1
|
Mussulini BHM, Maruszczak KK, Draczkowski P, Borrero-Landazabal MA, Ayyamperumal S, Wnorowski A, Wasilewski M, Chacinska A. MIA40 suppresses cell death induced by apoptosis-inducing factor 1. EMBO Rep 2025; 26:1835-1862. [PMID: 40055465 PMCID: PMC11976965 DOI: 10.1038/s44319-025-00406-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/08/2025] [Accepted: 01/24/2025] [Indexed: 04/09/2025] Open
Abstract
Mitochondria harbor respiratory complexes that perform oxidative phosphorylation. Complex I is the first enzyme of the respiratory chain that oxidizes NADH. A dysfunction in complex I can result in higher cellular levels of NADH, which in turn strengthens the interaction between apoptosis-inducing factor 1 (AIFM1) and Mitochondrial intermembrane space import and assembly protein 40 (MIA40) in the mitochondrial intermembrane space. We investigated whether MIA40 modulates the activity of AIFM1 upon increased NADH/NAD+ balance. We found that in model cells characterized by an increase in NADH the AIFM1-MIA40 interaction is strengthened and these cells demonstrate resistance to AIFM1-induced cell death. Either silencing of MIA40, rescue of complex I, or depletion of NADH through the expression of yeast NADH-ubiquinone oxidoreductase-2 sensitized NDUFA13-KO cells to AIFM1-induced cell death. These findings indicate that the complex of MIA40 and AIFM1 suppresses AIFM1-induced cell death in a NADH-dependent manner. This study identifies an effector complex involved in regulating the programmed cell death that accommodates the metabolic changes in the cell and provides a molecular explanation for AIFM1-mediated chemoresistance of cancer cells.
Collapse
Affiliation(s)
- Ben Hur Marins Mussulini
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland
- IMol Polish Academy of Sciences, Warsaw, Poland
| | | | - Piotr Draczkowski
- National Bioinformatics Infrastructure Sweden, SciLifeLab, Solna, Sweden
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Lublin, Poland
| | | | | | - Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, Lublin, Poland
| | - Michal Wasilewski
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland
- IMol Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Chacinska
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland.
- IMol Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
2
|
Fang J, Zhou G, Zhao H, Xie D, Zhang J, Kües U, Xiao Y, Fang Z, Liu J. An apoptosis-inducing factor controls programmed cell death and laccase expression during fungal interactions. Appl Microbiol Biotechnol 2024; 108:135. [PMID: 38229306 PMCID: PMC10787690 DOI: 10.1007/s00253-023-12988-1] [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: 10/24/2023] [Revised: 12/15/2023] [Accepted: 12/24/2023] [Indexed: 01/18/2024]
Abstract
Apoptotic-like programmed cell death (PCD) is one of the main strategies for fungi to resist environmental stresses and maintain homeostasis. The apoptosis-inducing factor (AIF) has been shown in different fungi to trigger PCD through upregulating reactive oxygen species (ROS). This study identified a mitochondrial localized AIF homolog, CcAIF1, from Coprinopsis cinerea monokaryon Okayama 7. Heterologous overexpression of CcAIF1 in Saccharomyces cerevisiae caused apoptotic-like PCD of the yeast cells. Ccaif1 was increased in transcription when C. cinerea interacted with Gongronella sp. w5, accompanied by typical apoptotic-like PCD in C. cinerea, including phosphatidylserine externalization and DNA fragmentation. Decreased mycelial ROS levels were observed in Ccaif1 silenced C. cinerea transformants during cocultivation, as well as reduction of the apoptotic levels, mycelial growth, and asexual sporulation. By comparison, Ccaif1 overexpression led to the opposite phenotypes. Moreover, the transcription and expression levels of laccase Lcc9 decreased by Ccaif1 silencing but increased firmly in Ccaif1 overexpression C. cinerea transformants in coculture. Thus, in conjunction with our previous report that intracellular ROS act as signal molecules to stimulate defense responses, we conclude that CcAIF1 is a regulator of ROS to promote apoptotic-like PCD and laccase expression in fungal-fungal interactions. In an axenic culture of C. cinerea, CcAIF1 overexpression and H2O2 stimulation together increased laccase secretion with multiplied production yield. The expression of two other normally silent isozymes, Lcc8 and Lcc13, was unexpectedly triggered along with Lcc9. KEY POINTS: • Mitochondrial CcAIF1 induces PCD during fungal-fungal interactions • CcAIF1 is a regulator of ROS to trigger the expression of Lcc9 for defense • CcAIF1 overexpression and H2O2 stimulation dramatically increase laccase production.
Collapse
Affiliation(s)
- Junnan Fang
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China
| | - Gang Zhou
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China
| | - Huifang Zhao
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China
| | - Dengdeng Xie
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China
| | - Jingna Zhang
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China
| | - Ursula Kües
- Molecular Wood Biotechnology and Technical Mycology, Büsgen‑Institute, University of Goettingen, Büsgenweg 2, 37077, Goettingen, Germany
| | - Yazhong Xiao
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China
| | - Zemin Fang
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China.
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China.
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China.
| | - Juanjuan Liu
- School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China.
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, Anhui, China.
- Anhui Provincial Engineering Technology Research Center of Microorganisms and Biocatalysis, Hefei, 230601, Anhui, China.
| |
Collapse
|
3
|
Li L, Du C. Fungal Apoptosis-Related Proteins. Microorganisms 2024; 12:2289. [PMID: 39597678 PMCID: PMC11596484 DOI: 10.3390/microorganisms12112289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Programmed cell death (PCD) plays a crucial role in the development and homeostasis maintenance of multicellular organisms. Apoptosis is a form of PCD that prevents pathological development by eliminating damaged or useless cells. Despite the complexity of fungal apoptosis mechanisms being similar to those of plants and metazoans, fungal apoptosis lacks the core regulatory elements of animal apoptosis. Apoptosis-like PCD in fungi can be triggered by a variety of internal and external factors, participating in biological processes such as growth, development, and stress response. Although the core regulatory elements are not fully understood, apoptosis-inducing factor and metacaspase have been found to be involved. This article summarizes various proteins closely related to fungal apoptosis, such as apoptosis-inducing factor, metacaspase, and inhibitors of apoptosis proteins, as well as their structures and functions. This research provides new strategies and ideas for the development of natural drugs targeting fungal apoptosis and the control of fungal diseases.
Collapse
Affiliation(s)
| | - Chunmei Du
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang Provincial Key Laboratory of Plant Genetic Engineering and Biological Fermentation Engineering for Cold Region, Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Sciences, Heilongjiang University, Harbin 150080, China;
| |
Collapse
|
4
|
Zehra K, Banu A, Can E, Hülya C. Fisetin and/or capecitabine causes changes in apoptosis pathways in capecitabine-resistant colorectal cancer cell lines. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7913-7926. [PMID: 38748229 PMCID: PMC11449987 DOI: 10.1007/s00210-024-03145-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 10/04/2024]
Abstract
Capecitabine is recommended as one of the first-line chemotherapy treatments for advanced or metastatic colorectal cancer. Researches have been conducted on capecitabine's impact on the viability of human colon cancer cells and its potential to induce apoptosis. However, even in cases initially responsive to treatment, the development of acquired resistance significantly limits its efficacy. Challenges still exist in effectively treating patients with chemotherapy, and developing new cytotoxic drugs is hindered by drug resistance. Fisetin alters the cell cycle, inducing apoptosis, inhibiting cancer cell proliferation, and enhancing the therapeutic effectiveness of chemotherapy drugs. This work aims to create a plan for reversing capecitabine resistance. For this purpose, the role of capecitabine and/or fisetin combinations in cell proliferation and apoptosis has been determined in both wild-type and capecitabine-resistant HT29 cells (CR/HT29). We developed capecitabine-resistant cell line from wild-type HT29 cells. This study demonstrated the effects of capecitabine, fisetin, and their combinations on both resistant and wild-type cells through experiments including cell survival skills, cell proliferation, wound healing, colony formation, hoechst staining, and western blot analysis. We established capecitabine-resistant cell lines. P-gp expression increased in CR/HT29 cells. Capecitabine effects on a CR/HT29 cells less than wild-type HT29 cells. The combination of fisetin and capecitabine in cell proliferation caused greater reductions in wild-type HT29 cells than in capecitabine-resistant cells. Fisetin has also additive effects on the apoptotic pathway in CR/HT29 cells. This study provides new perspectives on the combination of capecitabine and/or flavonoid treatment in resistant cells.
Collapse
Affiliation(s)
- Kanli Zehra
- Institute of Health Sciences, Marmara University, Basibuyuk-Maltepe, Istanbul, 34854, Turkey
| | - Aydin Banu
- School of Medicine, Department of Biophysics, Marmara University, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| | - Erzik Can
- School of Medicine, Department of Medical Biology, Marmara University, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| | - Cabadak Hülya
- School of Medicine, Department of Biophysics, Marmara University, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey.
| |
Collapse
|
5
|
Li M, Cong R, Wang H, Ma C, Lv Y, Zheng Y, Zhao Y, Fu Q, Li L. What happens to the osteoporotic bone mesenchymal stem cells? Evidence from RNA sequencing. Int J Med Sci 2024; 21:95-106. [PMID: 38164361 PMCID: PMC10750345 DOI: 10.7150/ijms.88146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/04/2023] [Indexed: 01/03/2024] Open
Abstract
Evidence presented that osteoporosis is closely related to the dysfunction of bone mesenchymal stem cells (BMSCs). But most studies are insufficient to reveal what actually happens to the osteoporotic BMSCs. In this study, BMSCs were harvested from ovariectomized and sham-operated rats. After checking the characteristics of rat models and stem cells, the BMSCs were carried out for RNA sequencing. Part of the findings were verified that seven mRNAs (Abi3bp, Aifm3, Ccl11, Cdkn1c, Chst10, Id2, Vcam1) were significantly up-regulated in osteoporotic BMSCs while seven mRNAs (Cep63, Fgfr3, Myc, Omd, Pou2f1, Smarcal1, Timm10b) were down-regulated. In addition, potential miRNA-mRNA and lncRNA-mRNA regulatory networks were illustrated. The changes in osteoporotic BMSCs covered a large set of biological processes, including cell viability, differentiation, immunoreaction, bone repairment and estrogen defect. This study enriched the pathophysiological mechanisms of BMSCs and osteporosis, as well as provided dozens of attractive RNA targets for further treatment.
Collapse
Affiliation(s)
- Mingyang Li
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Rong Cong
- Senior Department of Obstetrics & Gynecology, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Huadong Wang
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Chao Ma
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Yongwei Lv
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Yang Zheng
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Yantao Zhao
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Qin Fu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Li Li
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, China
| |
Collapse
|
6
|
Shoshan-Barmatz V, Arif T, Shteinfer-Kuzmine A. Apoptotic proteins with non-apoptotic activity: expression and function in cancer. Apoptosis 2023; 28:730-753. [PMID: 37014578 PMCID: PMC10071271 DOI: 10.1007/s10495-023-01835-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
Apoptosis is a process of programmed cell death in which a cell commits suicide while maintaining the integrity and architecture of the tissue as a whole. Apoptosis involves activation of one of two major pathways: the extrinsic pathway, where extracellular pro-apoptotic signals, transduced through plasma membrane death receptors, activate a caspase cascade leading to apoptosis. The second, the intrinsic apoptotic pathway, where damaged DNA, oxidative stress, or chemicals, induce the release of pro-apoptotic proteins from the mitochondria, leading to the activation of caspase-dependent and independent apoptosis. However, it has recently become apparent that proteins involved in apoptosis also exhibit non-cell death-related physiological functions that are related to the cell cycle, differentiation, metabolism, inflammation or immunity. Such non-conventional activities were predominantly reported in non-cancer cells although, recently, such a dual function for pro-apoptotic proteins has also been reported in cancers where they are overexpressed. Interestingly, some apoptotic proteins translocate to the nucleus in order to perform a non-apoptotic function. In this review, we summarize the unconventional roles of the apoptotic proteins from a functional perspective, while focusing on two mitochondrial proteins: VDAC1 and SMAC/Diablo. Despite having pro-apoptotic functions, these proteins are overexpressed in cancers and this apparent paradox and the associated pathophysiological implications will be discussed. We will also present possible mechanisms underlying the switch from apoptotic to non-apoptotic activities although a deeper investigation into the process awaits further study.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Beer Sheva, Israel.
| | - Tasleem Arif
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | |
Collapse
|
7
|
Alimba CG, Sivanesan S, Krishnamurthi K. Mitochondrial dysfunctions elicited by solid waste leachates provide insights into mechanisms of leachates induced cell death and pathophysiological disorders. CHEMOSPHERE 2022; 307:136085. [PMID: 36007733 DOI: 10.1016/j.chemosphere.2022.136085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 06/15/2023]
Abstract
Emissions (mainly leachates and landfill gases) from solid waste facilities are laden with mixtures of dangerous xenobiotics implicated with significant increase in various pathophysiological disorders including cancer, and eventual mortality of exposed wildlife and humans. However, the molecular mechanisms of solid waste leachates induce pathophysiological disorders and cell death are still largely unknown. Although, evolving evidence implicated generation of reactive oxygen species and oxidative stress as the possible mechanism. Recent scientific reports are linking reactive oxygen species and mitochondrial dysfunctions as the player mechanism in pathophysiological disorder and apoptosis induced by xenobiotics in solid waste leachates. This systematic review presents an explicit discussion of recent scientific findings on the structural and functional alterations in mitochondria induced by solid waste leachates as the molecular mechanisms plausibly responsible for the pathophysiological disorders, cancer and cell death reported in landfill toxicology and epidemiological studies. This review aims to increase scientific understanding on solid waste leachate induced mitochondria dysfunctions as the key player in molecular mechanisms of solid waste induced toxicity. The findings in this review were mainly from using primary cells, cell lines, Drosophila and fish. Whether the findings will similarly be observed in mammalian test systems in vivo and particularly in exposed humans, remained to be investigated. Improvement in technological advancements, enforcement of legislation and regulations, and creation of sophisticated health surveillance against exposure to solid waste leachates, will expectedly mitigate human exposure to solid waste emissions and contamination of the environment.
