dephosphorylated IF1 interacts with the enzyme. Upon IF1 interaction with the ATP synthase both the synthetic and hydrolytic activities of the engine of OXPHOS are inhibited. These findings are further placed into the physiological context to stress the emerging roles played by IF1 in metabolic reprogramming in cancer, in hypoxia and in cellular differentiation. We review also the implication of IF1 in other cellular situations that involve the malfunctioning of mitochondria. Special emphasis is given to the role of IF1 as driver of the generation of a reactive oxygen species signal that, emanating from mitochondria, is able to reprogram the nucleus of the cell to confer by various signaling pathways a cell-death resistant phenotype against oxidative stress. Overall, our intention is to highlight the urgent need of further investigations in the molecular and cellular biology of IF1 and of its target, the ATP synthase, to unveil new therapeutic strategies in human pathology. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.
Collapse
Affiliation(s)
- Javier García-Bermúdez
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049, Madrid, Spain.
| |
Collapse
|
27
|
Andrés MT, Acosta-Zaldívar M, Fierro JF. Antifungal Mechanism of Action of Lactoferrin: Identification of H+-ATPase (P3A-Type) as a New Apoptotic-Cell Membrane Receptor. Antimicrob Agents Chemother 2016; 60:4206-16. [PMID: 27139463 PMCID: PMC4914641 DOI: 10.1128/aac.03130-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/27/2016] [Indexed: 11/20/2022] Open
Abstract
Human lactoferrin (hLf) is a protein of the innate immune system which induces an apoptotic-like process in yeast. Determination of the susceptibility to lactoferrin of several yeast species under different metabolic conditions, respiratory activity, cytoplasmic ATP levels, and external medium acidification mediated by glucose assays suggested plasma membrane Pma1p (P3A-type ATPase) as the hLf molecular target. The inhibition of plasma membrane ATPase activity by hLf and the identification of Pma1p as the hLf-binding membrane protein confirmed the previous physiological evidence. Consistent with this, cytoplasmic ATP levels progressively increased in hLf-treated Candida albicans cells. However, oligomycin, a specific inhibitor of the mitochondrial F-type ATPase proton pump (mtATPase), abrogated the antifungal activity of hLf, indicating a crucial role for mtATPase in the apoptotic process. We suggest that lactoferrin targeted plasma membrane Pma1p H(+)-ATPase, perturbing the cytoplasmic ion homeostasis (i.e., cytoplasmic H(+) accumulation and subsequent K(+) efflux) and inducing a lethal mitochondrial dysfunction. This initial event involved a normal mitochondrial ATP synthase activity responsible for both the ATP increment and subsequent hypothetical mitochondrial proton flooding process. We conclude that human lactoferrin inhibited Pma1p H(+)-ATPase, inducing an apoptotic-like process in metabolically active yeast. Involvement of mitochondrial H(+)-ATPase (nonreverted) was essential for the progress of this programmed cell death in which the ionic homeostasis perturbation seems to precede classical nonionic apoptotic events.
Collapse
Affiliation(s)
- María T Andrés
- Laboratory of Oral Microbiology, School of Stomatology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | - Maikel Acosta-Zaldívar
- Laboratory of Oral Microbiology, School of Stomatology, Faculty of Medicine, University of Oviedo, Oviedo, Spain Department of Functional Biology (Microbiology), Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | - José F Fierro
- Laboratory of Oral Microbiology, School of Stomatology, Faculty of Medicine, University of Oviedo, Oviedo, Spain Department of Functional Biology (Microbiology), Faculty of Medicine, University of Oviedo, Oviedo, Spain
| |
Collapse
|
28
|
Santacatterina F, Sánchez-Cenizo L, Formentini L, Mobasher MA, Casas E, Rueda CB, Martínez-Reyes I, de Arenas CN, García-Bermúdez J, Zapata JM, Sánchez-Aragó M, Satrústegui J, Valverde ÁM, Cuezva JM. Down-regulation of oxidative phosphorylation in the liver by expression of the ATPase inhibitory factor 1 induces a tumor-promoter metabolic state. Oncotarget 2016; 7:490-508. [PMID: 26595676 PMCID: PMC4808013 DOI: 10.18632/oncotarget.6357] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/14/2015] [Indexed: 02/07/2023] Open
Abstract
The ATPase Inhibitory Factor 1 (IF1) is an inhibitor of the mitochondrial H+-ATP synthase that regulates the activity of both oxidative phosphorylation (OXPHOS) and cell death. Here, we have developed transgenic Tet-On and Tet-Off mice that express a mutant active form of hIF1 in the hepatocytes to restrain OXPHOS in the liver to investigate the relevance of mitochondrial activity in hepatocarcinogenesis. The expression of hIF1 promotes the inhibition of OXPHOS in both Tet-On and Tet-Off mouse models and induces a state of metabolic preconditioning guided by the activation of the stress kinases AMPK and p38 MAPK. Expression of the transgene significantly augmented proliferation and apoptotic resistance of carcinoma cells, which contributed to an enhanced diethylnitrosamine-induced liver carcinogenesis. Moreover, the expression of hIF1 also diminished acetaminophen-induced apoptosis, which is unrelated to differences in permeability transition pore opening. Mechanistically, cell survival in hIF1-preconditioned hepatocytes results from a nuclear factor-erythroid 2-related factor (Nrf2)-guided antioxidant response. The results emphasize in vivo that a metabolic phenotype with a restrained OXPHOS in the liver is prone to the development of cancer.
