1
|
Kang S, Antoniewicz MR, Hay N. Metabolic and transcriptomic reprogramming during contact inhibition-induced quiescence is mediated by YAP-dependent and YAP-independent mechanisms. Nat Commun 2024; 15:6777. [PMID: 39117624 PMCID: PMC11310444 DOI: 10.1038/s41467-024-51117-y] [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: 10/06/2023] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Metabolic rewiring during the proliferation-to-quiescence transition is poorly understood. Here, using a model of contact inhibition-induced quiescence, we conducted 13C-metabolic flux analysis in proliferating (P) and quiescent (Q) mouse embryonic fibroblasts (MEFs) to investigate this process. Q cells exhibit reduced glycolysis but increased TCA cycle flux and mitochondrial respiration. Reduced glycolytic flux in Q cells correlates with reduced glycolytic enzyme expression mediated by yes-associated protein (YAP) inhibition. The increased TCA cycle activity and respiration in Q cells is mediated by induced mitochondrial pyruvate carrier (MPC) expression, rendering them vulnerable to MPC inhibition. The malate-to-pyruvate flux, which generates NADPH, is markedly reduced by modulating malic enzyme 1 (ME1) dimerization in Q cells. Conversely, the malate dehydrogenase 1 (MDH1)-mediated oxaloacetate-to-malate flux is reversed and elevated in Q cells, driven by high mitochondrial-derived malate levels, reduced cytosolic oxaloacetate, elevated MDH1 levels, and a high cytoplasmic NAD+/NADH ratio. Transcriptomic analysis revealed large number of genes are induced in Q cells, many of which are associated with the extracellular matrix (ECM), while YAP-dependent and cell cycle-related genes are repressed. The results suggest that high TCA cycle flux and respiration in Q cells are required to generate ATP and amino acids to maintain de-novo ECM protein synthesis and secretion.
Collapse
Affiliation(s)
- Soeun Kang
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
2
|
Khan H, Verma Y, Rana SVS. Combined Effects of Fluoride and Arsenic on Mitochondrial Function in the Liver of Rat. Appl Biochem Biotechnol 2023; 195:6856-6866. [PMID: 36947368 DOI: 10.1007/s12010-023-04401-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 03/23/2023]
Abstract
Biochemical and/or molecular mechanisms of arsenic or fluoride toxicity in experimental animals have been widely investigated in the recent past. However, their combined effects on target cells/organelle are poorly understood. The present study was executed to delineate their combined effects on mitochondrial function in the liver of rat. Female Wistar rats (140 ± 20 g) were force fed individually or in combination with sodium arsenate (4 mg/kg body weight) and sodium fluoride (4 mg/kg body weight) for 90 days. Thereafter, established markers of mitochondrial function viz. mitochondrial lipid peroxidation, oxidative phosphorylation, ATPase, succinic dehydrogenase, and caspase-3 activity were determined. Cytochrome C release and oxidative DNA damage were also estimated in the liver of respective groups of rats. The study showed significant differences in these results amongst the three groups. Observations on parameters viz. LPO, cytochrome-C, caspase-3, and 8-OHdG suggested an antagonistic relationship between these two elements. Results on ATPase, SDH, and ADP:O ratio indicated synergism. It is concluded that AsIII + F in combination may express differential effects on signalling pathways and proapoptotic/antiapoptotic proteins/genes that contribute to liver cell death. Interaction of As and F with mitochondria.
Collapse
Affiliation(s)
- Huma Khan
- Department of Toxicology, Ch. Charan Singh University, Meerut, 250 004, India
| | - Yeshvandra Verma
- Department of Toxicology, Ch. Charan Singh University, Meerut, 250 004, India
| | - S V S Rana
- Department of Toxicology, Ch. Charan Singh University, Meerut, 250 004, India.