Collapse
Affiliation(s)
- Chibuisi Gideon Alimba
- Cell Biology and Genetics Unit, Department of Zoology, University of Ibadan, Ibadan, Nigeria; Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, 44139, Dortmund, Germany.
| | - Saravanadevi Sivanesan
- Health and Toxicity Cell (HTC), CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India; Academy of Scientific, Innovative Research (AcSIR), Ghaziabad, U.P, India
| | - Kannan Krishnamurthi
- Health and Toxicity Cell (HTC), CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India; Academy of Scientific, Innovative Research (AcSIR), Ghaziabad, U.P, India.
| |
Collapse
|
8
|
An HG, Shin S, Lee B, Kwon Y, Kwon TU, Kwon YJ, Chun YJ. Induction of synergistic apoptosis by tetramethoxystilbene and nutlin-3a in human cervical cancer cells. Toxicol Res 2022; 38:591-600. [PMID: 36277372 PMCID: PMC9532473 DOI: 10.1007/s43188-022-00150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022] Open
Abstract
2,4,3',5'-Tetramethoxystilbene (TMS) is a selective inhibitor of cytochrome P450 1B1 to block the conversion from estradiol to 4-OH-estradiol. Several studies suggested that TMS may act as a potent anti-cancer agent for hormone-related cancer including cervical cancer. Nutlin-3a is a cis-imidazoline analog that interferes with the interaction between mouse double minute 2 homolog (MDM2) and the tumor suppressor p53. The purpose of the study was to compare the cytotoxic effect of TMS and nutlin-3a treatment individually and in combination in HeLa cells. To assess the potential synergistic effects between TMS and nutlin-3a, low concentrations of TMS and nutlin-3a were simultaneously treated in HeLa cells. Based on cell viability, apoptosis assays, and the increase in cleaved caspase-3 and poly (ADP-ribose) polymerase cleavage, it was demonstrated that the combination with TMS and nutlin-3a exerts a synergistic effect on cancer cell death. Isobologram analysis of HeLa cells noted synergism between TMS and nutlin-3a. The combined treatment increased the expression of mitochondrial pro-apoptotic factors such as Bax and Bak, and decreased the expression of the XIAP. In addition, combination treatment significantly enhanced the translocation of AIF to the nucleus in HeLa cells. In conclusion, the results demonstrate that the combination of TMS and nutlin-3a induces synergistic apoptosis in HeLa cells, suggesting the possibility that this combination can be applied as a novel therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Hong-Gyu An
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Sangyun Shin
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Boyoung Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Yeonju Kwon
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Tae-Uk Kwon
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, 06974 Seoul, Republic of Korea
| |
Collapse
|
9
|
Alimba CG, Rudrashetti AP, Sivanesan S, Krishnamurthi K. Landfill soil leachates from Nigeria and India induced DNA damage and alterations in genes associated with apoptosis in Jurkat cell. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:5256-5268. [PMID: 34417692 DOI: 10.1007/s11356-021-15985-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/11/2021] [Indexed: 06/13/2023]
Abstract
Landfill soil leachates, containing myriad of xenobiotics, increase genotoxic and cytotoxic stress-induced cell death. However, the underlying mechanism involved in the elimination of the damaged cells is yet to be fully elucidated. This study investigated the apoptotic processes induced in lymphoma (Jurkat) cells by landfill soil leachates from Olusosun (OSL, Nigeria) and Nagpur (NPL, India). Jurkat was incubated with sub-lethal concentrations of OSL and NPL for 24 h and analyzed for DNA fragmentation and apoptosis using agarose gel electrophoresis and Hoechst 33258-PI staining, respectively. Complementary DNA expression profiling of some pro-apoptotic and anti-apoptotic genes regulating apoptosis was also analyzed using real-time PCR (RT-PCR) method. Agarose gel electrophoresis revealed DNA fragmentations in OSL and NPL-treated cells. Hoecsht-33258 - Propidium Iodide (PI) based apoptotic analysis confirmed apoptotic cell death in exposed Jurkat. RT-PCR analysis revealed different fold changes in the pro- and anti-apoptotic genes in OSL and NPL-treated Jurkat. There was significant increase in fold change of the up-regulated genes; apoptosis inducing factor mitochondrion-associated 2 (AIFM2), Fas-associated death domain (FADD), Caspase-2, Caspase-6, BH3 interacting domain death agonist (BID), tumor suppressor (p53), and BCL2 associated agonist of cell death (BAD) and down-regulation of apoptosis inhibitor 5 (API5). Results suggest that OSL and NPL elicited genotoxic stress-related apoptosis in Jurkat. The dysregulation in the expression of genes involved in apoptotic processes in wildlife and human exposed to landfill emissions may increase aetiology of various pathological diseases including cancer.
Collapse
Affiliation(s)
- Chibuisi G Alimba
- Cell Biology and Genetics Unit, Department of Zoology, University of Ibadan, Ibadan, Nigeria.
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, 44139, Dortmund, Germany.
| | - Ashwinkumar P Rudrashetti
- Environmental Biotechnology and Genomic Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India
| | - Saravanadevi Sivanesan
- Health and Toxicity Cell (HTC), CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India
- Academy of Scientific, Innovative Research (AcSIR), Ghaziabad, U.P., India
| | - Kannan Krishnamurthi
- Health and Toxicity Cell (HTC), CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, 440020, India.
- Academy of Scientific, Innovative Research (AcSIR), Ghaziabad, U.P., India.
| |
Collapse
|
10
|
Ullah R, Rauf N, Nabi G, Yi S, Yu-Dong Z, Fu J. Mechanistic insight into high-fat diet-induced metabolic inflammation in the arcuate nucleus of the hypothalamus. Biomed Pharmacother 2021; 142:112012. [PMID: 34388531 DOI: 10.1016/j.biopha.2021.112012] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
A high-fat diet (HFD) is linked with cytokines production by non-neuronal cells within the hypothalamus, which mediates metabolic inflammation. These cytokines then activate different inflammatory mediators in the arcuate nucleus of the hypothalamus (ARC), a primary hypothalamic area accommodating proopiomelanocortin (POMC) and agouti-related peptide (AGRP) neurons, first-order neurons that sense and integrate peripheral metabolic signals and then respond accordingly. These mediators, such as inhibitor of κB kinase-β (IKKβ), suppression of cytokine signaling 3 (SOCS3), c-Jun N-terminal kinases (JNKs), protein kinase C (PKC), etc., cause insulin and leptin resistance in POMC and AGRP neurons and support obesity and related metabolic complications. On the other hand, inhibition of these mediators has been shown to counteract the impaired metabolism. Therefore, it is important to discuss the contribution of neuronal and non-neuronal cells in HFD-induced hypothalamic inflammation. Furthermore, understanding few other questions, such as the diets causing hypothalamic inflammation, the gender disparity in response to HFD feeding, and how hypothalamic inflammation affects ARC neurons to cause impaired metabolism, will be helpful for the development of therapeutic approaches to prevent or treat HFD-induced obesity.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China; Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Naveed Rauf
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Ghulam Nabi
- Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China; Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| | - Shen Yi
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Zhou Yu-Dong
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China; National Clinical Research Center for Child Health, Hangzhou 310052, China; National Children's Regional Medical Center, Hangzhou 310052, China.
| |
Collapse
|
11
|
Novo N, Ferreira P, Medina M. The apoptosis-inducing factor family: Moonlighting proteins in the crosstalk between mitochondria and nuclei. IUBMB Life 2021; 73:568-581. [PMID: 33035389 DOI: 10.1002/iub.2390] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022]
Abstract
In Homo sapiens, the apoptosis-inducing factor (AIF) family is represented by three different proteins, known as AIF, AMID and AIFL, that have in common the mitochondrial localisation in healthy cells, the presence of FAD- and NADH-dependent domains involved in an -albeit yet not well understood- oxidoreductase function and their capability to induce programmed cell death. AIF is the best characterised family member, while the information about AMID and AIFL is much scarcer. Nonetheless, available data support different roles as well as mechanisms of action of their particular apoptogenic and redox domains regarding both pro-apoptotic and anti-apoptotic activities. Moreover, diverse cellular functions, to date far from fully clarified, are envisaged for the transcripts corresponding to these three proteins. Here, we review the so far available knowledge on the moonlighting human AIF family from their molecular properties to their relevance in health and disease, through the evaluation of their potential cell death and redox functions in their different subcellular locations. This picture emerging from the current knowledge of the AIF family envisages its contribution to regulate signalling and transcription machineries in the crosstalk among mitochondria, the cytoplasm and the nucleus.
Collapse
Affiliation(s)
- Nerea Novo
- Department of Biochemistry and Molecular and Cellular Biology, Faculty of Sciences, University of Zaragoza, Zaragoza, Spain
- Institute of Biocomputation and Physics of Complex Systems (BIFI-IQFR and CBsC-CSIC Joint Units, University of Zaragoza, Zaragoza, Spain
| | - Patricia Ferreira
- Department of Biochemistry and Molecular and Cellular Biology, Faculty of Sciences, University of Zaragoza, Zaragoza, Spain
- Institute of Biocomputation and Physics of Complex Systems (BIFI-IQFR and CBsC-CSIC Joint Units, University of Zaragoza, Zaragoza, Spain
| | - Milagros Medina
- Department of Biochemistry and Molecular and Cellular Biology, Faculty of Sciences, University of Zaragoza, Zaragoza, Spain
- Institute of Biocomputation and Physics of Complex Systems (BIFI-IQFR and CBsC-CSIC Joint Units, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
12
|
CHCHD4 (MIA40) and the mitochondrial disulfide relay system. Biochem Soc Trans 2021; 49:17-27. [PMID: 33599699 PMCID: PMC7925007 DOI: 10.1042/bst20190232] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/19/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Mitochondria are pivotal for normal cellular physiology, as they perform a crucial role in diverse cellular functions and processes, including respiration and the regulation of bioenergetic and biosynthetic pathways, as well as regulating cellular signalling and transcriptional networks. In this way, mitochondria are central to the cell's homeostatic machinery, and as such mitochondrial dysfunction underlies the pathology of a diverse range of diseases including mitochondrial disease and cancer. Mitochondrial import pathways and targeting mechanisms provide the means to transport into mitochondria the hundreds of nuclear-encoded mitochondrial proteins that are critical for the organelle's many functions. One such import pathway is the highly evolutionarily conserved disulfide relay system (DRS) within the mitochondrial intermembrane space (IMS), whereby proteins undergo a form of oxidation-dependent protein import. A central component of the DRS is the oxidoreductase coiled-coil-helix-coiled-coil-helix (CHCH) domain-containing protein 4 (CHCHD4, also known as MIA40), the human homologue of yeast Mia40. Here, we summarise the recent advances made to our understanding of the role of CHCHD4 and the DRS in physiology and disease, with a specific focus on the emerging importance of CHCHD4 in regulating the cellular response to low oxygen (hypoxia) and metabolism in cancer.