Collapse
Affiliation(s)
- Fulvio Santacatterina
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| | - Laura Sánchez-Cenizo
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| | - Laura Formentini
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| | - Maysa A. Mobasher
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Estela Casas
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| | - Carlos B. Rueda
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Inmaculada Martínez-Reyes
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| | - Cristina Núñez de Arenas
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Javier García-Bermúdez
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| | - Juan M. Zapata
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
| | - María Sánchez-Aragó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| | - Jorgina Satrústegui
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Ángela M. Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - José M. Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Centro de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain
| |
Collapse
|
29
|
Palchaudhuri R, Lambrecht MJ, Botham RC, Partlow KC, van Ham TJ, Putt KS, Nguyen LT, Kim SH, Peterson RT, Fan TM, Hergenrother PJ. A Small Molecule that Induces Intrinsic Pathway Apoptosis with Unparalleled Speed. Cell Rep 2015; 13:2027-36. [PMID: 26655912 DOI: 10.1016/j.celrep.2015.10.042] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/08/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022] Open
Abstract
Apoptosis is generally believed to be a process that requires several hours, in contrast to non-programmed forms of cell death that can occur in minutes. Our findings challenge the time-consuming nature of apoptosis as we describe the discovery and characterization of a small molecule, named Raptinal, which initiates intrinsic pathway caspase-dependent apoptosis within minutes in multiple cell lines. Comparison to a mechanistically diverse panel of apoptotic stimuli reveals that Raptinal-induced apoptosis proceeds with unparalleled speed. The rapid phenotype enabled identification of the critical roles of mitochondrial voltage-dependent anion channel function, mitochondrial membrane potential/coupled respiration, and mitochondrial complex I, III, and IV function for apoptosis induction. Use of Raptinal in whole organisms demonstrates its utility for studying apoptosis in vivo for a variety of applications. Overall, rapid inducers of apoptosis are powerful tools that will be used in a variety of settings to generate further insight into the apoptotic machinery.
Collapse
Affiliation(s)
- Rahul Palchaudhuri
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Michael J Lambrecht
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rachel C Botham
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kathryn C Partlow
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tjakko J van Ham
- Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Karson S Putt
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Laurie T Nguyen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Seok-Ho Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Randall T Peterson
- Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
30
|
Abstract
Permeabilization of the outer mitochondrial membrane that leads to the release of cytochrome c and several other apoptogenic proteins from mitochondria into cytosol represents a commitment point of apoptotic pathway in mammalian cells. This crucial event is governed by proteins of the Bcl-2 family. Molecular mechanisms, by which Bcl-2 family proteins permeabilize mitochondrial membrane, remain under dispute. Although yeast does not have apparent homologues of these proteins, when mammalian members of Bcl-2 family are expressed in yeast, they retain their activity, making yeast an attractive model system, in which to study their action. This review focuses on using yeast expressing mammalian proteins of the Bcl-2 family as a tool to investigate mechanisms, by which these proteins permeabilize mitochondrial membranes, mechanisms, by which pro- and antiapoptotic members of this family interact, and involvement of other cellular components in the regulation of programmed cell death by Bcl-2 family proteins.
Collapse
Affiliation(s)
- Peter Polčic
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Petra Jaká
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Marek Mentel
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
31
|
Martínez-Reyes I, Cuezva JM. The H+-ATP synthase: A gate to ROS-mediated cell death or cell survival. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2014; 1837:1099-112. [DOI: 10.1016/j.bbabio.2014.03.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/03/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022]
|
32
|
Formentini L, Pereira MP, Sánchez-Cenizo L, Santacatterina F, Lucas JJ, Navarro C, Martínez-Serrano A, Cuezva JM. In vivo inhibition of the mitochondrial H+-ATP synthase in neurons promotes metabolic preconditioning. EMBO J 2014; 33:762-78. [PMID: 24521670 PMCID: PMC4000092 DOI: 10.1002/embj.201386392] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 01/08/2014] [Accepted: 01/13/2014] [Indexed: 12/25/2022] Open
Abstract
A key transducer in energy conservation and signaling cell death is the mitochondrial H(+)-ATP synthase. The expression of the ATPase inhibitory factor 1 (IF1) is a strategy used by cancer cells to inhibit the activity of the H(+)-ATP synthase to generate a ROS signal that switches on cellular programs of survival. We have generated a mouse model expressing a mutant of human IF1 in brain neurons to assess the role of the H(+)-ATP synthase in cell death in vivo. The expression of hIF1 inhibits the activity of oxidative phosphorylation and mediates the shift of neurons to an enhanced aerobic glycolysis. Metabolic reprogramming induces brain preconditioning affording protection against quinolinic acid-induced excitotoxicity. Mechanistically, preconditioning involves the activation of the Akt/p70S6K and PARP repair pathways and Bcl-xL protection from cell death. Overall, our findings provide the first in vivo evidence highlighting the H(+)-ATP synthase as a target to prevent neuronal cell death.