| |
Collapse
|
3
|
Jia Y, Yu X, Liu R, Shi L, Jin H, Yang D, Zhang X, Shen Y, Feng Y, Zhang P, Yang Y, Zhang L, Zhang P, Li Z, He A, Kong G. PRMT1 methylation of WTAP promotes multiple myeloma tumorigenesis by activating oxidative phosphorylation via m6A modification of NDUFS6. Cell Death Dis 2023; 14:512. [PMID: 37558663 PMCID: PMC10412649 DOI: 10.1038/s41419-023-06036-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
Epigenetic modifications play important roles during the pathogenesis of multiple myeloma (MM). Herein, we found that protein arginine methyltransferase 1 (PRMT1) was highly expressed in MM patients, which was positively correlated with MM stages. High PRMT1 expression was correlated with adverse prognosis in MM patients. We further showed that silencing PRMT1 inhibited MM proliferation and tumorigenesis in vitro and in vivo. Mechanistically, we revealed that the knockdown of PRMT1 reduced the oxidative phosphorylation (OXPHOS) of MM cells through NDUFS6 downregulation. Meanwhile, we identified that WTAP, a key component of the m6A methyltransferase complex, was methylated by PRMT1, and NDUFS6 was identified as a bona fide m6A target of WTAP. Finally, we found that the combination of PRMT1 inhibitor and bortezomib synergistically inhibited MM progression. Collectively, our results demonstrate that PRMT1 plays a crucial role during MM tumorigenesis and suggeste that PRMT1 could be a potential therapeutic target in MM.
Collapse
Affiliation(s)
- Yachun Jia
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Xiao Yu
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Luyi Shi
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Hua Jin
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Dan Yang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Xiaofeng Zhang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Ying Shen
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Yuandong Feng
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Peihua Zhang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Yi Yang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Linlin Zhang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Pengyu Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Zongfang Li
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
| | - Aili He
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
| | - Guangyao Kong
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
| |
Collapse
|
4
|
Li Z, Zou J, Chen X. In Response to Precision Medicine: Current Subcellular Targeting Strategies for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209529. [PMID: 36445169 DOI: 10.1002/adma.202209529] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/08/2022] [Indexed: 05/26/2023]
Abstract
Emerging as a potent anticancer treatment, subcellular targeted cancer therapy has drawn increasing attention, bringing great opportunities for clinical application. Here, two targeting strategies for four main subcellular organelles (mitochondria, lysosome, endoplasmic reticulum, and nucleus), including molecule- and nanomaterial (inorganic nanoparticles, micelles, organic polymers, and others)-based targeted delivery or therapeutic strategies, are summarized. Phototherapy, chemotherapy, radiotherapy, immunotherapy, and "all-in-one" combination therapy are among the strategies covered in detail. Such materials are constructed based on the specific properties and relevant mechanisms of organelles, enabling the elimination of tumors by inducing dysfunction in the corresponding organelles or destroying specific structures. The challenges faced by organelle-targeting cancer therapies are also summarized. Looking forward, a paradigm for organelle-targeting therapy with enhanced therapeutic efficacy compared to current clinical approaches is envisioned.
Collapse
Affiliation(s)
- Zheng Li
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
5
|
Petrelli F, Scandella V, Montessuit S, Zamboni N, Martinou JC, Knobloch M. Mitochondrial pyruvate metabolism regulates the activation of quiescent adult neural stem cells. SCIENCE ADVANCES 2023; 9:eadd5220. [PMID: 36857455 PMCID: PMC9977184 DOI: 10.1126/sciadv.add5220] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Cellular metabolism is important for adult neural stem/progenitor cell (NSPC) behavior. However, its role in the transition from quiescence to proliferation is not fully understood. We here show that the mitochondrial pyruvate carrier (MPC) plays a crucial and unexpected part in this process. MPC transports pyruvate into mitochondria, linking cytosolic glycolysis to mitochondrial tricarboxylic acid cycle and oxidative phosphorylation. Despite its metabolic key function, the role of MPC in NSPCs has not been addressed. We show that quiescent NSPCs have an active mitochondrial metabolism and express high levels of MPC. Pharmacological MPC inhibition increases aspartate and triggers NSPC activation. Furthermore, genetic Mpc1 ablation in vitro and in vivo also activates NSPCs, which differentiate into mature neurons, leading to overall increased hippocampal neurogenesis in adult and aged mice. These findings highlight the importance of metabolism for NSPC regulation and identify an important pathway through which mitochondrial pyruvate import controls NSPC quiescence and activation.