Collapse
|
13
|
Letkovska K, Babal P, Cierna Z, Schmidtova S, Liskova V, Kalavska K, Miskovska V, Horak S, Rejlekova K, Chovanec M, Mardiak J, Janega P, Mego M. Prognostic Value of Apoptosis-Inducing Factor (AIF) in Germ Cell Tumors. Cancers (Basel) 2021; 13:cancers13040776. [PMID: 33668443 PMCID: PMC7917670 DOI: 10.3390/cancers13040776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 01/28/2023] Open
Abstract
Simple Summary Germ cell tumors (GCTs) are the most common solid malignancies in young men. GCTs are extraordinary sensitive to chemotherapy and represent a model of curable cancer. However, in a small proportion of patients the disease progresses or relapses despite administration of salvage chemotherapy. Apoptosis is a form of programmed cell death that occurs in multicellular organisms. It is well established that dysregulation of apoptosis plays an important role in pathogenesis of malignant diseases and may be associated with tumor progression and resistance to cytotoxic treatment. This study aimed to evaluate expression of apoptosis inducing factor (AIF) in GCTs. We observed lower AIF expression in GCTs compared to normal testicular tissue. We also showed prognostic significance of AIF in GCTs. AIF downregulation might represent one of the mechanisms of inhibition of apoptosis with subsequent facilitation of cell survival and metastatic dissemination of GCTs and perhaps could serve as a potential therapeutic target. Abstract Apoptosis is a strictly regulated process essential for preservation of tissue homeostasis. This study aimed to evaluate expression of apoptosis inducing factor (AIF) in testicular germ cell tumors (GCTs) and to correlate expression patterns with clinicopathological variables. Formalin-fixed and paraffin-embedded specimens of non-neoplastic testicular tissue and GCTs obtained from 216 patients were included in the study. AIF expression was detected by immunohistochemistry, scored by the multiplicative quickscore method (QS). Normal testicular tissue exhibits higher cytoplasmic granular expression of AIF compared to GCTs (mean QS = 12.77 vs. 4.80, p < 0.0001). Among invasive GCTs, mean QS was the highest in embryonal carcinoma, yolk sac tumor and seminoma, lower in teratoma and the lowest in choriocarcinoma. No nuclear translocation of AIF was observed. Nonpulmonary visceral metastases were associated with lower AIF expression. Metastatic GCTs patients with high AIF expression had better overall survival compared to patients with low AIF expression (HR = 0.26, 95% CI 0.11–0.62, p = 0.048). We observed significantly lower AIF expression in GCTs compared to normal testicular tissue, which is an uncommon finding in malignant tumors. AIF downregulation might represent one of the mechanisms of inhibition of apoptosis and promotion of cell survival in GCTs.
Collapse
Affiliation(s)
- Katarina Letkovska
- Department of Pathology, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia; (K.L.); (P.B.); (Z.C.); (S.H.); (P.J.)
| | - Pavel Babal
- Department of Pathology, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia; (K.L.); (P.B.); (Z.C.); (S.H.); (P.J.)
| | - Zuzana Cierna
- Department of Pathology, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia; (K.L.); (P.B.); (Z.C.); (S.H.); (P.J.)
- Department of Pathology, Faculty Hospital, A. Zarnova, 917 75 Trnava, Slovakia
| | - Silvia Schmidtova
- Translational Research Unit, 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, 833 10 Bratislava, Slovakia; (S.S.); (K.K.)
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, 814 39 Bratislava, Slovakia
| | - Veronika Liskova
- Institute of Clinical and Translational Research, Biomedical Research Center of the Slovak Academy of Sciences, 845 05 Bratislava, Slovakia;
| | - Katarína Kalavska
- Translational Research Unit, 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, 833 10 Bratislava, Slovakia; (S.S.); (K.K.)
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, 814 39 Bratislava, Slovakia
| | - Vera Miskovska
- 1st Department of Oncology, Comenius University, Faculty of Medicine, St. Elisabeth Cancer Institute, 812 50 Bratislava, Slovakia;
| | - Samuel Horak
- Department of Pathology, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia; (K.L.); (P.B.); (Z.C.); (S.H.); (P.J.)
| | - Katarina Rejlekova
- 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, 833 10 Bratislava, Slovakia; (K.R.); (M.C.); (J.M.)
| | - Michal Chovanec
- 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, 833 10 Bratislava, Slovakia; (K.R.); (M.C.); (J.M.)
| | - Jozef Mardiak
- 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, 833 10 Bratislava, Slovakia; (K.R.); (M.C.); (J.M.)
| | - Pavel Janega
- Department of Pathology, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia; (K.L.); (P.B.); (Z.C.); (S.H.); (P.J.)
| | - Michal Mego
- Translational Research Unit, 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, 833 10 Bratislava, Slovakia; (S.S.); (K.K.)
- 2nd Department of Oncology, Comenius University, Faculty of Medicine, National Cancer Institute, 833 10 Bratislava, Slovakia; (K.R.); (M.C.); (J.M.)
- Correspondence: ; Tel.: +421-2-59378366; Fax: +421-2-54774943
| |
Collapse
|
14
|
Zhang M, Zhang X, Chu X, Cheng L, Cai C. Long non-coding RNA MALAT1 plays a protective role in bronchopulmonary dysplasia via the inhibition of apoptosis and interaction with the Keap1/Nrf2 signal pathway. Transl Pediatr 2021; 10:265-275. [PMID: 33708512 PMCID: PMC7944181 DOI: 10.21037/tp-20-200] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a common respiratory disease in premature infants and is characterized by alveolar and pulmonary vascular dysplasia. Long-term oxygen exposure can cause BPD in preterm infants. Numerous studies have shown that long non-coding ribonucleic acid (lncRNA) is involved in the process of biological metabolism; however, its role in the development of BPD is unclear. Apoptosis-induced factor (AIF) is a key component involved in apoptosis. The Kelch-like ECH-associated protein 1/nuclear factor erythroid-2-related factor 2 (Keap1/Nrf2) signaling pathway is a body-derived antioxidant signaling pathway. METHODS In this study, the relative expression of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), AIF, Keap1, and Nrf2 was detected by real-time polymerase chain reaction (PCR). Also, the apoptosis of A549 cells was detected by flow cytometry. RESULTS The results showed that, compared to the control group, the expression of MALAT1 increased significantly, and AIF decreased substantially in BPD premature infants. In the A549 hyperoxic lung injury model, compared with the air group, the expression of MALAT1 in the hyperoxia group decreased markedly, while the expression of Keap1 and Nrf2 increased considerably. Furthermore, compared with the control plasmid transfection air group (NC group), the expression of Keap1 and Nrf2 increased significantly in the small interfering RNA (siRNA) group. CONCLUSIONS These results indicate that MALAT1 can play a protective role in BPD via the reduction of apoptosis and anti-oxidation, offering clinicians a new way to prevent and treat BPD.
Collapse
Affiliation(s)
- Min Zhang
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyue Zhang
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyun Chu
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lihua Cheng
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Cai
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Herrmann JM, Riemer J. Apoptosis inducing factor and mitochondrial NADH dehydrogenases: redox-controlled gear boxes to switch between mitochondrial biogenesis and cell death. Biol Chem 2020; 402:289-297. [PMID: 32769219 DOI: 10.1515/hsz-2020-0254] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023]
Abstract
The mitochondrial complex I serves as entry point for NADH into the electron transport chain. In animals, fungi and plants, additional NADH dehydrogenases carry out the same electron transfer reaction, however they do not pump protons. The apoptosis inducing factor (AIF, AIFM1 in humans) is a famous member of this group as it was the first pro-apoptotic protein identified that can induce caspase-independent cell death. Recent studies on AIFM1 and the NADH dehydrogenase Nde1 of baker's yeast revealed two independent and experimentally separable activities of this class of enzymes: On the one hand, these proteins promote the functionality of mitochondrial respiration in different ways: They channel electrons into the respiratory chain and, at least in animals, promote the import of Mia40 (named MIA40 or CHCHD4 in humans) and the assembly of complex I. On the other hand, they can give rise to pro-apoptotic fragments that are released from the mitochondria to trigger cell death. Here we propose that AIFM1 and Nde1 serve as conserved redox switches which measure metabolic conditions on the mitochondrial surface and translate it into a binary life/death decision. This function is conserved among eukaryotic cells and apparently used to purge metabolically compromised cells from populations.
Collapse
Affiliation(s)
- Johannes M Herrmann
- Department of Cell Biology, University of Kaiserslautern, Erwin-Schrödinger-Strasse 13, D-67663Kaiserslautern, Germany
| | - Jan Riemer
- Department of Biochemistry, University of Cologne, Zülpicher Str. 47A, D-50674Cologne, Germany
| |
Collapse
|
16
|
Sahu P, Mohan KV, Aggarwal S, Arindkar S, Mahesh Kumar J, Kumar Upadhyay P, Ramakrishna G, Nagarajan P. Apoptosis-inducing factor deficient mice fail to develop hepatic steatosis under high fat high fructose diet or bile duct ligation. Cell Biochem Funct 2020; 39:296-307. [PMID: 32767404 DOI: 10.1002/cbf.3579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/22/2020] [Accepted: 07/05/2020] [Indexed: 11/08/2022]
Abstract
Apoptosis-inducing factor (AIF) is a mitochondrial flavoprotein involved in redox signalling and programmed cell death. The role of AIF has been well recognized in diabetes and obesity. However, the aspect of AIF deficiency in the development of hepatic steatosis and liver injury is unknown. Therefore, in the current study, Harlequin (Hq mutant) mouse with markedly reduced content of AIF was investigated to explore the role of AIF on the initiation of liver injury. The wild type (WT) developed physiological and pathological features of non-alcoholic fatty liver disease (NAFLD) that were not seen in the Hq mice with AIF deficiency, when fed on high fat high fructose (HFHF) diet. Following bile duct ligation (BDL), the liver associated pathological changes were less conspicuous in Hq mice as compared to WT mice. The expression of AIF protein and apoptosis was markedly lesser as compared to their respective control in Hq mice on HFHF diet. Furthermore, the genes involved in fatty acid metabolism were also altered in the group of treated Hq mice. In conclusion, Hq mice failed to develop diet induced hepatic steatosis, suggestive of a role of AIF mediated pathway in the initiation and progression of liver inflammation. Thus, partial loss of AIF appears to be hepatoprotective. SIGNIFICANCE OF THE STUDY: AIF deficiency has multiple roles in altered pathology processes and cellular metabolism, thereby compromising the cellular homeostasis. Considering the molecular functions of AIF in other organ pathology little is known about its role in diet induced liver injury. Hence, the aim of the current study was to investigate the role of AIF deficiency in liver injury and diseases with focus on NAFLD. The study will help to deliniate the mechanisms of NAFLD using Harliquin Mice.
Collapse
Affiliation(s)
- Parul Sahu
- National Institute of Immunology, New Delhi, India
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Sirt3 Exerts Its Tumor-Suppressive Role by Increasing p53 and Attenuating Response to Estrogen in MCF-7 Cells. Antioxidants (Basel) 2020; 9:antiox9040294. [PMID: 32244715 PMCID: PMC7222218 DOI: 10.3390/antiox9040294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Estrogen (E2) is a major risk factor for the initiation and progression of malignancy in estrogen receptor (ER) positive breast cancers, whereas sirtuin 3 (Sirt3), a major mitochondrial NAD+-dependent deacetylase, has the inhibitory effect on the tumorigenic properties of ER positive MCF-7 breast cancer cells. Since it is unclear if this effect is mediated through the estrogen receptor alpha (ERα) signaling pathway, in this study, we aimed to determine if the tumor-suppressive function of Sirt3 in MCF-7 cells interferes with their response to E2. Although we found that Sirt3 improves the antioxidative response and mitochondrial fitness of the MCF-7 cells, it also increases DNA damage along with p53, AIF, and ERα expression. Moreover, Sirt3 desensitizes cells to the proliferative effect of E2, affects p53 by disruption of the ERα–p53 interaction, and decreases proliferation, colony formation, and migration of the cells. Our observations indicate that these tumor-suppressive effects of Sirt3 could be reversed by E2 treatment only to a limited extent which is not sufficient to recover the tumorigenic properties of the MCF-7 cells. This study provides new and interesting insights with respect to the functional role of Sirt3 in the E2-dependent breast cancers.
Collapse
|
18
|
Kadam A, Mehta D, Jubin T, Mansuri MS, Begum R. Apoptosis inducing factor: Cellular protective function in Dictyostelium discoideum. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2020; 1861:148158. [PMID: 31991113 DOI: 10.1016/j.bbabio.2020.148158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 01/31/2023]
Abstract
Apoptosis Inducing Factor (AIF), a nuclear encoded mitochondrial inter-membrane space flavoprotein with intrinsic NADH oxidase activity, plays an important role in inducing cell death mechanisms. In response to cell death signals, it undergoes mitochondrio-nuclear translocation leading to DNA fragmentation. In addition to its role in cell death, AIF has a pro-survival role, wherein it contributes to the maintenance of mitochondrial structure and function in a coordinated manner. However, its exact mechanism of controlling mitochondrial homeostasis is unclear. The current study aims to explore the protective functions of AIF by its downregulation and overexpression in Dictyostelium discoideum. Constitutive AIF downregulated (dR) cells exhibited compromised oxidative phosphorylation along with elevated levels of cellular ROS. Interestingly, constitutive AIF dR cells showed amelioration in the activity of the ETC complexes upon antioxidant treatment, strengthening AIF's role as an ROS regulator, by virtue of its oxidoreductase property. Also, constitutive AIF dR cells showed lower transcript levels of the various subunits of ETC. Moreover, loss of AIF affected mtDNA content and mitochondrial fusion-fission mechanism, which subsequently caused morphometric mitochondrial alterations. Constitutive AIF overexpressed (OE) cells also showed higher cellular ROS and mitochondrial fission genes transcript levels along with reduced mitochondrial fusion genes transcript levels and mtDNA content. Thus, the results of the current study provide a paradigm where AIF is implicated in cell survival by maintaining mitochondrial bioenergetics, morphology and fusion-fission mechanism in D. discoideum, an evolutionarily significant model organism for mitochondrial diseases.