Collapse
Affiliation(s)
- Laura Formentini
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| | - Marta P Pereira
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
| | - Laura Sánchez-Cenizo
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| | - Fulvio Santacatterina
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| | - José J Lucas
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadrid, Spain
| | - Carmen Navarro
- Departamento de Patología y Neuropatología, Instituto de Investigación Biomédica de Vigo (IBIV)Vigo, Spain
| | - Alberto Martínez-Serrano
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| |
Collapse
|
33
|
Ghaffarpour M, Mahdian R, Fereidooni F, Kamalidehghan B, Moazami N, Houshmand M. The mitochondrial ATPase6 gene is more susceptible to mutation than the ATPase8 gene in breast cancer patients. Cancer Cell Int 2014; 14:21. [PMID: 24588805 PMCID: PMC3942513 DOI: 10.1186/1475-2867-14-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 02/20/2014] [Indexed: 12/15/2022] Open
Abstract
Background Breast cancer is the most common malignancy in women throughout the world. Mitochondria play important roles in cellular energy production, free radical generation and apoptosis. Identification of mitochondrial DNA mutations and/or polymorphisms as cancer biomarkers is rapidly developing in molecular oncology research. Methods In this study, the DNA alterations of the mitochondrial ATPase 6 and 8 genes were investigated in 49 breast cancer patients using PCR amplification and direct DNA sequencing on mtDNA. A possible association between these variants and tumorigenesis was assessed. Furthermore, the impact of non-synonymous substitutions on the amino acid sequence was evaluated using the PolyPhen-2 software. Results Twenty eight distinct somatic mitochondrial DNA variants were detected in tumor tissues but not in the corresponding adjacent non-tumor tissues. Among these variants, 9 were observed for the first time in breast cancer patients. The mtDNA variants of A8384 (T7A), T8567C (I14T), G8572A (G16S), A9041G (H172R) and G9055A (A177T) showed the most significant effects probably due to damaging changes to the resulting protein. Furthermore, non-synonymous amino acid changing variants were more frequent in the ATPase6 gene compared to the ATPase8 gene. Conclusion Our results showed that the ATPase6 gene is more susceptible to variations in breast cancer and may play an important role in tumorigenesis by changing the energy metabolism level in cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Massoud Houshmand
- Medical Genetics Department, National Institute for Genetic Engineering & Biotechnology, Tehran, Iran.
| |
Collapse
|
34
|
Tapia-Limonchi R, Díaz I, Cahuana GM, Bautista M, Martín F, Soria B, Tejedo JR, Bedoya FJ. Impact of exposure to low concentrations of nitric oxide on protein profile in murine and human pancreatic islet cells. Islets 2014; 6:e995997. [PMID: 25658244 PMCID: PMC4398281 DOI: 10.1080/19382014.2014.995997] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Homeostatic levels of nitric oxide (NO) protect efficiently against apoptotic death in both human and rodent pancreatic β cells, but the protein profile of this action remains to be determined. We have applied a 2 dimensional LC-MS-MALDI-TOF/TOF-based analysis to study the impact of protective NO in rat insulin-producing RINm5F cell line and in mouse and human pancreatic islets (HPI) exposed to serum deprivation condition. 24 proteins in RINm5F and 22 in HPI were identified to undergo changes in at least one experimental condition. These include stress response mitochondrial proteins (UQCRC2, VDAC1, ATP5C1, ATP5A1) in RINm5F cells and stress response endoplasmic reticulum proteins (HSPA5, PDIA6, VCP, GANAB) in HPI. In addition, metabolic and structural proteins, oxidoreductases and chaperones related with protein metabolism are also regulated by NO treatment. Network analysis of differentially expressed proteins shows their interaction in glucocorticoid receptor and NRF2-mediated oxidative stress response pathways and eNOS signaling. The results indicate that exposure to exogenous NO counteracts the impact of serum deprivation on pancreatic β cell proteome. Species differences in the proteins involved are apparent.
Collapse
Affiliation(s)
- Rafael Tapia-Limonchi
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)- Pablo de Olavide University; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
| | - Irene Díaz
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)- Pablo de Olavide University; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
| | - Gladys M Cahuana
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)- Pablo de Olavide University; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
| | - Mario Bautista
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)- Pablo de Olavide University; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
| | - Franz Martín
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)- Pablo de Olavide University; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
| | - Bernat Soria
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)-Fundación Progreso y Salud; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
| | - Juan R Tejedo
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)- Pablo de Olavide University; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
| | - Francisco J Bedoya
- Andalusian Center for Molecular Biology and
Regenerative Medicine (CABIMER)- Pablo de Olavide University; Biomedical Research
Network (CIBER) of Diabetes and Related Metabolic Diseases; RED-TERCEL;
Seville, Spain
- Correspondence to: Francisco J. Bedoya;
| |
Collapse
|
35
|
Xiao X, Yang J, Li R, Liu S, Xu Y, Zheng W, Yi Y, Luo Y, Gong F, Peng H, Pei M, Deng M, Zhang G. Deregulation of mitochondrial ATPsyn-β in acute myeloid leukemia cells and with increased drug resistance. PLoS One 2013; 8:e83610. [PMID: 24391795 PMCID: PMC3877068 DOI: 10.1371/journal.pone.0083610] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 11/06/2013] [Indexed: 01/24/2023] Open
Abstract