Collapse
Affiliation(s)
- Francesco Petrelli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Department of Cell Biology, University of Geneva, Geneva, Switzerland
| | - Valentina Scandella
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sylvie Montessuit
- Department of Cell Biology, University of Geneva, Geneva, Switzerland
| | - Nicola Zamboni
- Institute for Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | | | - Marlen Knobloch
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
6
|
Pourgholamali B, Sohrabi B, Salbi M, Akbari S, Rastan I, Sayaf M, Jalil AT, Kadhim MM, Sheervalilou R, Mehrzad N. Bioinformatic Analysis Divulged Novel Prognostic Circulating MicroRNAs and Their Potential Target Genes in Breast Cancer. Appl Biochem Biotechnol 2023; 195:283-297. [PMID: 36074234 DOI: 10.1007/s12010-022-04151-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2022] [Indexed: 01/13/2023]
Abstract
Breast cancer (BC) is both an inherited and environmental-based disease which is the leading cause of death among women. Early detection of BC can prevent invasion and metastasis in patients. Currently, researchers endeavor to find non-invasive biological markers from body fluids. Circulating non-coding RNAs such as microRNAs (miRNAs) can potentially be valuable prognostic and detective biomarkers. To identify novel miRNA-based biomarkers, we utilized bioinformatic tools. To reach this goal, the miRNA expression profiles of GSE31309, GSE 44,281, GSE98181, and GSE118782 were analyzed through a limma package of R. Target gene prediction of differentially expressed miRNAs, called differentially expressed miRNAs (DEMs), between samples of healthy individuals and BC patients was implemented through Multimir package of R. Functional enrichment analysis of predicted target genes through Enrich R (online database) revealed that most of the genes are enriched in the mitochondrial outer membrane for cellular component, intrinsic apoptotic signaling regulations for biological processes, transcription co-receptor activity for molecular functions, and dopaminergic synapse pathway. Furthermore, our survival analysis results revealed that miR-29c and mir-361 have the potential to serve as prognostic biomarkers.
Collapse
Affiliation(s)
- Babak Pourgholamali
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Behnoush Sohrabi
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Mandana Salbi
- Department of Microbiology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | | | - Iman Rastan
- Department of Electronic and Electrical Engineering, Shiraz Azad University, Shiraz, Iran
| | - Masoud Sayaf
- Azad University Central Tehran Branch Faculty of Basic Sciences, Department of Cellular and Molecular Biology, Tehran, Iran
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Mustafa M Kadhim
- Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, Iraq.,Department of Dentistry, Kut University College, Kut, Wasit, Iraq
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Nazanin Mehrzad
- Department of Biology, Science and Research Branch Islamic Azad university, Tehran, Iran.
| |
Collapse
|
7
|
Marotta C, Giorgi E, Binacchi F, Cirri D, Gabbiani C, Pratesi A. An overview of recent advancements in anticancer Pt(IV) prodrugs: New smart drug combinations, activation and delivery strategies. Inorganica Chim Acta 2023. [DOI: 10.1016/j.ica.2023.121388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
8
|
Witkowska M, Maciejewska N, Ryczkowska M, Olszewski M, Bagiński M, Makowiec S. From tryptophan to novel mitochondria-disruptive agent, synthesis and biological evaluation of 1,2,3,6-tetrasubstituted carbazoles. Eur J Med Chem 2022; 238:114453. [DOI: 10.1016/j.ejmech.2022.114453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/27/2022] [Accepted: 05/07/2022] [Indexed: 11/30/2022]
|
9
|
Gayan S, Joshi G, Dey T. Biomarkers of mitochondrial origin: a futuristic cancer diagnostic. Integr Biol (Camb) 2022; 14:77-88. [PMID: 35780307 DOI: 10.1093/intbio/zyac008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/17/2022] [Accepted: 05/27/2022] [Indexed: 11/12/2022]
Abstract
Cancer is a highly fatal disease without effective early-stage diagnosis and proper treatment. Along with the oncoproteins and oncometabolites, several organelles from cancerous cells are also emerging as potential biomarkers. Mitochondria isolated from cancer cells are one such biomarker candidates. Cancerous mitochondria exhibit different profiles compared with normal ones in morphology, genomic, transcriptomic, proteomic and metabolic landscape. Here, the possibilities of exploring such characteristics as potential biomarkers through single-cell omics and Artificial Intelligence (AI) are discussed. Furthermore, the prospects of exploiting the biomarker-based diagnosis and its futuristic utilization through circulatory tumor cell technology are analyzed. A successful alliance of circulatory tumor cell isolation protocols and a single-cell omics platform can emerge as a next-generation diagnosis and personalized treatment procedure.