Collapse
Affiliation(s)
- Ashlesha Kadam
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Darshan Mehta
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Tina Jubin
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Mohmmad Shoab Mansuri
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India.
| |
Collapse
|
19
|
Monroe JD, Hodzic D, Millay MH, Patty BG, Smith ME. Anti-Cancer and Ototoxicity Characteristics of the Curcuminoids, CLEFMA and EF24, in Combination with Cisplatin. Molecules 2019; 24:molecules24213889. [PMID: 31671767 PMCID: PMC6864451 DOI: 10.3390/molecules24213889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/23/2022] Open
Abstract
In this study, we investigated whether the curcuminoids, CLEFMA and EF24, improved cisplatin efficacy and reduced cisplatin ototoxicity. We used the lung cancer cell line, A549, to determine the effects of the curcuminoids and cisplatin on cell viability and several apoptotic signaling mechanisms. Cellular viability was measured using the MTT assay. A scratch assay was used to measure cell migration and fluorescent spectrophotometry to measure reactive oxygen species (ROS) production. Western blots and luminescence assays were used to measure the expression and activity of apoptosis-inducing factor (AIF), caspases-3/7, -8, -9, and -12, c-Jun N-terminal kinases (JNK), mitogen-activated protein kinase (MAPK), and proto-oncogene tyrosine-protein kinase (Src). A zebrafish model was used to evaluate auditory effects. Cisplatin, the curcuminoids, and their combinations had similar effects on cell viability (IC50 values: 2-16 μM) and AIF, caspase-12, JNK, MAPK, and Src expression, while caspase-3/7, -8, and -9 activity was unchanged or decreased. Cisplatin increased ROS yield (1.2-fold), and curcuminoid and combination treatments reduced ROS (0.75-0.85-fold). Combination treatments reduced A549 migration (0.51-0.53-fold). Both curcuminoids reduced auditory threshold shifts induced by cisplatin. In summary, cisplatin and the curcuminoids might cause cell death through AIF and caspase-12. The curcuminoids may potentiate cisplatin's effect against A549 migration, but may counteract cisplatin's effect to increase ROS production. The curcuminoids might also prevent cisplatin ototoxicity.
Collapse
Affiliation(s)
- Jerry D Monroe
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, USA.
| | - Denis Hodzic
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, USA.
| | - Matthew H Millay
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, USA.
| | - Blaine G Patty
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, USA.
| | - Michael E Smith
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, USA.
| |
Collapse
|
20
|
Sinha S, Renganathan A, Nagendra PB, Bhat V, Mathew BS, Rao MRS. AEBP1 down regulation induced cell death pathway depends on PTEN status of glioma cells. Sci Rep 2019; 9:14577. [PMID: 31601918 PMCID: PMC6787275 DOI: 10.1038/s41598-019-51068-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common aggressive form of brain cancer with overall dismal prognosis (10–12 months) despite all current multimodal treatments. Previously we identified adipocyte enhancer binding protein 1 (AEBP1) as a differentially regulated gene in GBM. On probing the role of AEBP1 over expression in glioblastoma, we found that both cellular proliferation and survival were affected upon AEBP1 silencing in glioma cells, resulting in cell death. In the present study we report that the classical caspase pathway components are not activated in cell death induced by AEBP1 down regulation in PTEN-deficient (U87MG and U138MG) cells. PARP-1 was not cleaved but over-activated under AEBP1 down regulation which leads to the synthesis of PAR in the nucleus triggering the release of AIF from the mitochondria. Subsequently, AIF translocates to the nucleus along with MIF causing chromatinolysis. AEBP1 positively regulates PI3KinaseCβ by the binding to AE-1 binding element in the PI3KinaseCβ promoter. Loss of PI3KinaseCβ expression under AEBP1 depleted condition leads to excessive DNA damage and activation of PARP-1. Furthermore, over expression of PIK3CB (in trans) in U138MG cells prevents DNA damage in these AEBP1 depleted cells. On the contrary, AEBP1 down regulation induces caspase-dependent cell death in PTEN-proficient (LN18 and LN229) cells. Ectopic expression of wild-type PTEN in PTEN-deficient U138MG cells results in the activation of canonical caspase and Akt dependent cell death. Collectively, our findings define AEBP1 as a potential oncogenic driver in glioma, with potential implications for therapeutic intervention.
Collapse
Affiliation(s)
- Swati Sinha
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, Karnataka, 560064, India
| | - Arun Renganathan
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, Karnataka, 560064, India.,Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Prathima B Nagendra
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, Karnataka, 560064, India.,Gynaecology Oncology Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Vasudeva Bhat
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, Karnataka, 560064, India.,Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Brian Steve Mathew
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, Karnataka, 560064, India
| | | |
Collapse
|
21
|
SNHG15 is a bifunctional MYC-regulated noncoding locus encoding a lncRNA that promotes cell proliferation, invasion and drug resistance in colorectal cancer by interacting with AIF. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:172. [PMID: 31014355 PMCID: PMC6480895 DOI: 10.1186/s13046-019-1169-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 04/07/2019] [Indexed: 02/08/2023]
Abstract
Background Thousands of long noncoding RNAs (lncRNAs) are aberrantly expressed in various types of cancers, however our understanding of their role in the disease is still very limited. Methods We applied RNAseq analysis from patient-derived data with validation in independent cohort of patients. We followed these studies with gene regulation analysis as well as experimental dissection of the role of the identified lncRNA by multiple in vitro and in vivo methods. Results We analyzed RNA-seq data from tumors of 456 CRC patients compared to normal samples, and identified SNHG15 as a potentially oncogenic lncRNA that encodes a snoRNA in one of its introns. The processed SNHG15 is overexpressed in CRC tumors and its expression is highly correlated with poor survival of patients. Interestingly, SNHG15 is more highly expressed in tumors with high levels of MYC expression, while MYC protein binds to two E-box motifs on SNHG15 sequence, indicating that SNHG15 transcription is directly regulated by the oncogene MYC. The depletion of SNHG15 by siRNA or CRISPR-Cas9 inhibits cell proliferation and invasion, decreases colony formation as well as the tumorigenic capacity of CRC cells, whereas its overexpression leads to opposite effects. Gene expression analysis performed upon SNHG15 inhibition showed changes in multiple relevant genes implicated in cancer progression, including MYC, NRAS, BAG3 or ERBB3. Several of these genes are functionally related to AIF, a protein that we found to specifically interact with SNHG15, suggesting that the SNHG15 acts, at least in part, by regulating the activity of AIF. Interestingly, ROS levels, which are directly regulated by AIF, show a significant reduction in SNHG15-depleted cells. Moreover, knockdown of SNHG15 increases the sensitiveness of the cells to 5-FU, while its overexpression renders them more resistant to the chemotherapeutic drug. Conclusion Altogether, these results describe an important role of SNHG15 in promoting colon cancer and mediating drug resistance, suggesting its potential as prognostic marker and target for RNA-based therapies. Electronic supplementary material The online version of this article (10.1186/s13046-019-1169-0) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Gu B, Liu H, Han Y, Chen Y, Jiang H. Integrated analysis of miRNA and mRNA expression profiles in 2-, 6-, and 12-month-old Small Tail Han Sheep ovaries reveals that oar-miR-432 downregulates RPS6KA1 expression. Gene 2019; 710:76-90. [PMID: 30898702 DOI: 10.1016/j.gene.2019.02.095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/23/2019] [Accepted: 02/28/2019] [Indexed: 01/08/2023]
Abstract
Small Tail Han Sheep are an excellent local sheep breed in China, and their outstanding reproductive performance is one of their very important biological characteristics. Clarifying the ovary development process of these ewes should provide a theoretical basis for improving their reproductive efficiency. In this study, we identified the differentially expressed (DE) microRNAs (miRNAs) in 2-, 6-, and 12-month-old small-tail Han sheep ovaries by constructing and analyzing the miRNA expression profiles. These findings clarify the molecular mechanisms regulating the excellent reproductive performance of small-tail Han ewes. We used RNA-Seq technology and bioinformatic to analyze these profiles. Eleven, 13, and 19 DE miRNAs were identified in the 2- vs 6-, 6- vs 12-, and 2- vs 12-month-old ovaries, respectively. In total, 54, 37, and 198 predicted target genes of these DE miRNAs were identified in these three groups, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that in the 2- vs 6-month-old ovaries, the target genes of DE known sheep miRNAs were involved in 102 GO terms and seven signaling pathways; in the 6- vs 12-month-old ovaries, the target genes of DE known sheep miRNAs were involved in 52 GO terms and three signaling pathways; and in the 2- vs 12-month-old ovaries, the target genes of DE known sheep miRNAs were involved in 88 GO terms and six signaling pathways. Three miRNA-target regulatory networks were constructed based on these DE miRNA-target interactions. Nine miRNAs were selected to confirm to the accuracy of the miRNA sequencing data with qRT-PCR. The site at which oar-miR-432 binds RPS6KA1 was determined with a dual-luciferase system. This is the first integrated analysis the expression profiles of miRNAs and their targets during ovarian development in small-tail Han sheep. These data clarify the molecular regulatory mechanisms underlying sheep ovarian development and identify biomarkers that influence the reproductive performance of small-tail Han ewes.
Collapse
Affiliation(s)
- Bo Gu
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Hang Liu
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Yue Han
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Yang Chen
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Huaizhi Jiang
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China.
| |
Collapse
|
23
|
Autophagy protects against redox-active trace metal-induced cell death in rabbit synovial fibroblasts through Toll-like receptor 4 activation. Exp Cell Res 2019; 374:19-28. [DOI: 10.1016/j.yexcr.2018.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/22/2018] [Accepted: 11/03/2018] [Indexed: 12/18/2022]
|
24
|
Timper K, Paeger L, Sánchez-Lasheras C, Varela L, Jais A, Nolte H, Vogt MC, Hausen AC, Heilinger C, Evers N, Pospisilik JA, Penninger JM, Taylor EB, Horvath TL, Kloppenburg P, Brüning JC. Mild Impairment of Mitochondrial OXPHOS Promotes Fatty Acid Utilization in POMC Neurons and Improves Glucose Homeostasis in Obesity. Cell Rep 2018; 25:383-397.e10. [PMID: 30304679 PMCID: PMC6349418 DOI: 10.1016/j.celrep.2018.09.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/06/2018] [Accepted: 09/11/2018] [Indexed: 01/19/2023] Open
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) and substrate utilization critically regulate the function of hypothalamic proopiomelanocortin (POMC)-expressing neurons. Here, we demonstrate that inactivation of apoptosis-inducing factor (AIF) in POMC neurons mildly impairs mitochondrial respiration and decreases firing of POMC neurons in lean mice. In contrast, under diet-induced obese conditions, POMC-Cre-specific inactivation of AIF prevents obesity-induced silencing of POMC neurons, translating into improved glucose metabolism, improved leptin, and insulin sensitivity, as well as increased energy expenditure in AIFΔPOMC mice. On a cellular level, AIF deficiency improves mitochondrial morphology, facilitates the utilization of fatty acids for mitochondrial respiration, and increases reactive oxygen species (ROS) formation in POMC neurons from obese mice, ultimately leading to restored POMC firing upon HFD feeding. Collectively, partial impairment of mitochondrial function shifts substrate utilization of POMC neurons from glucose to fatty acid metabolism and restores their firing properties, resulting in improved systemic glucose and energy metabolism in obesity.
Collapse
Affiliation(s)
- Katharina Timper
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Lars Paeger
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Biocenter, University of Cologne, Cologne, Germany
| | - Carmen Sánchez-Lasheras
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Luis Varela
- Department of Biochemistry and Fraternal Order of Eagles Diabetes Research Center, Caver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Alexander Jais
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Hendrik Nolte
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Merly C Vogt
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - A Christine Hausen
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Christian Heilinger
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Nadine Evers
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - J Andrew Pospisilik
- Max-Planck-Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Eric B Taylor
- Department of Biochemistry and Fraternal Order of Eagles Diabetes Research Center, Caver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Tamas L Horvath
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Biocenter, University of Cologne, Cologne, Germany
| | - Jens Claus Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
25
|
Scott AJ, Walker SA, Krank JJ, Wilkinson AS, Johnson KM, Lewis EM, Wilkinson JC. AIF promotes a JNK1-mediated cadherin switch independently of respiratory chain stabilization. J Biol Chem 2018; 293:14707-14722. [PMID: 30093403 DOI: 10.1074/jbc.ra118.004022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/26/2018] [Indexed: 12/18/2022] Open
Abstract
Apoptosis-inducing factor (AIF) is a mitochondrial flavoprotein occasionally involved in cell death that primarily regulates mitochondrial energy metabolism under normal cellular conditions. AIF catalyzes the oxidation of NADH in vitro, yet the significance of this redox activity in cells remains unclear. Here, we show that through its enzymatic activity AIF is a critical factor for oxidative stress-induced activation of the mitogen-activated protein kinases JNK1 (c-Jun N-terminal kinase), p38, and ERK (extracellular signal-regulated kinase). AIF-dependent JNK1 signaling culminates in the cadherin switch, and genetic reversal of this switch leads to apoptosis when AIF is suppressed. Notably, this widespread ability of AIF to promote JNK signaling can be uncoupled from its more limited role in respiratory chain stabilization. Thus, AIF is a transmitter of extra-mitochondrial signaling cues with important implications for human development and disease.