The mechanisms underlying the development of multidrug resistance in acute myeloid leukemia are not fully understood. Here we analyzed the expressions of mitochondrial ATPsyn-β in adriamycin-resistant cell line HL-60/ADM and its parental cell line HL-60. Meanwhile we compared the differences of mitochondrial ATPsyn-β expression and ATP synthase activity in 110 acute myeloid leukemia (AML, non-M3) patients between relapsed/refractory and those in remission. Our results showed that down-regulation of ATPsyn-β expression by siRNA in HL-60 cells increased cell viability and apoptotic resistance to adriamycin, while up-regulation of mitochondrial ATPsyn-β in HL-60/ADM cells enhanced cell sensitivity to adriamycin and promoted apoptosis. Mitochondrial ATPsyn-β expression and ATP synthase activity in relapsed/refractory acute myeloid leukemia patients were downregulated. This downregulated ATPsyn-β expression exhibited a positive correlation with the response to adriamycin of primary cells. A lower expression of ATPsyn-β in newly diagnosed or relapsed/refractory patients was associated with a shorter first remission duration or overall survival. Our findings show mitochondrial ATPsyn-β plays an important role in the mechanism of multidrug resistance in AML thus may present both a new marker for prognosis assessment and a new target for reversing drug resistance.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/genetics
- Apoptosis/physiology
- Cell Survival/drug effects
- Cell Survival/genetics
- Cell Survival/physiology
- Down-Regulation
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/genetics
- Drug Resistance, Multiple/physiology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/physiology
- Female
- HL-60 Cells
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Male
- Middle Aged
- Mitochondria/enzymology
- Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors
- Mitochondrial Proton-Translocating ATPases/genetics
- Mitochondrial Proton-Translocating ATPases/metabolism
- Prognosis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA, Small Interfering/genetics
- Young Adult
Collapse
Affiliation(s)
- Xiang Xiao
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jingke Yang
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Ruijuan Li
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Sufang Liu
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yunxiao Xu
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Wenli Zheng
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yan Yi
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yunya Luo
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Fanjie Gong
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Honglin Peng
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Minfei Pei
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Mingyang Deng
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Guangsen Zhang
- Division of Hematology, Institution of Molecular Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- * E-mail:
| |
Collapse
|
36
|
Guaragnella N, Palermo V, Galli A, Moro L, Mazzoni C, Giannattasio S. The expanding role of yeast in cancer research and diagnosis: insights into the function of the oncosuppressors p53 and BRCA1/2. FEMS Yeast Res 2013; 14:2-16. [PMID: 24103154 DOI: 10.1111/1567-1364.12094] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/26/2013] [Accepted: 09/12/2013] [Indexed: 12/16/2022] Open
Abstract
When the glucose supply is high, despite the presence of oxygen, Saccharomyces cerevisiae uses fermentation as its main metabolic pathway and switches to oxidative metabolism only when this carbon source is limited. There are similarities between glucose-induced repression of oxidative metabolism of yeast and metabolic reprogramming of tumor cells. The glucose-induced repression of oxidative metabolism is regulated by oncogene homologues in yeast, such as RAS and Sch9p, the yeast homologue of Akt. Yeast also undergoes an apoptosis-like programmed cell death process sharing several features with mammalian apoptosis, including oxidative stress and a major role played by mitochondria. Evasion of apoptosis and sustained proliferative signaling are hallmarks of cancer. This, together with the possibility of heterologous expression of human genes in yeast, has allowed new insights to be obtained into the function of mammalian oncogenes/oncosuppressors. Here, we elaborate on the similarities between tumor and yeast cells underpinning the use of this model organism in cancer research. We also review the achievements obtained through heterologous expression in yeast of p53, BRCA1, and BRCA2, which are among the best-known cancer-susceptibility genes, with the aim of understanding their role in tumorigenesis. Yeast-cell-based functional assays for cancer genetic testing will also be dealt with.
Collapse
|
37
|
Liu XF, Zhang LM, Guan HN, Zhang ZW, Xu SW. Effects of oxidative stress on apoptosis in manganese-induced testicular toxicity in cocks. Food Chem Toxicol 2013; 60:168-76. [DOI: 10.1016/j.fct.2013.07.058] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 04/19/2013] [Accepted: 07/22/2013] [Indexed: 01/21/2023]
|
38
|
Sánchez-Aragó M, Formentini L, Cuezva JM. Mitochondria-mediated energy adaption in cancer: the H(+)-ATP synthase-geared switch of metabolism in human tumors. Antioxid Redox Signal 2013; 19:285-98. [PMID: 22901241 PMCID: PMC3691914 DOI: 10.1089/ars.2012.4883] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Since the signing of the National Cancer Act in 1971, cancer still remains a major cause of death despite significant progresses made in understanding the biology and treatment of the disease. After many years of ostracism, the peculiar energy metabolism of tumors has been recognized as an additional phenotypic trait of the cancer cell. RECENT ADVANCES While the enhanced aerobic glycolysis of carcinomas has already been translated to bedside for precise tumor imaging and staging of cancer patients, accepting that an impaired bioenergetic function of mitochondria is pivotal to understand energy metabolism of tumors and in its progression is debated. However, mitochondrial bioenergetics and cell death are tightly connected. CRITICAL ISSUES Recent clinical findings indicate that H(+)-ATP synthase, a core component of mitochondrial oxidative phosphorylation, is repressed at both the protein and activity levels in human carcinomas. This review summarizes the relevance that mitochondrial function has to understand energy metabolism of tumors and explores the connection between the bioenergetic function of the organelle and the activity of mitochondria as tumor suppressors. FUTURE DIRECTIONS The reversible nature of energy metabolism in tumors highlights the relevance that the microenvironment has for tumor progression. Moreover, the stimulation of mitochondrial activity or the inhibition of glycolysis suppresses tumor growth. Future research should elucidate the mechanisms promoting the silencing of oxidative phosphorylation in carcinomas. The aim is the development of new therapeutic strategies tackling energy metabolism to eradicate tumors or at least, to maintain tumor dormancy and transform cancer into a chronic disease.