Collapse
Affiliation(s)
- Sukanya Gayan
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Gargee Joshi
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Tuli Dey
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
10
|
Vulczak A, Alberici LC. Physical Exercise and Tumor Energy Metabolism. Cancer Treat Res Commun 2022; 32:100600. [PMID: 35811248 DOI: 10.1016/j.ctarc.2022.100600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/15/2022]
Abstract
Evidence supports the antitumoral effects of physical activity, either in experimental animal models or humans. However, the biological mechanisms by which physical exercise modulates tumoral development are still unclear. An important feature of the tumor cells is the altered energy metabolism, often associated with definitions of tumor aggressiveness. Nevertheless, exercise can cause global metabolic changes in the body, as well as modulate tumor metabolism. Here we specifically discuss the metabolic changes found in tumors and how exercise can contribute to anti-tumoral effects by modulating the mitochondrial function, and tricarboxylic acid cycle-related metabolites of cancer cells. The effect of physical exercise on tumor metabolism is a new possibility for comprehension of cancer biology and developing therapies focused on tumor energy metabolism.
Collapse
Affiliation(s)
- Anderson Vulczak
- Department of Biomolecular Sciences - School of Pharmaceutical Sciences of Ribeirao Preto - University of Sao Paulo, RibeirãoPreto, SP, Brazil
| | - Luciane Carla Alberici
- Department of Biomolecular Sciences - School of Pharmaceutical Sciences of Ribeirao Preto - University of Sao Paulo, RibeirãoPreto, SP, Brazil.
| |
Collapse
|
11
|
Hu M, Wang B, Zhang H, Wang H, Li H, Zhang X, Zhang J, Lu Q, Fang G, Wang J, Dong B. A Dual-Labeling Probe for Super-Resolution Imaging to Detect Mitochondrial Reactive Sulfur Species in Live Cells. Front Pharmacol 2022; 13:871059. [PMID: 35721202 PMCID: PMC9198575 DOI: 10.3389/fphar.2022.871059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Mitochondria are the main sites of reactive sulfur species (RSS) production in living cells. RSS in mitochondria play an important role in physiological and pathological processes of life. In this study, a dual-labeling probe that could simultaneously label the mitochondrial membrane and matrix was designed to quantitatively detect RSS of mitochondria in living cells using nano-level super-resolution imaging. Methods: A fluorescent probe CPE was designed and synthesized. The cytotoxicity of CPE was determined and co-localization of CPE with a commercial mitochondrial probe was analyzed in HeLa cells. Then, the uptake patterns of CPE in HeLa cells at different temperatures and endocytosis levels were investigated. The staining characteristics of CPE under different conditions were imaged and quantitated under structured illumination microscopy. Results: A fluorescence probe CPE reacting to RSS was developed, which could simultaneously label the mitochondrial membrane with green fluorescence and the mitochondrial matrix with red fluorescence. CPE was able to demonstrate the mitochondrial morphology and detect the changes of RSS in mitochondria. With the increase of mitochondrial RSS concentration, the light of the red matrix will be quenched. Conclusion: CPE provides a strategy for the design of probes and an attractive tool for accurate examination to changes of mitochondrial morphology and RSS in mitochondria in living cells at the nanoscale.
Collapse
Affiliation(s)
- Maomao Hu
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Boyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Hongdan Zhang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Han Wang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Huixin Li
- Department of Cardiology, Shandong Traditional Chinese Medicine University, Jinan, China
| | - Xinyu Zhang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Jinjin Zhang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Qianrun Lu
- Shandong Cancer Hospital and Institute, Jinan, China
| | - Guiqian Fang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Juan Wang
- Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China.,Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Cardiology, Shandong Traditional Chinese Medicine University, Jinan, China
| |
Collapse
|
12
|
Mendelsohn DH, Schnabel K, Mamilos A, Sossalla S, Pabel S, Duerr GD, Keller K, Schmitt VH, Barsch F, Walter N, Wong RMY, El Khassawna T, Niedermair T, Alt V, Rupp M, Brochhausen C. Structural Analysis of Mitochondrial Dynamics-From Cardiomyocytes to Osteoblasts: A Critical Review. Int J Mol Sci 2022; 23:4571. [PMID: 35562962 PMCID: PMC9101187 DOI: 10.3390/ijms23094571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/04/2022] Open
Abstract
Mitochondria play a crucial role in cell physiology and pathophysiology. In this context, mitochondrial dynamics and, subsequently, mitochondrial ultrastructure have increasingly become hot topics in modern research, with a focus on mitochondrial fission and fusion. Thus, the dynamics of mitochondria in several diseases have been intensively investigated, especially with a view to developing new promising treatment options. However, the majority of recent studies are performed in highly energy-dependent tissues, such as cardiac, hepatic, and neuronal tissues. In contrast, publications on mitochondrial dynamics from the orthopedic or trauma fields are quite rare, even if there are common cellular mechanisms in cardiovascular and bone tissue, especially regarding bone infection. The present report summarizes the spectrum of mitochondrial alterations in the cardiovascular system and compares it to the state of knowledge in the musculoskeletal system. The present paper summarizes recent knowledge regarding mitochondrial dynamics and gives a short, but not exhaustive, overview of its regulation via fission and fusion. Furthermore, the article highlights hypoxia and its accompanying increased mitochondrial fission as a possible link between cardiac ischemia and inflammatory diseases of the bone, such as osteomyelitis. This opens new innovative perspectives not only for the understanding of cellular pathomechanisms in osteomyelitis but also for potential new treatment options.