Collapse
Affiliation(s)
- Andrew J Scott
- From the Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108 and
| | - Sierra A Walker
- From the Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108 and
| | - Joshua J Krank
- From the Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108 and
| | - Amanda S Wilkinson
- From the Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108 and
| | - Kaitlyn M Johnson
- From the Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108 and
| | - Eric M Lewis
- the Department of Chemistry, Mathematics and Physics, Clarion University of Pennsylvania, Clarion, Pennsylvania 16214
| | - John C Wilkinson
- From the Department of Chemistry and Biochemistry, North Dakota State University, Fargo, North Dakota 58108 and
| |
Collapse
|
26
|
Lee JE, Kwon YJ, Baek HS, Ye DJ, Cho E, Choi HK, Oh KS, Chun YJ. Synergistic induction of apoptosis by combination treatment with mesupron and auranofin in human breast cancer cells. Arch Pharm Res 2017; 40:746-759. [DOI: 10.1007/s12272-017-0923-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 05/26/2017] [Indexed: 12/14/2022]
|
27
|
Ramsay RR, Majekova M, Medina M, Valoti M. Key Targets for Multi-Target Ligands Designed to Combat Neurodegeneration. Front Neurosci 2016; 10:375. [PMID: 27597816 PMCID: PMC4992697 DOI: 10.3389/fnins.2016.00375] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022] Open
Abstract
HIGHLIGHTS Compounds that interact with multiple targets but minimally with the cytochrome P450 system (CYP) address the many factors leading to neurodegeneration.Acetyl- and Butyryl-cholineEsterases (AChE, BChE) and Monoamine Oxidases A/B (MAO A, MAO B) are targets for Multi-Target Designed Ligands (MTDL).ASS234 is an irreversible inhibitor of MAO A >MAO B and has micromolar potency against the cholinesterases.ASS234 is a poor CYP substrate in human liver, yielding the depropargylated metabolite.SMe1EC2, a stobadine derivative, showed high radical scavenging property, in vitro and in vivo giving protection in head trauma and diabetic damage of endothelium.Control of mitochondrial function and morphology by manipulating fission and fusion is emerging as a target area for therapeutic strategies to decrease the pathological outcome of neurodegenerative diseases. Growing evidence supports the view that neurodegenerative diseases have multiple and common mechanisms in their aetiologies. These multifactorial aspects have changed the broadly common assumption that selective drugs are superior to "dirty drugs" for use in therapy. This drives the research in studies of novel compounds that might have multiple action mechanisms. In neurodegeneration, loss of neuronal signaling is a major cause of the symptoms, so preservation of neurotransmitters by inhibiting the breakdown enzymes is a first approach. Acetylcholinesterase (AChE) inhibitors are the drugs preferentially used in AD and that one of these, rivastigmine, is licensed also for PD. Several studies have shown that monoamine oxidase (MAO) B, located mainly in glial cells, increases with age and is elevated in Alzheimer (AD) and Parkinson's Disease's (PD). Deprenyl, a MAO B inhibitor, significantly delays the initiation of levodopa treatment in PD patients. These indications underline that AChE and MAO are considered a necessary part of multi-target designed ligands (MTDL). However, both of these targets are simply symptomatic treatment so if new drugs are to prevent degeneration rather than compensate for loss of neurotransmitters, then oxidative stress and mitochondrial events must also be targeted. MAO inhibitors can protect neurons from apoptosis by mechanisms unrelated to enzyme inhibition. Understanding the involvement of MAO and other proteins in the induction and regulation of the apoptosis in mitochondria will aid progress toward strategies to prevent the loss of neurons. In general, the oxidative stress observed both in PD and AD indicate that antioxidant properties are a desirable part of MTDL molecules. After two or more properties are incorporated into one molecule, the passage from a lead compound to a therapeutic tool is strictly linked to its pharmacokinetic and toxicity. In this context the interaction of any new molecules with cytochrome P450 and other xenobiotic metabolic processes is a crucial point. The present review covers the biochemistry of enzymes targeted in the design of drugs against neurodegeneration and the cytochrome P450-dependent metabolism of MTDLs.
Collapse
Affiliation(s)
- Rona R. Ramsay
- Biomedical Sciences Research Complex, University of St. AndrewsSt. Andrews, UK
| | - Magdalena Majekova
- Department of Biochemical Pharmacology, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of SciencesBratislava, Slovakia
| | - Milagros Medina
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias and BIFI, Universidad de ZaragozaZaragoza, Spain
| | - Massimo Valoti
- Dipartimento di Scienze della Vita, Università degli Studi di SienaSiena, Italy
| |
Collapse
|
28
|
High expression of apoptosis-inducing factor, mitochondrion-associated 3 (AIFM3) in human cholangiocarcinoma. Tumour Biol 2016; 37:13659-13667. [DOI: 10.1007/s13277-016-5204-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/13/2016] [Indexed: 12/27/2022] Open
|
29
|
Lenhausen AM, Wilkinson AS, Lewis EM, Dailey KM, Scott AJ, Khan S, Wilkinson JC. Apoptosis Inducing Factor Binding Protein PGAM5 Triggers Mitophagic Cell Death That Is Inhibited by the Ubiquitin Ligase Activity of X-Linked Inhibitor of Apoptosis. Biochemistry 2016; 55:3285-302. [PMID: 27218139 DOI: 10.1021/acs.biochem.6b00306] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Apoptosis inducing factor (AIF) plays a well-defined role in controlling cell death but is also a critical factor for maintaining mitochondrial energy homeostasis; how these dueling activities are balanced has remained largely elusive. To identify new AIF binding partners that may define the continuum of AIF cellular regulation, a biochemical screen was performed that identified the mitochondrial phosphoglycerate mutase 5 (PGAM5) as an AIF associated factor. AIF binds both the short and long isoforms of PGAM5 and can reduce the ability of PGAM5 to control antioxidant responses. Transient overexpression of either PGAM5 isoform triggers caspase activation and cell death, and while AIF could reduce this caspase activation neither AIF expression nor caspase activity is required for PGAM5-mediated death. PGAM5 toxicity morphologically and biochemically resembles mitophagic cell death and is inhibited by the AIF binding protein X-linked inhibitor of apoptosis (XIAP) in a manner that depends on the ubiquitin ligase activity of XIAP. The phosphatase activity of PGAM5 was not required for cell death, and comparison of phosphatase activity between short and long PGAM5 isoforms suggested that only the long isoform is catalytically competent. This property correlated with an increased ability of PGAM5L to form dimers and/or higher order oligomers in intact cells compared to PGAM5S. Overall this study identifies an AIF/PGAM5/XIAP axis that can regulate PGAM5 activities related to the antioxidant response and mitophagy.
Collapse
Affiliation(s)
- Audrey M Lenhausen
- Department of Biochemistry, Wake Forest School of Medicine , Winston-Salem, North Carolina 27157, United States
| | - Amanda S Wilkinson
- Department of Chemistry and Biochemistry, North Dakota State University , Fargo, North Dakota 58108, United States
| | - Eric M Lewis
- Department of Biochemistry, Wake Forest School of Medicine , Winston-Salem, North Carolina 27157, United States
| | - Kaitlin M Dailey
- Department of Chemistry and Biochemistry, North Dakota State University , Fargo, North Dakota 58108, United States
| | - Andrew J Scott
- Department of Chemistry and Biochemistry, North Dakota State University , Fargo, North Dakota 58108, United States
| | - Shahzeb Khan
- Department of Chemistry and Biochemistry, North Dakota State University , Fargo, North Dakota 58108, United States
| | - John C Wilkinson
- Department of Chemistry and Biochemistry, North Dakota State University , Fargo, North Dakota 58108, United States
| |
Collapse
|
30
|
Basal metabolic state governs AIF-dependent growth support in pancreatic cancer cells. BMC Cancer 2016; 16:286. [PMID: 27108222 PMCID: PMC4841948 DOI: 10.1186/s12885-016-2320-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/15/2016] [Indexed: 01/15/2023] Open
Abstract
Background Apoptosis-inducing factor (AIF), named for its involvement in cell death pathways, is a mitochondrial protein that regulates metabolic homeostasis. In addition to supporting the survival of healthy cells, AIF also plays a contributory role to the development of cancer through its enzymatic activity, and we have previously shown that AIF preferentially supports advanced-stage prostate cancer cells. Here we further evaluated the role of AIF in tumorigenesis by exploring its function in pancreatic cancer, a disease setting that most often presents at an advanced stage by the time of diagnosis. Methods A bioinformatics approach was first employed to investigate AIF mRNA transcript levels in pancreatic tumor specimens vs. normal tissues. AIF-deficient pancreatic cancer cell lines were then established via lentiviral infection. Immunoblot analysis was used to determine relative protein quantities within cells. Cell viability was measured by flow cytometry; in vitro and Matrigel™ growth/survival using Coulter™ counting and phase contrast microscopy; and glucose consumption in the absence and presence of Matrigel™ using spectrophotometric methods. Results Archival gene expression data revealed a modest elevation of AIF transcript levels in subsets of pancreatic tumor specimens, suggesting a possible role in disease progression. AIF expression was then suppressed in a panel of five pancreatic cancer cell lines that display diverse metabolic phenotypes. AIF ablation selectively crippled the growth of cells in vitro in a manner that directly correlated with the loss of mitochondrial respiratory chain subunits and altered glucose metabolism, and these effects were exacerbated in the presence of Matrigel™ substrate. This suggests a critical metabolic role for AIF to pancreatic tumorigenesis, while the spectrum of sensitivities to AIF ablation depends on basal cellular metabolic phenotypes. Conclusions Altogether these data indicate that AIF supports the growth and survival of metabolically defined pancreatic cancer cells and that this metabolic function may derive from a novel mechanism so far undocumented in other cancer types.