Collapse
Affiliation(s)
- María Sánchez-Aragó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Centro de Investigación Biomédica en Red de Enfermedades Raras, Centro de Investigación Hospital 12 de Octubre, Madrid, Spain
| | | | | |
Collapse
|
39
|
Eißmann M, Schwamb B, Melzer IM, Moser J, Siele D, Köhl U, Rieker RJ, Wachter DL, Agaimy A, Herpel E, Baumgarten P, Mittelbronn M, Rakel S, Kögel D, Böhm S, Gutschner T, Diederichs S, Zörnig M. A functional yeast survival screen of tumor-derived cDNA libraries designed to identify anti-apoptotic mammalian oncogenes. PLoS One 2013; 8:e64873. [PMID: 23717670 PMCID: PMC3661464 DOI: 10.1371/journal.pone.0064873] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 04/19/2013] [Indexed: 11/29/2022] Open
Abstract
Yeast cells can be killed upon expression of pro-apoptotic mammalian proteins. We have established a functional yeast survival screen that was used to isolate novel human anti-apoptotic genes overexpressed in treatment-resistant tumors. The screening of three different cDNA libraries prepared from metastatic melanoma, glioblastomas and leukemic blasts allowed for the identification of many yeast cell death-repressing cDNAs, including 28% of genes that are already known to inhibit apoptosis, 35% of genes upregulated in at least one tumor entity and 16% of genes described as both anti-apoptotic in function and upregulated in tumors. These results confirm the great potential of this screening tool to identify novel anti-apoptotic and tumor-relevant molecules. Three of the isolated candidate genes were further analyzed regarding their anti-apoptotic function in cell culture and their potential as a therapeutic target for molecular therapy. PAICS, an enzyme required for de novo purine biosynthesis, the long non-coding RNA MALAT1 and the MAST2 kinase are overexpressed in certain tumor entities and capable of suppressing apoptosis in human cells. Using a subcutaneous xenograft mouse model, we also demonstrated that glioblastoma tumor growth requires MAST2 expression. An additional advantage of the yeast survival screen is its universal applicability. By using various inducible pro-apoptotic killer proteins and screening the appropriate cDNA library prepared from normal or pathologic tissue of interest, the survival screen can be used to identify apoptosis inhibitors in many different systems.
Collapse
Affiliation(s)
- Moritz Eißmann
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Bettina Schwamb
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Inga Maria Melzer
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Julia Moser
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Dagmar Siele
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Ulrike Köhl
- Institute of Cellular Therapeutics, IFB-Tx, Hannover Medical School, Hannover, Germany
| | | | | | - Abbas Agaimy
- Institute for Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Esther Herpel
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Peter Baumgarten
- Institute of Neurology (Edinger Institute), Frankfurt/Main, Germany
| | | | - Stefanie Rakel
- Experimental Neurosurgery, Center for Neurology and Neurosurgery, Goethe University Hospital Frankfurt, Frankfurt/Main, Germany
| | - Donat Kögel
- Experimental Neurosurgery, Center for Neurology and Neurosurgery, Goethe University Hospital Frankfurt, Frankfurt/Main, Germany
| | - Stefanie Böhm
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt/Main, Germany
| | - Tony Gutschner
- Helmholtz-University-Group Molecular RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sven Diederichs
- Helmholtz-University-Group Molecular RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Zörnig
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt/Main, Germany
- * E-mail:
| |
Collapse
|
40
|
Abstract
Recent findings in colon cancer cells indicate that inhibition of the mitochondrial H+-adenosine triphosphate (ATP) synthase by the ATPase inhibitory factor 1 (IF1) promotes aerobic glycolysis and a reactive oxygen species (ROS)-mediated signal that enhances proliferation and cell survival. Herein, we have studied the expression, biological relevance, mechanism of regulation and potential clinical impact of IF1 in some prevalent human carcinomas. We show that IF1 is highly overexpressed in most (>90%) of the colon (n=64), lung (n=30), breast (n=129) and ovarian (n=10) carcinomas studied as assessed by different approaches in independent cohorts of cancer patients. The expression of IF1 in the corresponding normal tissues is negligible. By contrast, the endometrium, stomach and kidney show high expression of IF1 in the normal tissue revealing subtle differences by carcinogenesis. The overexpression of IF1 also promotes the activation of aerobic glycolysis and a concurrent ROS signal in mitochondria of the lung, breast and ovarian cancer cells mimicking the activity of oligomycin. IF1-mediated ROS signaling activates cell-type specific adaptive responses aimed at preventing death in these cell lines. Remarkably, regulation of IF1 expression in the colon, lung, breast and ovarian carcinomas is exerted at post-transcriptional levels. We demonstrate that IF1 is a short-lived protein (t1/2 ∼100 min) strongly implicating translation and/or protein stabilization as main drivers of metabolic reprogramming and cell survival in these human cancers. Analysis of tumor expression of IF1 in cohorts of breast and colon cancer patients revealed its relevance as a predictive marker for clinical outcome, emphasizing the high potential of IF1 as therapeutic target.