Collapse
Affiliation(s)
- Daniel H. Mendelsohn
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (D.H.M.); (K.S.); (A.M.); (T.N.)
- Central Biobank Regensburg, University Regensburg, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany; (N.W.); (V.A.); (M.R.)
| | - Katja Schnabel
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (D.H.M.); (K.S.); (A.M.); (T.N.)
- Central Biobank Regensburg, University Regensburg, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Andreas Mamilos
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (D.H.M.); (K.S.); (A.M.); (T.N.)
| | - Samuel Sossalla
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (S.S.); (S.P.)
| | - Steffen Pabel
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (S.S.); (S.P.)
| | - Georg Daniel Duerr
- Department of Cardiovascular Surgery, University Medical Center Mainz (Johannes Gutenberg-University Mainz), 55131 Mainz, Germany;
| | - Karsten Keller
- Department of Cardiology, Cardiology I, University Medical Center Mainz (Johannes Gutenberg-University Mainz), 55131 Mainz, Germany; (K.K.); (V.H.S.)
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz (Johannes Gutenberg-University Mainz), 55131 Mainz, Germany
- Department of Sports Medicine, Medical Clinic VII, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Volker H. Schmitt
- Department of Cardiology, Cardiology I, University Medical Center Mainz (Johannes Gutenberg-University Mainz), 55131 Mainz, Germany; (K.K.); (V.H.S.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, 55131 Mainz, Germany
| | - Friedrich Barsch
- Institute for Exercise and Occupational Medicine, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany;
| | - Nike Walter
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany; (N.W.); (V.A.); (M.R.)
| | - Ronald Man Yeung Wong
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China;
| | - Thaqif El Khassawna
- Department of Experimental Trauma Surgery, Justus-Liebig-University Giessen, 35390 Giessen, Germany;
| | - Tanja Niedermair
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (D.H.M.); (K.S.); (A.M.); (T.N.)
- Central Biobank Regensburg, University Regensburg, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Volker Alt
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany; (N.W.); (V.A.); (M.R.)
| | - Markus Rupp
- Department of Trauma Surgery, University Medical Centre Regensburg, 93053 Regensburg, Germany; (N.W.); (V.A.); (M.R.)
| | - Christoph Brochhausen
- Institute of Pathology, University Regensburg, 93053 Regensburg, Germany; (D.H.M.); (K.S.); (A.M.); (T.N.)
- Central Biobank Regensburg, University Regensburg, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
13
|
Wang J, Huang CLH, Zhang Y. Complement C1q Binding Protein (C1QBP): Physiological Functions, Mutation-Associated Mitochondrial Cardiomyopathy and Current Disease Models. Front Cardiovasc Med 2022; 9:843853. [PMID: 35310974 PMCID: PMC8924301 DOI: 10.3389/fcvm.2022.843853] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/25/2022] [Indexed: 12/03/2022] Open
Abstract
Complement C1q binding protein (C1QBP, p32) is primarily localized in mitochondrial matrix and associated with mitochondrial oxidative phosphorylative function. C1QBP deficiency presents as a mitochondrial disorder involving multiple organ systems. Recently, disease associated C1QBP mutations have been identified in patients with a combined oxidative phosphorylation deficiency taking an autosomal recessive inherited pattern. The clinical spectrum ranges from intrauterine growth restriction to childhood (cardio) myopathy and late-onset progressive external ophthalmoplegia. This review summarizes the physiological functions of C1QBP, its mutation-associated mitochondrial cardiomyopathy shown in the reported available patients and current experimental disease platforms modeling these conditions.