Collapse
|
31
|
Arif T, Krelin Y, Shoshan-Barmatz V. Reducing VDAC1 expression induces a non-apoptotic role for pro-apoptotic proteins in cancer cell differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1228-1242. [PMID: 27080741 DOI: 10.1016/j.bbabio.2016.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 12/30/2022]
Abstract
Proteins initially identified as essential for apoptosis also mediate a wide range of non-apoptotic functions that include cell cycle progression, differentiation and metabolism. As this phenomenon was mostly reported with non-cancer cells, we considered non-conventional roles for the apoptotic machinery in the cancer setting. We found that treating glioblastoma (GBM) tumors with siRNA against VDAC1, a mitochondrial protein found at the crossroads of metabolic and survival pathways and involved in apoptosis, inhibited tumor growth while leading to differentiation of tumor cells into neuronal-like cells, as reflected in the expression of specific markers. Although VDAC1 depletion did not induce apoptosis, the expression levels of several pro-apoptotic regulatory proteins were changed. Specifically, VDAC1 deletion led to up-regulation of caspases, p53, cytochrome c, and down-regulation of SMAC/Diablo, AIF and TSPO. The down-regulated group was highly expressed in U-87MG xenografts, as well as in GBMs from human patients. We also showed that the rewired cancer-cell metabolism resulting from VDAC1 depletion reinforced cell growth arrest and differentiation via alterations in the transcription factors p53, c-Myc, HIF-1α and NF-κB. The decrease in c-Myc, HIF-1α and NF-κB levels was in accord with reduced cell proliferation, whereas increased p53 expression promoted differentiation. Thus, upon metabolic re-programing induced by VDAC1 depletion, the levels of pro-apoptotic proteins associated with cell growth decreased, while those connected to cell differentiation increased, converting GBM cells into astrocyte- and neuron-like cells. The results reveal that in tumors, pro-apoptotic proteins can perform non-apoptotic functions, acting as regulators of cell growth and differentiation, making these molecules potential new targets for cancer therapy. 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)
- Tasleem Arif
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yakov Krelin
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Varda Shoshan-Barmatz
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| |
Collapse
|
32
|
Amigoni L, Frigerio G, Martegani E, Colombo S. Involvement of Aif1 in apoptosis triggered by lack of Hxk2 in the yeastSaccharomyces cerevisiae. FEMS Yeast Res 2016; 16:fow016. [DOI: 10.1093/femsyr/fow016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2016] [Indexed: 12/26/2022] Open
|
33
|
Fowler PA, Filis P, Bhattacharya S, le Bizec B, Antignac JP, Morvan ML, Drake AJ, Soffientini U, O'Shaughnessy PJ. Human anogenital distance: an update on fetal smoke-exposure and integration of the perinatal literature on sex differences. Hum Reprod 2016; 31:463-72. [PMID: 26732622 PMCID: PMC4716811 DOI: 10.1093/humrep/dev323] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 11/20/2015] [Indexed: 01/08/2023] Open
Abstract
STUDY QUESTION Do sex and maternal smoking effects on human fetal anogenital distance (AGD) persist in a larger study and how do these data integrate with the wider literature on perinatal human AGD, especially with respect to sex differences? SUMMARY ANSWER Second trimester sex differences in AGD are broadly consistent with neonatal and infant measures of AGD and maternal cigarette smoking is associated with a temporary increase in male AGD in the absence of changes in circulating testosterone. WHAT IS KNOWN ALREADY AGD is a biomarker of fetal androgen exposure, a reduced AGD in males being associated with cryptorchidism, hypospadias and reduced penile length. Normative fetal AGD data remain partial and windows of sensitivity of human fetal AGD to disruption are not known. STUDY DESIGN, SIZE, DURATION The effects of fetal sex and maternal cigarette smoking on the second trimester (11–21 weeks of gestation) human fetal AGD were studied, along with measurement of testosterone and testicular transcripts associated with apoptosis and proliferation. PARTICIPANTS/MATERIALS, SETTING METHODS AGD, measured from the centre of the anus to the posterior/caudal root of penis/clitoris (AGDapp) was determined in 56 female and 70 male morphologically normal fetuses. These data were integrated with current literature on perinatal AGD in humans. MAIN RESULTS AND THE ROLE OF CHANCE At 11–13 weeks of gestation male fetal AGDapp was 61% (P< 0.001) longer than in females, increasing to 70% at 17–21 weeks. This sexual dimorphism was independent of growth characteristics (fetal weight, length, gonad weight). We confirmed that at 14–16 weeks of gestation male fetal AGDapp was increased 28% (P < 0.05) by in utero cigarette smoke exposure. Testosterone levels were not affected by smoking. To develop normative data, our findings have been integrated with available data from in vivo ultrasound scans and neonatal studies. Inter-study variations in male/female AGD differences lead to the conclusion that normalization and standardization approaches should be developed to enable confidence in comparing data from different perinatal AGD studies. LIMITATIONS, REASONS FOR CAUTION Sex differences, and a smoking-dependent increase in male fetal AGD at 14–16 weeks, identified in a preliminary study, were confirmed with a larger number of fetuses. However, human fetal AGD should, be re-assessed once much larger numbers of fetuses have been studied and this should be integrated with more detailed analysis of maternal lifestyle. Direct study of human fetal genital tissues is required for further mechanistic insights. WIDER IMPLICATIONS OF THE FINDINGS Fetal exposure to cigarette smoke chemicals is known to lead to reduced fertility in men and women. Integration of our data into the perinatal human AGD literature shows that more work needs to be done to enable reliable inter-study comparisons. STUDY FUNDING/COMPETING INTEREST(S) The study was supported by grants from the Chief Scientist Office (Scottish Executive, CZG/1/109 & CZG/4/742), NHS Grampian Endowments (08/02), the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no 212885 and the Medical Research Council, UK (MR/L010011/1). The authors declare they have no competing interests, be it financial, personal or professional.
Collapse
Affiliation(s)
- Paul A Fowler
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Panagiotis Filis
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Siladitya Bhattacharya
- Institute of Applied Health Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Bruno le Bizec
- USC INRA 1329 Laboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, Oniris, Nantes F-44307, France
| | - Jean-Philippe Antignac
- USC INRA 1329 Laboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, Oniris, Nantes F-44307, France
| | - Marie-Line Morvan
- USC INRA 1329 Laboratoire d'Etude des Résidus et Contaminants dans les Aliments, LUNAM Université, Oniris, Nantes F-44307, France
| | - Amanda J Drake
- Endocrinology Unit, Queen's Medical Research Institute, University/BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Ugo Soffientini
- Institute of Biodiversity, Animal Health & Comparative Medicine (IBAHCM), College of Medical, Veterinary & Life Sciences, University of Glasgow, Bearsden Rd, Glasgow G61 1QH, UK
| | - Peter J O'Shaughnessy
- Institute of Biodiversity, Animal Health & Comparative Medicine (IBAHCM), College of Medical, Veterinary & Life Sciences, University of Glasgow, Bearsden Rd, Glasgow G61 1QH, UK
| |
Collapse
|
34
|
Elvitigala DAS, Jayasooriya RGPT, Whang I, Lee J. First report on the gastropod proapoptotic AIF3 counterpart from disk abalone (Haliotis discus discus) deciphering its transcriptional modulation by induced pathogenic stress. FISH & SHELLFISH IMMUNOLOGY 2015; 47:697-705. [PMID: 26455649 DOI: 10.1016/j.fsi.2015.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/02/2015] [Accepted: 10/05/2015] [Indexed: 06/05/2023]
Abstract
Apoptosis inducing factor (AIF) is a flavoprotein that is involved in oxidative phosphorylation and induces apoptosis in eukaryotic cells. There are three isozymes of AIF that have been identified to date, designated as AIF1, AIF2, and AIF3; the human AIF3 is also known as an AIF-like protein (AIFL). This study aimed to identify and characterize a homologue of AIF3 from disk abalone (AbAIF3) that belongs to the phylum Mollusca. The open reading frame (ORF) of AbAIF3 is 1749 base pairs (bp) in length and encodes a protein of 583 amino acids, with a predicted molecular mass of 63.14 kDa. Based on our in-silico analysis, the AbAIF3 protein harbored the typical domain architecture as that of the known AIF family proteins, consisting of N-terminal Rieske and pyridine nucleotide-disulphide oxidoreductase domain. Comparative protein sequence analysis confirmed that AbAIF3 is a homolog of AIF3. Moreover, our phylogenetic analysis revealed that AbAIF3 had a close evolutionary relationship with the molluscan counterparts. Interestingly, AbAIF3 was shown to induce apoptosis in HEK293T cells using transfection assays followed by flow cytometric analysis. In addition, we found that AbAIF3 mRNA expression was ubiquitous in physiologically important tissues, and significantly modulated upon experimental immune stimulations in hemocytes. Collectively, our study illustrates the indispensable role of AbAIF3 in inducing apoptosis in disk abalones, which in turn might be involved in hosts' immune defense mechanisms against microbial infections.
Collapse
Affiliation(s)
- Don Anushka Sandaruwan Elvitigala
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea
| | - R G P T Jayasooriya
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea
| | - Ilson Whang
- Fish Vaccine Development Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea.
| |
Collapse
|
35
|
Chen TC, Yu MC, Chien CC, Wu MS, Lee YC, Chen YC. Nilotinib reduced the viability of human ovarian cancer cells via mitochondria-dependent apoptosis, independent of JNK activation. Toxicol In Vitro 2015; 31:1-11. [PMID: 26549707 DOI: 10.1016/j.tiv.2015.11.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 09/12/2015] [Accepted: 11/04/2015] [Indexed: 11/25/2022]
Abstract
Nilotinib (AMN) induces apoptosis in various cancer cells; however the effect of AMN on human ovarian cancer cells is still unclear. A reduction in cell viability associated with the occurrence of apoptotic characteristics was observed in human SKOV-3 ovarian cancer cells under AMN but not sorafenib (SORA) or imatinib (STI) stimulation. Activation of apoptotic pathway including increased caspase (Casp)-3 and poly(ADP-ribose) polymerase 1 (PARP1) protein cleavage by AMN was detected with disrupted mitochondrial membrane potential (MMP) accompanied by decreased Bcl-2 protein and increased cytosolic cytochrome (Cyt) c/cleaved Casp-9 protein expressions was found, and AMN-induced cell death was inhibited by peptidyl Casp inhibitors, VAD, DEVD and LEHD. Increased phosphorylated c-Jun N-terminal kinase (JNK) protein expression was detected in AMN- but not SORA- or STI-treated SKOV-3 cells, and the JNK inhibitors, SP600125 and JNKI, showed slight but significant enhancement of AMN-induced cell death in SKOV-3 cells. The intracellular peroxide level was elevated by AMN and H2O2, and N-acetylcysteine (NAC) prevented H2O2- but not AMN-induced peroxide production and apoptosis in SKOV-3 cells. AMN induction of apoptosis with increased intracellular peroxide production and JNK protein phosphorylation was also identified in human A2780 ovarian cancer cells, cisplatin-resistant A2780CP cells, and clear ES-2 cells. The evidence supporting AMN effectively reducing the viability of human ovarian cancer cells via mitochondrion-dependent apoptosis is provided.
Collapse
Affiliation(s)
- Tze-Chien Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Ming-Chih Yu
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chiang Chien
- Department of Nephrology, Chi-Mei Medical Center, Tainan, Taiwan; Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Ming-Shun Wu
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Yu-Chieh Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yen-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Cancer Research Center and Orthopedics Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
36
|
Shen SM, Guo M, Xiong Z, Yu Y, Zhao XY, Zhang FF, Chen GQ. AIF inhibits tumor metastasis by protecting PTEN from oxidation. EMBO Rep 2015; 16:1563-80. [PMID: 26415504 PMCID: PMC4641507 DOI: 10.15252/embr.201540536] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/26/2015] [Indexed: 12/27/2022] Open
Abstract
Apoptosis-inducing factor (AIF) exerts dual roles on cell death and survival, but its substrates as a putative oxidoreductase and roles in tumorigenesis remain elusive. Here, we report that AIF physically interacts with and inhibits the oxidation of phosphatase and tensin homolog on chromosome ten (PTEN), a tumor suppressor susceptible for oxidation-mediated inactivation. More intriguingly, we also identify PTEN as a mitochondrial protein and the ectopic expression of mitochondrial targeting sequence-carrying PTEN almost completely inhibits Akt phosphorylation in PTEN-deficient cells. AIF knockdown causes oxidation-mediated inactivation of the lipid phosphatase activity of PTEN, with ensuing activation of Akt kinase, phosphorylation of the Akt substrate GSK-3β, and activation of β-catenin signaling in cancer cells. Through its effect on β-catenin signaling, AIF inhibits epithelial–mesenchymal transition (EMT) and metastasis of cancer cells in vitro and in orthotopically implanted xenografts. Accordingly, the expression of AIF is correlated with the survival of human patients with cancers of multiple origins. These results identify PTEN as the substrate of AIF oxidoreductase and reveal a novel function for AIF in controlling tumor metastasis.
Collapse
Affiliation(s)
- Shao-Ming Shen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Meng Guo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Zhong Xiong
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Yun Yu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xu-Yun Zhao
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fei-Fei Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences-SJTU-SM, Shanghai, China
| |
Collapse
|
37
|
Cobanoglu B, Ceyran AB, Simsek M, Şenol S. Immunohistochemical analysis of Bax and AIF in colorectal tumors. Int J Clin Exp Med 2015; 8:16071-16076. [PMID: 26629114 PMCID: PMC4659003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 08/03/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND/AIMS This study had three aims. The first was to determine whether Bax (Bcl-2-associated X protein) and AIF (apoptosis-inducing factor) are expressed in tissue sections of colorectal tumors. The second was to ascertain whether there is any difference in Bax and AIF expression between colorectal polyps, adenomas, and carcinomas. The third aim was to determine whether there is any difference between Bax and AIF expression in colorectal tumors. MATERIALS AND METHODS Bax and AIF expression were determined in 20 hyperplastic polyps (HPs), 20 adenomatous polyps (APs), 20 samples of colorectal carcinomas, and 20 samples of normal mucosa by immunohistochemical methods. RESULTS The staining level of Bax and AIF in adenomas and carcinomas was significantly higher than in normal tissues (P<0.01). There was also a significant difference between HPs and APs (P<0.01). The level of Bax and AIF in carcinomas was higher than in adenomas, and the difference was of statistical significance (P<0.01). CONCLUSION This study may be of interest in future research to confirm whether the changed expression of Bax and/or AIF between benign and malignant tumors can provide valuable information for the evaluation of colon or other tumors.