Collapse
|
41
|
Bonora M, Bononi A, De Marchi E, Giorgi C, Lebiedzinska M, Marchi S, Patergnani S, Rimessi A, Suski JM, Wojtala A, Wieckowski MR, Kroemer G, Galluzzi L, Pinton P. Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle 2013; 12:674-83. [PMID: 23343770 PMCID: PMC3594268 DOI: 10.4161/cc.23599] [Citation(s) in RCA: 361] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The term "mitochondrial permeability transition" (MPT) refers to an abrupt increase in the permeability of the inner mitochondrial membrane to low molecular weight solutes. Due to osmotic forces, MPT is paralleled by a massive influx of water into the mitochondrial matrix, eventually leading to the structural collapse of the organelle. Thus, MPT can initiate mitochondrial outer membrane permeabilization (MOMP), promoting the activation of the apoptotic caspase cascade as well as of caspase-independent cell death mechanisms. MPT appears to be mediated by the opening of the so-called "permeability transition pore complex" (PTPC), a poorly characterized and versatile supramolecular entity assembled at the junctions between the inner and outer mitochondrial membranes. In spite of considerable experimental efforts, the precise molecular composition of the PTPC remains obscure and only one of its constituents, cyclophilin D (CYPD), has been ascribed with a crucial role in the regulation of cell death. Conversely, the results of genetic experiments indicate that other major components of the PTPC, such as voltage-dependent anion channel (VDAC) and adenine nucleotide translocase (ANT), are dispensable for MPT-driven MOMP. Here, we demonstrate that the c subunit of the FO ATP synthase is required for MPT, mitochondrial fragmentation and cell death as induced by cytosolic calcium overload and oxidative stress in both glycolytic and respiratory cell models. Our results strongly suggest that, similar to CYPD, the c subunit of the FO ATP synthase constitutes a critical component of the PTPC.
Collapse
Affiliation(s)
- Massimo Bonora
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| | - Angela Bononi
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| | - Elena De Marchi
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| | - Magdalena Lebiedzinska
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
- Department of Biochemistry; Nencki Institute of Experimental Biology; Warsaw, Poland
| | - Saverio Marchi
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| | - Simone Patergnani
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| | - Alessandro Rimessi
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| | - Jan M. Suski
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
- Department of Biochemistry; Nencki Institute of Experimental Biology; Warsaw, Poland
| | - Aleksandra Wojtala
- Department of Biochemistry; Nencki Institute of Experimental Biology; Warsaw, Poland
| | - Mariusz R. Wieckowski
- Department of Biochemistry; Nencki Institute of Experimental Biology; Warsaw, Poland
| | - Guido Kroemer
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- U848; INSERM; Villejuif, France
- Metabolomics Platform; Institut Gustave Roussy; Villejuif, France
- Equipe 11 Labelisée par la Ligue Contre le cancer; Centre de Recherche des Cordeliers; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Institut Gustave Roussy; Villejuif, France
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine; Section of General Pathology; Interdisciplinary Center for the Study of Inflammation (ICSI); Laboratory for Technologies of Advanced Therapies (LTTA); University of Ferrara; Ferrara, Italy
| |
Collapse
|
42
|
Andersen JL, Kornbluth S. The tangled circuitry of metabolism and apoptosis. Mol Cell 2013; 49:399-410. [PMID: 23395270 PMCID: PMC3801185 DOI: 10.1016/j.molcel.2012.12.026] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 12/11/2012] [Accepted: 12/26/2012] [Indexed: 12/15/2022]
Abstract
For single-cell organisms, nutrient uptake and metabolism are central to the fundamental decision of whether to grow or divide. In metazoans, cell fate decisions are more complex: organismal homeostasis must be strictly maintained by balancing cell proliferation and death. Despite this increased complexity, cell fate within multicellular organisms is also influenced by metabolism; recent studies, triggered in part by an interest in tumor metabolism, are beginning to illuminate the mechanisms through which proliferation, death, and metabolism are intertwined. In particular, work on Bcl-2 family proteins suggests that the signaling pathways governing metabolism and apoptosis are inextricably linked. Here we review the crosstalk between these pathways, emphasizing recent work that illustrates the emerging dual nature of several core apoptotic proteins in regulating both metabolism and cell death.
Collapse
Affiliation(s)
- Joshua L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | | |
Collapse
|
43
|
Zhou K, Ren Y, Lv J, Wang Y, Liu F, Zhou F, Zhao S, Chen S, Peng C, Zhang X, Guo X, Cheng Z, Wang J, Wu F, Jiang L, Wan J. Young Leaf Chlorosis 1, a chloroplast-localized gene required for chlorophyll and lutein accumulation during early leaf development in rice. PLANTA 2013; 237:279-92. [PMID: 23053539 DOI: 10.1007/s00425-012-1756-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 08/28/2012] [Indexed: 05/08/2023]
Abstract
Chlorophyll (Chl) and lutein are the two most abundant and essential components in photosynthetic apparatus, and play critical roles in plant development. In this study, we characterized a rice mutant named young leaf chlorosis 1 (ylc1) from a ⁶⁰Co-irradiated population. Young leaves of the ylc1 mutant showed decreased levels of Chl and lutein compared to those of wild type, and transmission electron microscopy analysis revealed that the thylakoid lamellar structures were obviously loosely arranged. Whereas, the mutant turns green gradually and approaches normal green at the maximum tillering stage. The Young Leaf Chlorosis 1 (YLC1) gene was isolated via map-based cloning and identified to encode a protein of unknown function belonging to the DUF3353 superfamily. Complementation and RNA-interference tests confirmed the role of the YLC1 gene, which expressed in all tested rice tissues, especially in the leaves. Real-time PCR analyses showed that the expression levels of the genes associated with Chl biosynthesis and photosynthesis were affected in ylc1 mutant at different temperatures. In rice protoplasts, the YLC1 protein displayed a typical chloroplast location pattern. The N-terminal 50 amino acid residues were confirmed to be necessary and sufficient for chloroplast targeting. These data suggested that the YLC1 protein may be involved in Chl and lutein accumulation and chloroplast development at early leaf development in rice.