Collapse
Affiliation(s)
- Jie Wang
- National Regional Children's Medical Center (Northwest), Xi'an, China
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an, China
- Shaanxi Institute for Pediatric Diseases, Xi'an, China
- Xi'an Key Laboratory of Children's Health and Diseases, Xi'an, China
| | | | - Yanmin Zhang
- National Regional Children's Medical Center (Northwest), Xi'an, China
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an, China
- Shaanxi Institute for Pediatric Diseases, Xi'an, China
- Xi'an Key Laboratory of Children's Health and Diseases, Xi'an, China
- Department of Cardiology of Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China
- *Correspondence: Yanmin Zhang
| |
Collapse
|
14
|
Singh R, Jain A, Palanichamy JK, Nag TC, Bakhshi S, Singh A. Ultrastructural changes in cristae of lymphoblasts in acute lymphoblastic leukemia parallel alterations in biogenesis markers. Appl Microsc 2021; 51:20. [PMID: 34964922 PMCID: PMC8716663 DOI: 10.1186/s42649-021-00069-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/18/2021] [Indexed: 11/10/2022] Open
Abstract
We explored the link between mitochondrial biogenesis and mitochondrial morphology using transmission electron microscopy (TEM) in lymphoblasts of pediatric acute lymphoblastic leukemia (ALL) patients and compared these characteristics between tumors and control samples. Gene expression of mitochondrial biogenesis markers was analysed in 23 ALL patients and 18 controls and TEM for morphology analysis was done in 15 ALL patients and 9 healthy controls. The area occupied by mitochondria per cell and the cristae cross-sectional area was observed to be significantly higher in patients than in controls (p-value = 0.0468 and p-value< 0.0001, respectively). The mtDNA copy numbers, TFAM, POLG, and c-myc gene expression were significantly higher in ALL patients than controls (all p-values< 0.01). Gene Expression of PGC-1α was higher in tumor samples. The analysis of the correlation between PGC-1α expression and morphology parameters i.e., both M/C ratio and cristae cross-sectional area revealed a positive trend (r = 0.3, p = 0.1). The increased area occupied by mitochondria and increased cristae area support the occurrence of cristae remodelling in ALL. These changes might reflect alterations in cristae dynamics to support the metabolic state of the cells by forming a more condensed network. Ultrastructural imaging can be useful for affirming changes occurring at a subcellular organellar level.
Collapse
Affiliation(s)
- Ritika Singh
- Department of Biochemistry, Teaching Block, All India Institute of Medical Sciences, Room 3044, New Delhi, 110029, India
| | - Ayushi Jain
- Department of Biochemistry, Teaching Block, All India Institute of Medical Sciences, Room 3044, New Delhi, 110029, India
| | - Jayanth Kumar Palanichamy
- Department of Biochemistry, Convergence Block, All India Institute of Medical Sciences, New Delhi, India
| | - T C Nag
- Department of Anatomy, Teaching Block, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Archna Singh
- Department of Biochemistry, Teaching Block, All India Institute of Medical Sciences, Room 3044, New Delhi, 110029, India.
| |
Collapse
|
15
|
Pedersen SF, Flinck M, Pardo LA. The Interplay between Dysregulated Ion Transport and Mitochondrial Architecture as a Dangerous Liaison in Cancer. Int J Mol Sci 2021; 22:ijms22105209. [PMID: 34069047 PMCID: PMC8156689 DOI: 10.3390/ijms22105209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Transport of ions and nutrients is a core mitochondrial function, without which there would be no mitochondrial metabolism and ATP production. Both ion homeostasis and mitochondrial phenotype undergo pervasive changes during cancer development, and both play key roles in driving the malignancy. However, the link between these events has been largely ignored. This review comprehensively summarizes and critically discusses the role of the reciprocal relationship between ion transport and mitochondria in crucial cellular functions, including metabolism, signaling, and cell fate decisions. We focus on Ca2+, H+, and K+, which play essential and highly interconnected roles in mitochondrial function and are profoundly dysregulated in cancer. We describe the transport and roles of these ions in normal mitochondria, summarize the changes occurring during cancer development, and discuss how they might impact tumorigenesis.
Collapse
Affiliation(s)
- Stine F. Pedersen
- Department of Biology, Faculty of Science, University of Copenhagen, 2100 Copenhagen, Denmark;
- Correspondence: (S.F.P.); (L.A.P.)
| | - Mette Flinck
- Department of Biology, Faculty of Science, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Luis A. Pardo
- Oncophysiology Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Correspondence: (S.F.P.); (L.A.P.)