Collapse
Affiliation(s)
- Bengu Cobanoglu
- Department of Pathology, Istanbul-Turkıye, Medeniyet University, Goztepe Education and Research HospitalTurkıye
| | - A Bahar Ceyran
- Department of Pathology, Istanbul-Turkıye, Medeniyet University, Goztepe Education and Research HospitalTurkıye
| | - Mustafa Simsek
- Department of Anesthesiology and Reanimation, Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research HospitalIstanbul, Turkıye
| | - Serkan Şenol
- Department of Pathology, Istanbul-Turkıye, Medeniyet University, Goztepe Education and Research HospitalTurkıye
| |
Collapse
|
38
|
Elguindy MM, Nakamaru-Ogiso E. Apoptosis-inducing Factor (AIF) and Its Family Member Protein, AMID, Are Rotenone-sensitive NADH:Ubiquinone Oxidoreductases (NDH-2). J Biol Chem 2015; 290:20815-20826. [PMID: 26063804 PMCID: PMC4543644 DOI: 10.1074/jbc.m115.641498] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/26/2015] [Indexed: 11/06/2022] Open
Abstract
Apoptosis-inducing factor (AIF) and AMID (AIF-homologous mitochondrion-associated inducer of death) are flavoproteins. Although AIF was originally discovered as a caspase-independent cell death effector, bioenergetic roles of AIF, particularly relating to complex I functions, have since emerged. However, the role of AIF in mitochondrial respiration and redox metabolism has remained unknown. Here, we investigated the redox properties of human AIF and AMID by comparing them with yeast Ndi1, a type 2 NADH:ubiquinone oxidoreductase (NDH-2) regarded as alternative complex I. Isolated AIF and AMID containing naturally incorporated FAD displayed no NADH oxidase activities. However, after reconstituting isolated AIF or AMID into bacterial or mitochondrial membranes, N-terminally tagged AIF and AMID displayed substantial NADH:O₂ activities and supported NADH-linked proton pumping activities in the host membranes almost as efficiently as Ndi1. NADH:ubiquinone-1 activities in the reconstituted membranes were highly sensitive to 2-n-heptyl-4-hydroxyquinoline-N-oxide (IC₅₀ = ∼1 μm), a quinone-binding inhibitor. Overexpressing N-terminally tagged AIF and AMID enhanced the growth of a double knock-out Escherichia coli strain lacking complex I and NDH-2. In contrast, C-terminally tagged AIF and NADH-binding site mutants of N-terminally tagged AIF and AMID failed to show both NADH:O₂ activity and the growth-enhancing effect. The disease mutant AIFΔR201 showed decreased NADH:O₂ activity and growth-enhancing effect. Furthermore, we surprisingly found that the redox activities of N-terminally tagged AIF and AMID were sensitive to rotenone, a well known complex I inhibitor. We propose that AIF and AMID are previously unidentified mammalian NDH-2 enzymes, whose bioenergetic function could be supplemental NADH oxidation in cells.
Collapse
Affiliation(s)
- Mahmoud M Elguindy
- Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Eiko Nakamaru-Ogiso
- Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| |
Collapse
|
39
|
Villanueva R, Ferreira P, Marcuello C, Usón A, Miramar MD, Peleato ML, Lostao A, Susin SA, Medina M. Key Residues Regulating the Reductase Activity of the Human Mitochondrial Apoptosis Inducing Factor. Biochemistry 2015; 54:5175-84. [PMID: 26237213 DOI: 10.1021/acs.biochem.5b00696] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The human Apoptosis Inducing Factor (hAIF) is a bifunctional NAD(P)H-dependent flavoreductase involved in both mitochondrial energy metabolism and caspase-independent cell death. Even though several studies indicate that both functions are redox controlled by NADH binding, the exact role of hAIF as a reductase in healthy mitochondria remains unknown. Upon reduction by NADH, hAIF dimerizes and produces very stable flavin/nicotinamide charge transfer complexes (CTC), by stacking of the oxidized nicotinamide moiety of the NAD(+) coenzyme against the re-face of the reduced flavin ring of its FAD cofactor. Such complexes are critical to restrict the hAIF efficiency as a reductase. The molecular basis of the hAIF reductase activity is here investigated by analyzing the role played by residues contributing to the interaction of the FAD isoalloxazine ring and of the nicotinamide moiety of NADH at the active site. Mutations at K177 and E314 produced drastic effects on the hAIF ability to retain the FAD cofactor, indicating that these residues are important to set up the holo-enzyme active site conformation. Characterization of P173G hAIF indicates that the stacking of P173 against the isoalloxazine ring is relevant to determine the flavin environment and to modulate the enzyme affinity for NADH. Finally, the properties of the F310G and H454S hAIF mutants indicate that these two positions contribute to form a compact active site essential for NADH binding, CTC stabilization, and NAD(+) affinity for the reduced state of hAIF. These features are key determinants of the particular behavior of hAIF as a NADH-dependent oxidoreductase.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Santos A Susin
- INSERM U1138, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche des Cordeliers , F-75006, Paris, France.,Université Pierre et Marie Curie-Sorbonne Universités , F-75006, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité , F-75006, Paris, France
| | | |
Collapse
|
40
|
Syed DN, Lall RK, Chamcheu JC, Haidar O, Mukhtar H. Involvement of ER stress and activation of apoptotic pathways in fisetin induced cytotoxicity in human melanoma. Arch Biochem Biophys 2014; 563:108-117. [PMID: 25016296 DOI: 10.1016/j.abb.2014.06.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/04/2014] [Accepted: 06/15/2014] [Indexed: 02/07/2023]
Abstract
The prognosis of malignant melanoma remains poor in spite of recent advances in therapeutic strategies for the deadly disease. Fisetin, a dietary flavonoid is currently being investigated for its growth inhibitory properties in various cancer models. We previously showed that fisetin inhibited melanoma growth in vitro and in vivo. Here, we evaluated the molecular basis of fisetin induced cytotoxicity in metastatic human melanoma cells. Fisetin treatment induced endoplasmic reticulum (ER) stress in highly aggressive A375 and 451Lu human melanoma cells, as revealed by up-regulation of ER stress markers including IRE1α, XBP1s, ATF4 and GRP78. Time course analysis indicated that the ER stress was associated with activation of the extrinsic and intrinsic apoptotic pathways. Fisetin treated 2-D melanoma cultures displayed autophagic response concomitant with induction of apoptosis. Prolonged treatment (16days) with fisetin in a 3-D reconstituted melanoma model resulted in inhibition of melanoma progression with significant apoptosis, as evidenced by increased staining of cleaved Caspase-3 in the treated constructs. However, no difference in the expression of autophagic marker LC-3 was noted between treated and control groups. Fisetin treatment to 2-D melanoma cultures resulted in phosphorylation and activation of the multifunctional AMP-activated protein kinase (AMPK) involved in the regulation of diverse cellular processes, including autophagy and apoptosis. Silencing of AMPK failed to prevent cell death indicating that fisetin induced cytotoxicity is mediated through both AMPK-dependent and -independent mechanisms. Taken together, our studies confirm apoptosis as the primary mechanism through which fisetin inhibits melanoma cell growth and that activation of both extrinsic and intrinsic pathways contributes to fisetin induced cytotoxicity.
Collapse
Affiliation(s)
- Deeba N Syed
- Department of Dermatology, University of Wisconsin, Madison
| | - Rahul K Lall
- Department of Dermatology, University of Wisconsin, Madison
| | | | - Omar Haidar
- Department of Dermatology, University of Wisconsin, Madison
| | - Hasan Mukhtar
- Department of Dermatology, University of Wisconsin, Madison
| |
Collapse
|
41
|
Xu S, Wu H, Nie H, Yue L, Jiang H, Xiao S, Li Y. AIF downregulation and its interaction with STK3 in renal cell carcinoma. PLoS One 2014; 9:e100824. [PMID: 24992339 PMCID: PMC4081115 DOI: 10.1371/journal.pone.0100824] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/30/2014] [Indexed: 01/06/2023] Open
Abstract
Apoptosis-inducing factor (AIF) plays a crucial role in caspase-independent programmed cell death by triggering chromatin condensation and DNA fragmentation. Therefore, it might be involved in cell homeostasis and tumor development. In this study, we report significant AIF downregulation in the majority of renal cell carcinomas (RCC). In a group of RCC specimens, 84% (43 out of 51) had AIF downregulation by immunohistochemistry stain. Additional 10 kidney tumors, including an oxyphilic adenoma, also had significant AIF downregulation by Northern blot analysis. The mechanisms of the AIF downregulation included both AIF deletion and its promoter methylation. Forced expression of AIF in RCC cell lines induced massive apoptosis. Further analysis revealed that AIF interacted with STK3, a known regulator of apoptosis, and enhanced its phosphorylation at Thr180. These results suggest that AIF downregulation is a common event in kidney tumor development. AIF loss may lead to decreased STK3 activity, defective apoptosis and malignant transformation.
Collapse
Affiliation(s)
- Shengqiang Xu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hongjin Wu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Huan Nie
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Lei Yue
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Huadong Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (SZ) (SX); (YL) (YL)
| | - Yu Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
- * E-mail: (SZ) (SX); (YL) (YL)
| |
Collapse
|
42
|
Yu W, Bonnet M, Farso M, Ma K, Chabot JG, Martin E, Torriglia A, Guan Z, McLaurin J, Quirion R, Krantic S. The expression of apoptosis inducing factor (AIF) is associated with aging-related cell death in the cortex but not in the hippocampus in the TgCRND8 mouse model of Alzheimer's disease. BMC Neurosci 2014; 15:73. [PMID: 24915960 PMCID: PMC4070095 DOI: 10.1186/1471-2202-15-73] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/30/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent evidence has suggested that Alzheimer's disease (AD)-associated neuronal loss may occur via the caspase-independent route of programmed cell death (PCD) in addition to caspase-dependent mechanisms. However, the brain region specificity of caspase-independent PCD in AD-associated neurodegeneration is unknown. We therefore used the transgenic CRND8 (TgCRND8) AD mouse model to explore whether the apoptosis inducing factor (AIF), a key mediator of caspase-independent PCD, contributes to cell loss in selected brain regions in the course of aging. RESULTS Increased expression of truncated AIF (tAIF), which is directly responsible for cell death induction, was observed at both 4- and 6-months of age in the cortex. Concomitant with the up-regulation of tAIF was an increase in the nuclear translocation of this protein. Heightened tAIF expression or translocation was not observed in the hippocampus or cerebellum, which were used as AD-vulnerable and relatively AD-spared regions, respectively. The cortical alterations in tAIF levels were accompanied by increased Bax expression and mitochondrial translocation. This effect was preceded by a significant reduction in ATP content and an increase in reactive oxygen species (ROS) production, detectable at 2 months of age despite negligible amounts of amyloid-beta peptides (Aβ). CONCLUSIONS Taken together, these data suggest that AIF is likely to play a region-specific role in AD-related caspase-independent PCD, which is consistent with aging-associated mitochondrial impairment and oxidative stress.
Collapse
Affiliation(s)
- Wenfeng Yu
- Key laboratory of Molecular Biology, Guiyang Medical University, Guiyang 550004, China
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Mathilde Bonnet
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Mark Farso
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Keran Ma
- Department Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jean-Guy Chabot
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | | | | | - Zhizhong Guan
- Key laboratory of Molecular Biology, Guiyang Medical University, Guiyang 550004, China
- Department of Pathology in the Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China
| | - JoAnne McLaurin
- Department Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Rémi Quirion
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Slavica Krantic
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
- Centre de Recherche des Cordeliers, UMRS872, Paris, France
| |
Collapse
|
43
|
Significant evidence of linkage for a gene predisposing to colorectal cancer and multiple primary cancers on 22q11. Clin Transl Gastroenterol 2014; 5:e50. [PMID: 24572700 PMCID: PMC3940837 DOI: 10.1038/ctg.2014.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 11/15/2013] [Accepted: 12/23/2013] [Indexed: 01/28/2023] Open
Abstract
Objectives: The genetic basis of colorectal cancer (CRC) is not completely specified. Part of the difficulty in mapping predisposition genes for CRC may be because of phenotypic heterogeneity. Using data from a population genealogy of Utah record linked to a statewide cancer registry, we identified a subset of CRC cases that exhibited familial clustering in excess of that expected for all CRC cases in general, which may represent a genetically homogeneous subset of CRC. Methods: Using a new familial aggregation method referred to as the subset genealogic index of familiality (subsetGIF), combined with detailed information from a statewide tumor registry, we identified a subset of CRC cases that exhibited excess familial clustering above that expected for CRC: CRC cases who had at least one other primary tumor at a different site. A genome-wide linkage analysis was performed on a set of high-risk CRC pedigrees that included multiple CRC cases with additional primaries to identify evidence for predisposition loci. Results: A total of 13 high-risk CRC pedigrees with multiple CRC cases with other primary cancers were identified. Linkage analysis identified one pedigree with a significant linkage signal at 22q11 (LOD (logarithm (base 10) of odds)=3.39). Conclusions: A predisposition gene or variant for CRC that also predisposes to other primary cancers likely resides on chromosome 22q11. The ability to use statewide population genealogy and tumor registry data was critical to identify an informative subset of CRC cases that is possibly more genetically homogeneous than CRC in general, and may have improved statistical power for predisposition locus identification in this study.