Collapse
Affiliation(s)
- Kunneng Zhou
- National Key Laboratory for Crop Genetics and Germplasm Enhancement, Jiangsu Plant Gene Engineering Research Center, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Guaragnella N, Zdralević M, Antonacci L, Passarella S, Marra E, Giannattasio S. The role of mitochondria in yeast programmed cell death. Front Oncol 2012; 2:70. [PMID: 22783546 PMCID: PMC3388595 DOI: 10.3389/fonc.2012.00070] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 06/14/2012] [Indexed: 01/02/2023] Open
Abstract
Mammalian apoptosis and yeast programmed cell death (PCD) share a variety of features including reactive oxygen species production, protease activity and a major role played by mitochondria. In view of this, and of the distinctive characteristics differentiating yeast and multicellular organism PCD, the mitochondrial contribution to cell death in the genetically tractable yeast Saccharomyces cerevisiae has been intensively investigated. In this mini-review we report whether and how yeast mitochondrial function and proteins belonging to oxidative phosphorylation, protein trafficking into and out of mitochondria, and mitochondrial dynamics, play a role in PCD. Since in PCD many processes take place over time, emphasis will be placed on an experimental model based on acetic acid-induced PCD (AA-PCD) which has the unique feature of having been investigated as a function of time. As will be described there are at least two AA-PCD pathways each with a multifaceted role played by mitochondrial components, in particular by cytochrome c.
Collapse
Affiliation(s)
- Nicoletta Guaragnella
- Institute of Biomembranes and Bioenergetics, National Research Council of Italy, Bari, Italy
| | | | | | | | | | | |
Collapse
|
45
|
Formentini L, Sánchez-Aragó M, Sánchez-Cenizo L, Cuezva JM. The mitochondrial ATPase inhibitory factor 1 triggers a ROS-mediated retrograde prosurvival and proliferative response. Mol Cell 2012; 45:731-42. [PMID: 22342343 DOI: 10.1016/j.molcel.2012.01.008] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 11/02/2011] [Accepted: 01/06/2012] [Indexed: 01/06/2023]
Abstract
Recent findings indicate that prevalent human carcinomas overexpress the mitochondrial ATPase Inhibitory Factor 1 (IF1). Overexpression of IF1 inhibits the synthase activity of the mitochondrial H(+)-ATP synthase and plays a crucial role in metabolic adaptation of cancer cells to enhanced aerobic glycolysis. Herein, we demonstrate that IF1 overexpression in colon cancer cells triggers mitochondrial hyperpolarization and the subsequent production of superoxide radical, a reactive oxygen species (ROS). ROS are required to promote the transcriptional activation of the NFκB pathway via phosphorylation-dependent IκBα degradation. Activation of NFκB results in a cellular adaptive response that includes proliferation and Bcl-xL mediated resistance to drug-induced cell death. Quenching the mitochondrial production of ROS prevents the activation of NFκB and abolishes the IF1-mediated cellular adaptive response. Overall, our findings provide evidence linking the activity of a mitochondrial protein with retrograde signaling to the nucleus to promote cellular proliferation and survival.
Collapse
Affiliation(s)
- Laura Formentini
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | | | | | | |
Collapse
|
46
|
Yang CA, Cheng CH, Lee JW, Lo CT, Liu SY, Peng KC. Monomeric L-amino acid oxidase-induced mitochondrial dysfunction in Rhizoctonia solani Reveals a novel antagonistic mechanism of Trichoderma harzianum ETS 323. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:2464-2471. [PMID: 22352318 DOI: 10.1021/jf203883u] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The monomeric L-amino acid oxidase (mTh-LAAO) of Trichoderma harzianum ETS 323 has been suggested to antagonize Rhizoctonia solani by an unknown mechanism. Here, the mTh-LAAO-treated R. solani exhibited hyphal lysis and apoptotic characteristics such as DNA fragmentation, reactive oxygen species (ROS) accumulation, lipid peroxidation, and mitochondrial membrane potential depolarization. This hyphal lysis was suppressed by the mitochondria-dependent apoptosis inhibitor oligomycin while accompanied by reduction of ROS accumulation. This result suggested that mitochondria-mediated apoptosis in R. solani was involved in mTh-LAAO-induced growth inhibition, which was supported by the evidence of cytocheome c release and activation of caspases 9 and 3. Furthermore, the data indicated that the mTh-LAAO-induced fungal cell death was also closely interrelated with the interaction of mTh-LAAO with R. solani hyphal cell wall proteins. These results illuminate the biological function and mechanism underlying the antagonistic action of T. harzianum mTh-LAAO against fungal pathogens.
Collapse
Affiliation(s)
- Chia-Ann Yang
- Institute of Medical Science, Tzu Chi University, Hualien, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
47
|
Pereira C, Coutinho I, Soares J, Bessa C, Leão M, Saraiva L. New insights into cancer-related proteins provided by the yeast model. FEBS J 2012; 279:697-712. [PMID: 22239976 DOI: 10.1111/j.1742-4658.2012.08477.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cancer is a devastating disease with a profound impact on society. In recent years, yeast has provided a valuable contribution with respect to uncovering the molecular mechanisms underlying this disease, allowing the identification of new targets and novel therapeutic opportunities. Indeed, several attributes make yeast an ideal model system for the study of human diseases. It combines a high level of conservation between its cellular processes and those of mammalian cells, with advantages such as a short generation time, ease of genetic manipulation and a wealth of experimental tools for genome- and proteome-wide analyses. Additionally, the heterologous expression of disease-causing proteins in yeast has been successfully used to gain an understanding of the functions of these proteins and also to provide clues about the mechanisms of disease progression. Yeast research performed in recent years has demonstrated the tremendous potential of this model system, especially with the validation of findings obtained with yeast in more physiologically relevant models. The present review covers the major aspects of the most recent developments in the yeast research area with respect to cancer. It summarizes our current knowledge on yeast as a cellular model for investigating the molecular mechanisms of action of the major cancer-related proteins that, even without yeast orthologues, still recapitulate in yeast some of the key aspects of this cellular pathology. Moreover, the most recent contributions of yeast genetics and high-throughput screening technologies that aim to identify some of the potential causes underpinning this disorder, as well as discover new therapeutic agents, are discussed.