| |
Collapse
|
16
|
Qiao H, Zhang L, Fang D, Zhu Z, He W, Hu L, Di L, Guo Z, Wang X. Surmounting tumor resistance to metallodrugs by co-loading a metal complex and siRNA in nanoparticles. Chem Sci 2021; 12:4547-4556. [PMID: 34163720 PMCID: PMC8179575 DOI: 10.1039/d0sc06680j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Copper complexes are promising anticancer agents widely studied to overcome tumor resistance to metal-based anticancer drugs. Nevertheless, copper complexes per se encounter drug resistance from time to time. Adenosine-5'-triphosphate (ATP)-responsive nanoparticles containing a copper complex CTND and B-cell lymphoma 2 (Bcl-2) small interfering RNA (siRNA) were constructed to cope with the resistance of cancer cells to the complex. CTND and siRNA can be released from the nanoparticles in cancer cells upon reacting with intracellular ATP. The resistance of B16F10 melanoma cells to CTND was terminated by silencing the cellular Bcl-2 gene via RNA interference, and the therapeutic efficacy was significantly enhanced. The nanoparticles triggered a cellular autophagy that amplified the apoptotic signals, thus revealing a novel mechanism for antagonizing the resistance of copper complexes. In view of the extensive association of Bcl-2 protein with cancer resistance to chemotherapeutics, this strategy may be universally applicable for overcoming the ubiquitous drug resistance to metallodrugs.
Collapse
Affiliation(s)
- Hongzhi Qiao
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China .,Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Lei Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Dong Fang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Zhenzhu Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 China
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Liuqing Di
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 China
| |
Collapse
|
17
|
Datta A, Alam MJ, Khaleda L, Al-Forkan M. Protective effects of Corchorus olitorius and Butea monosperma against Arsenic induced aberrant methylation and mitochondrial DNA damage in wistar rat model. Toxicol Rep 2021; 8:30-37. [PMID: 33391994 PMCID: PMC7772721 DOI: 10.1016/j.toxrep.2020.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/07/2020] [Accepted: 12/16/2020] [Indexed: 11/26/2022] Open
Abstract
Arsenic exposure through food causes genotoxic effects in vivo. Sub chronic exposure to Arsenic causes pre-cancerous changes. C. olitorious & B. monosperma have significant nephro & hepato protective potential. Protective effect of both plants species is evident in molecular level. Potent use as readily available food supplement to minimize Arsenic toxicity.
Millions of people around the world are chronically exposed to Arsenic (As) through food and drinking water. Studies revealed that Arsenic is genotoxic and causes damage to DNA. In this study, we evaluated Corchorus olitorius and Butea monosperma for their alleviative properties against Arsenic induced genotoxicity in vivo using Wistar Rat model. Arsenic exposed rats were given C. olitorius leaf powder and B. monosperma flower powder as supplementation with normal food. Methylation status of p53 promoter was measured using Methylation Sensitive Restriction Endonuclease PCR (MSRE-PCR) assay and mitochondrial DNA (mtDNA) copy number as well as occurrence of a common deletion in mtDNA in liver and kidney tissue was determined through quantitative realtime PCR (qPCR). Arsenic exposed rats after supplementation showed relatively less severe effects of toxicity evident by significantly higher amount of (p<0.05) mtDNA copy number and reduced occurrence of deletion containing mtDNA as well as lower levels of methylation in p53 gene promoter. Histopathological analysis revealed less severe histopathological changes of liver and kidney and normal liver and kidney function parameters in supplemented rats. So, the protective properties of B. monosperma and C. olitorius against Arsenic toxicity is evident in molecular level.
Collapse
Affiliation(s)
- Amit Datta
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Md Jibran Alam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Laila Khaleda
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Mohammad Al-Forkan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, 4331, Bangladesh
| |
Collapse
|
18
|
Jiang C, Moorthy BT, Patel DM, Kumar A, Morgan WM, Alfonso B, Huang J, Lampidis TJ, Isom DG, Barrientos A, Fontanesi F, Zhang F. Regulation of Mitochondrial Respiratory Chain Complex Levels, Organization, and Function by Arginyltransferase 1. Front Cell Dev Biol 2020; 8:603688. [PMID: 33409279 PMCID: PMC7779560 DOI: 10.3389/fcell.2020.603688] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/23/2020] [Indexed: 12/24/2022] Open
Abstract
Arginyltransferase 1 (ATE1) is an evolutionary-conserved eukaryotic protein that localizes to the cytosol and nucleus. It is the only known enzyme in metazoans and fungi that catalyzes posttranslational arginylation. Lack of arginylation has been linked to an array of human disorders, including cancer, by altering the response to stress and the regulation of metabolism and apoptosis. Although mitochondria play relevant roles in these processes in health and disease, a causal relationship between ATE1 activity and mitochondrial biology has yet to be established. Here, we report a phylogenetic analysis that traces the roots of ATE1 to alpha-proteobacteria, the mitochondrion microbial ancestor. We then demonstrate that a small fraction of ATE1 localizes within mitochondria. Furthermore, the absence of ATE1 influences the levels, organization, and function of respiratory chain complexes in mouse cells. Specifically, ATE1-KO mouse embryonic fibroblasts have increased levels of respiratory supercomplexes I+III2+IVn. However, they have decreased mitochondrial respiration owing to severely lowered complex II levels, which leads to accumulation of succinate and downstream metabolic effects. Taken together, our findings establish a novel pathway for mitochondrial function regulation that might explain ATE1-dependent effects in various disease conditions, including cancer and aging, in which metabolic shifts are part of the pathogenic or deleterious underlying mechanism.