Collapse
|
44
|
Prabhu SB, Khalsa JK, Banerjee H, Das A, Srivastava S, Mattoo HR, Thyagarajan K, Tanwar S, Das DS, Majumdar SS, George A, Bal V, Durdik JM, Rath S. Role of apoptosis-inducing factor (Aif) in the T cell lineage. Indian J Med Res 2013; 138:577-90. [PMID: 24434313 PMCID: PMC3928691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Multiple checkpoints regulating finely balanced death-versus-survival decisions characterize both thymic development and peripheral homeostasis of T lymphocytes. While exploring the mechanisms of T cell death involved at various stages during the life of a T cell, we have observed and reported a variety of non-redundant roles for apoptosis inducing factor (Aif), a mitochondrial flavoprotein. Aif is ubiquitously expressed in all cell lineages and functions as an NADH oxidase in its mitochondrial location. It is released following the mitochondrial death signals, whereupon it translocates to the nucleus, binds to DNA and causes large-scale DNA fragmentation. During T cell development, Aif is important for developing thymocytes to navigate the double negative (DN)3 to DN4 transition (beta-selection), via its oxidoreductase property which protects the rapidly proliferating cells from death due to reactive oxygen species (ROS). In peripheral mature T cells, Aif deficiency leads to an increased susceptibility of T cell blasts to activation induced cell death (AICD), possibly mediated by its antioxidant function, and decreased sensitivity to neglect-induced death (NID). Thus, Aif seems to have pro-apoptotic and anti-apoptotic roles in the same lineage in different contexts and at different stages. Surprisingly, in the closely related B lymphocyte lineage, Aif deficiency does not result in any abnormality. These findings generate the possibility of specific T cell dysfunction in human disease caused by Aif deficiency, as well as in mitochondriopathies due to other causes. Also, these data raise questions regarding the basis of lineage-specific consequences of the dysfunction/deficiency of apparently ubiquitous molecules.
Collapse
Affiliation(s)
- Savit B. Prabhu
- National Institute of Immunology, New Delhi, India,Reprint requests: Dr Savit B. Prabhu / Dr Satyajit Rath, National Institute of Immunology, Aruna Asaf Ali Road, New Delhi 110 067, India e-mail: /
| | | | - Hridesh Banerjee
- National Institute of Immunology, New Delhi, India,Laboratory of Molecular Biology & Immunology, National Institute of Aging, 251 Bayview Boulevard, Baltimore MD 21224, USA
| | - Abhishek Das
- National Institute of Immunology, New Delhi, India,Immune Disease Institute, Children's Hospital Boston, 3 Blackfan Circle, Boston MA 02115, USA
| | - Smita Srivastava
- National Institute of Immunology, New Delhi, India,Division of Infectious Diseases, Department of Medicine, New York University School of Medicine, Smilow 901, 550 First Avenue, New York NY 10016, USA
| | - Hamid R. Mattoo
- National Institute of Immunology, New Delhi, India,MGH Cancer Center, Building 149, 13th Street, Charlestown, MA 02129, USA
| | - Krishnamurthy Thyagarajan
- National Institute of Immunology, New Delhi, India,Department of Surgery, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston SC 29425, USA
| | | | | | | | - Anna George
- National Institute of Immunology, New Delhi, India
| | - Vineeta Bal
- National Institute of Immunology, New Delhi, India
| | - Jeannine M. Durdik
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Satyajit Rath
- National Institute of Immunology, New Delhi, India,Reprint requests: Dr Savit B. Prabhu / Dr Satyajit Rath, National Institute of Immunology, Aruna Asaf Ali Road, New Delhi 110 067, India e-mail: /
| |
Collapse
|
45
|
Wu MS, Lien GS, Shen SC, Yang LY, Chen YC. N
-acetyl-L
-cysteine enhances fisetin-induced cytotoxicity via induction of ROS-independent apoptosis in human colonic cancer cells. Mol Carcinog 2013; 53 Suppl 1:E119-29. [DOI: 10.1002/mc.22053] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Revised: 04/18/2013] [Accepted: 05/03/2013] [Indexed: 01/11/2023]
Affiliation(s)
- Ming-Shun Wu
- Graduate Institute of Clinical Medicine; College of Medicine, Taipei Medical University; Taipei Taiwan
- Division of Gastroenterology; Department of Internal Medicine; Wan Fang Hospital; Taipei Medical University; Taipei Taiwan
| | - Gi-Shih Lien
- Graduate Institute of Clinical Medicine; College of Medicine, Taipei Medical University; Taipei Taiwan
- Division of Gastroenterology; Department of Internal Medicine; Wan Fang Hospital; Taipei Medical University; Taipei Taiwan
| | - Shing-Chuan Shen
- Graduate Institute of Medical Sciences; Taipei Medical University; Taipei Taiwan
| | - Liang-Yo Yang
- Department of Physiology and Graduate Institute of Neuroscience; Taipei Medical University; Taipei Taiwan
| | - Yen-Chou Chen
- Graduate Institute of Medical Sciences; Taipei Medical University; Taipei Taiwan
- Cancer Research Center and Orthopedics Research Center; Taipei Medical University Hospital; Taipei Taiwan
| |
Collapse
|
46
|
Feng X, Koh DW. Roles of poly(ADP-ribose) glycohydrolase in DNA damage and apoptosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 304:227-81. [PMID: 23809438 DOI: 10.1016/b978-0-12-407696-9.00005-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Poly(ADP-ribose) glycohydrolase (PARG) is the primary enzyme that catalyzes the hydrolysis of poly(ADP-ribose) (PAR), an essential biopolymer that is synthesized by poly(ADP-ribose) polymerases (PARPs) in the cell. By regulating the hydrolytic arm of poly(ADP-ribosyl)ation, PARG participates in a number of biological processes, including the repair of DNA damage, chromatin dynamics, transcriptional regulation, and cell death. Collectively, the research investigating the roles of PARG in the cell has identified the importance of PARG and its value as a therapeutic target. However, the biological role of PARG remains less understood than the role of PAR synthesis by the PARPs. Further complicating the study of PARG is the existence of multiple PARG isoforms in the cell, the lack of optimal PARG inhibitors, and the lack of viable PARG-null animals. This review will present our current knowledge of PARG, with a focus on its roles in DNA-damage repair and cell death.
Collapse
Affiliation(s)
- Xiaoxing Feng
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Pullman, Washington, USA
| | | |
Collapse
|
47
|
Polster BM. AIF, reactive oxygen species, and neurodegeneration: a "complex" problem. Neurochem Int 2012; 62:695-702. [PMID: 23246553 DOI: 10.1016/j.neuint.2012.12.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 11/26/2012] [Accepted: 12/05/2012] [Indexed: 12/21/2022]
Abstract
Apoptosis-inducing factor (AIF) is a flavin-binding mitochondrial intermembrane space protein that is implicated in diverse but intertwined processes that include maintenance of electron transport chain function, reactive oxygen species regulation, cell death, and neurodegeneration. In acute brain injury, AIF acquires a pro-death role upon translocation from the mitochondria to the nucleus, where it initiates chromatin condensation and large-scale DNA fragmentation. Although harlequin mice exhibiting an 80-90% global reduction in AIF protein are resistant to numerous forms of acute brain injury, they paradoxically undergo slow, progressive neurodegeneration beginning at three months of age. Brain deterioration, accompanied by markers of oxidative stress, is most pronounced in the cerebellum and retina, although it also occurs in the cortex, striatum, and thalamus. Loss of an AIF pro-survival function linked to assembly or stabilization of electron transport chain complex I underlies chronic neurodegeneration. To date, most studies of neurodegeneration have failed to adequately separate the relative importance of the mitochondrial and nuclear functions of AIF in determining the extent of injury, or whether oxidative stress plays a causative role. This review explores the complicated relationship among AIF, complex I, and the regulation of mitochondrial reactive oxygen species levels. It also discusses the controversial role of complex I deficiency in Parkinson's disease, and what can be learned from the AIF- and complex I-depleted harlequin mouse.
Collapse
Affiliation(s)
- Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore St., MSTF 5-34, Baltimore, MD 21201, USA.
| |
Collapse
|
48
|
Lewis EM, Wilkinson AS, Jackson JS, Mehra R, Varambally S, Chinnaiyan AM, Wilkinson JC. The enzymatic activity of apoptosis-inducing factor supports energy metabolism benefiting the growth and invasiveness of advanced prostate cancer cells. J Biol Chem 2012; 287:43862-75. [PMID: 23118229 DOI: 10.1074/jbc.m112.407650] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Apoptosis-inducing factor (AIF) promotes cell death yet also controls mitochondrial homeostasis and energy metabolism. It is unclear how these activities are coordinated, and the impact of AIF upon human disease, in particular cancer, is not well documented. In this study we have explored the contribution of AIF to the progression of prostate cancer. Analysis of archival gene expression data demonstrated that AIF transcript levels are elevated in human prostate cancer, and we found that AIF protein is increased in prostate tumors. Suppression of AIF expression in the prostate cancer cell lines LNCaP, DU145, and PC3 demonstrated that AIF does not contribute to cell toxicity via a variety of chemical death triggers, and growth under nutrient-rich conditions is largely unaffected by AIF ablation. However, under growth stress conditions, AIF depletion from DU145 and PC3 cell lines led to significant reductions in cell survival and growth that were not observed in LNCaP cells. Moreover AIF-deficient PC3 cells exhibited substantial reduction of tumorigenic growth in vivo. This reduced survival correlated with decreased expression of mitochondrial complex I protein subunits and concomitant changes in glucose metabolism. Finally, restoration of AIF-deficient PC3 cells with AIF variants demonstrated that the enzymatic activity of AIF is required for aggressive growth. Overall these studies show that AIF is an important factor for advanced prostate cancer cells and that through control of energy metabolism and redox balance, the enzymatic activity of AIF is critical for this support.
Collapse
Affiliation(s)
- Eric M Lewis
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Banerjee H, Das A, Srivastava S, Mattoo HR, Thyagarajan K, Khalsa JK, Tanwar S, Das DS, Majumdar SS, George A, Bal V, Durdik JM, Rath S. A role for apoptosis-inducing factor in T cell development. J Exp Med 2012; 209:1641-53. [PMID: 22869892 PMCID: PMC3428951 DOI: 10.1084/jem.20110306] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 07/20/2012] [Indexed: 12/14/2022] Open
Abstract
Apoptosis-inducing factor (Aif) is a mitochondrial flavoprotein that regulates cell metabolism and survival in many tissues. We report that aif-hypomorphic harlequin (Hq) mice show thymic hypocellularity and a cell-autonomous thymocyte developmental block associated with apoptosis at the β-selection stage, independent of T cell receptor β recombination. No abnormalities are observed in the B cell lineage. Transgenes encoding wild-type or DNA-binding-deficient mutant Aif rectify the thymic defect, but a transgene encoding oxidoreductase activity-deficient mutant Aif does not. The Hq thymic block is reversed in vivo by antioxidant treatment, and Hq T but not B lineage cells show enhanced oxidative stress. Thus, Aif, a ubiquitous protein, serves a lineage-specific nonredundant antiapoptotic role in the T cell lineage by regulating reactive oxygen species during thymic β-selection.
Collapse
Affiliation(s)
| | - Abhishek Das
- National Institute of Immunology, New Delhi 110067, India
| | | | | | | | | | - Shalini Tanwar
- National Institute of Immunology, New Delhi 110067, India
| | | | | | - Anna George
- National Institute of Immunology, New Delhi 110067, India
| | - Vineeta Bal
- National Institute of Immunology, New Delhi 110067, India
| | - Jeannine M. Durdik
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701
| | - Satyajit Rath
- National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
50
|
Wang D, Liang J, Zhang Y, Gui B, Wang F, Yi X, Sun L, Yao Z, Shang Y. Steroid receptor coactivator-interacting protein (SIP) inhibits caspase-independent apoptosis by preventing apoptosis-inducing factor (AIF) from being released from mitochondria. J Biol Chem 2012; 287:12612-21. [PMID: 22371500 DOI: 10.1074/jbc.m111.334151] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Apoptosis-inducing factor (AIF) is a caspase-independent death effector. Normally residing in the mitochondrial intermembrane space, AIF is released and translocated to the nucleus in response to proapoptotic stimuli. Nuclear AIF binds to DNA and induces chromatin condensation and DNA fragmentation, characteristics of apoptosis. Until now, it remained to be clarified how the mitochondrial-nuclear translocation of AIF is regulated. Here we report that steroid receptor coactivator-interacting protein (SIP) interacts directly with AIF in mitochondria and specifically inhibits caspase-independent and AIF-dependent apoptosis. Challenging cells with apoptotic stimuli leads to rapid degradation of SIP, and subsequently AIF is liberated from mitochondria and translocated to the nucleus to induce apoptosis. Together, our data demonstrate that SIP is a novel regulator in caspase-independent and AIF-mediated apoptosis.
Collapse
Affiliation(s)
- Dandan Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100191, China
| | | | | | | | | | | | | | | | | |
Collapse
|