Collapse
Affiliation(s)
- Clara Pereira
- REQUIMTE, Department of Biological Sciences, Laboratory of Microbiology, University of Porto, Portugal
| | | | | | | | | | | |
Collapse
|
48
|
Preuss B, Berg C, Dengjel J, Stevanovic S, Klein R. Relevance of the inner mitochondrial membrane enzyme F1F0-ATPase as an autoantigen in autoimmune liver disorders. Liver Int 2012; 32:249-57. [PMID: 22098431 DOI: 10.1111/j.1478-3231.2011.02630.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 07/29/2011] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Recently, a non-M2-related mitochondrial 60 kDa protein found to be recognized by antimitochondrial antibody (AMA) negative sera from patients with primary biliary cirrhosis (PBC) has been shown to contain parts of the five F(1)-ATPase subunits α, β, γ, δ and ε. In this study, we examined whether this enzyme is, indeed, a target antigen in PBC. METHODS Analysed were 60 AMA-positive/anti-M2-negative and 103 anti-M2-positive PBC patients, 46 patients with autoimmune hepatitis (AIH), 35 patients with primary sclerosing cholangitis (PSC), 110 patients with viral hepatitis, 40 patients with inflammatory bowel diseases (IBD), 33 patients with connective tissue diseases (systemic lupus erythematosus, mixed connective tissue disease, Sjögren disease, systemic sclerosis) and 25 blood donors. The F(1)-ATPase-subunits α-δ were recombinantly expressed in Escherichia coli, purified and applied to ELISA and Western blotting. RESULTS In all, 40 of the 60 AMA-positive/anti-M2-negative (67%) and 44 (43%) of the 103 anti-M2-positive PBC-sera reacted with at least one of the F(1)-subunits α-δ. The β- and γ-subunits were preferentially recognized. However, also up to 57% of patients with AIH and 34% of patients with PSC had anti-β- or γ-antibodies, while patients with viral hepatitis had these antibodies in up to 13%. Patients with IBD had anti-β and anti-γ-antibodies in up to 20 and 5% respectively. None of the patients with connective tissue diseases had antibodies to the β- and only 6% to the γ-subunit. Sera from healthy blood donors were negative. CONCLUSIONS Antibodies to the β- and γ-subunits of F(1)-ATPase are further AMAs in PBC but occur also in other autoimmune liver disorders; they may be, therefore, indicators for a general autoimmune process of the liver.
Collapse
Affiliation(s)
- Beate Preuss
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | | | | | | | | |
Collapse
|
49
|
He L, Jang JH, Choi HG, Lee SM, Nan MH, Jeong SJ, Dong Z, Kwon YT, Lee KS, Lee KW, Chung JK, Ahn JS, Kim BY. Oligomycin A enhances apoptotic effect of TRAIL through CHOP-mediated death receptor 5 expression. Mol Carcinog 2011; 52:85-93. [DOI: 10.1002/mc.21831] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 10/06/2011] [Accepted: 10/12/2011] [Indexed: 11/06/2022]
|
50
|
Ponnala S, Chetty C, Veeravalli KK, Dinh DH, Klopfenstein JD, Rao JS. Metabolic remodeling precedes mitochondrial outer membrane permeabilization in human glioma xenograft cells. Int J Oncol 2011; 40:509-18. [PMID: 22076676 DOI: 10.3892/ijo.2011.1255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Accepted: 09/09/2011] [Indexed: 11/06/2022] Open
Abstract
Glioma cancer cells adapt to changing microenvironment and shift from mitochondrial oxidative phosphorylation to aerobic glycolysis for their metabolic needs irrespective of oxygen availability. In the present study, we show that silencing MMP-9 in combination with uPAR/cathepsin B switch the glycolytic metabolism of glioma cells to oxidative phosphorylation (OXPHOS) and generate reactive oxygen species (ROS) to predispose glioma cells to mitochondrial outer membrane permeabilization. shRNA for MMP-9 and uPAR (pMU) as well as shRNA for MMP-9 and cathepsin B (pMC) activated complexes of mitochondria involved in OXPHOS and inhibited glycolytic hexokinase expression. The decreased interaction of hexokinase 2 with mitochondria in the treated cells indicated the inhibition of glycolysis activation. Overexpression of Akt reversed the pMU- and pMC-mediated OXPHOS to glycolysis switch. The OXPHOS un-coupler oligomycin A altered the expression levels of the Bcl-2 family of proteins; treatment with pMU or pMC reversed this effect and induced mitochondrial outer membrane permeabilization. In addition, our results show changes in mitochondrial pore transition to release cytochrome c due to changes in the VDAC-Bcl-XL and BAX-BAK interaction with pMU and pMC treatments. Taken together, our results suggest that pMU and pMC treatments switch glioma cells from the glycolytic to the OXPHOS pathway through an inhibitory effect on Akt, ROS induction and an increase of cytosolic cytochrome c accumulation. These results demonstrate the potential of pMU and pMC as therapeutic candidates for the treatment of glioma.
Collapse
Affiliation(s)
- Shivani Ponnala
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | | | | | | | | | | |
Collapse
|