Collapse
Affiliation(s)
- Chunhua Jiang
- Department of Molecular & Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Balaji T Moorthy
- Department of Molecular & Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Devang M Patel
- Department of Molecular & Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Akhilesh Kumar
- Department of Molecular & Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - William M Morgan
- Department of Molecular & Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Belkis Alfonso
- Department of Human Genetics, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Jingyu Huang
- Department of Human Genetics, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Theodore J Lampidis
- Department of Cell Biology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Daniel G Isom
- Department of Molecular & Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Institute for Data Science and Computing, University of Miami, Coral Gables, FL, United States
| | - Antoni Barrientos
- Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Department of Biochemistry & Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Flavia Fontanesi
- Department of Biochemistry & Molecular Biology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Fangliang Zhang
- Department of Molecular & Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
19
|
Miller JM, Brambley CA, Marsee JD. Examination of the Role of Mg 2+ in the Mechanism of Nucleotide Binding to the Monomeric YME1L AAA+ Domain. Biochemistry 2020; 59:4303-4320. [PMID: 33155794 DOI: 10.1021/acs.biochem.0c00699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The first line of defense in the mitochondrial quality control network involves the stress response from a family of ATP-dependent proteases. We have reported that a solubilized version of the mitochondrial inner membrane ATP-dependent protease YME1L displays nucleotide binding kinetics that are sensitive to the reactive oxygen species hydrogen peroxide under a limiting ATP concentration. Our observations were consistent with an altered YME1L conformational ensemble leading to increased nucleotide binding site accessibility under oxidative stress conditions. To examine this hypothesis further, we report here the results of a comprehensive study of the thermodynamic and kinetic properties underlying the binding of nucleoside di- and triphosphate to the isolated YME1L AAA+ domain (YME1L-AAA+). A combination of fluorescence titrations, molecular dynamics, and stopped-flow fluorescence experiments have demonstrated similarity between nucleotide binding behaviors for YME1L under oxidative conditions and the isolated AAA+ domain. Our data demonstrate that YME1L-AAA+ binds ATP and ADP with affinities equal to ∼30 and 5 μM, respectively, in the absence of Mg2+. We note a negative heterotropic linkage effect between Mg2+ and ATP that arises as the MgCl2 concentration is increased such that the affinity of YME1L-AAA+ for ATP decreases to ∼60 μM in the presence of 10 mM MgCl2. Molecular dynamics methods allow for structural rationalization by revealing condition-dependent conformational populations for YME1L-AAA+. Taken together, these data suggest a preliminary model in which YME1L modulates its affinity for the nucleotide to stabilize against degradation or instability inherent to such stress conditions.
Collapse
Affiliation(s)
- Justin M Miller
- Department of Chemistry, Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, United States
| | - Chad A Brambley
- Department of Chemistry, Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, United States
| | - Justin D Marsee
- Department of Chemistry, Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, United States
| |
Collapse
|
20
|
Estrogen Regulation of mTOR Signaling and Mitochondrial Function in Invasive Lobular Carcinoma Cell Lines Requires WNT4. Cancers (Basel) 2020; 12:cancers12102931. [PMID: 33053661 PMCID: PMC7650584 DOI: 10.3390/cancers12102931] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Invasive lobular carcinoma (ILC) is a common but understudied breast cancer subtype. ILC is presumed to be a low-risk disease in part because nearly all ILCs contain the estrogen receptor (ER). However, we previously showed that ER has unique functions in ILC cells, including driving expression of the Wnt ligand WNT4. WNT4 signaling is required for ILC cell proliferation and survival, but the mechanisms and targets of WNT4 signaling in ILC is unknown. We found that WNT4 regulates mTOR signaling via S6 kinase, and controls levels of MCL-1 protein, ultimately regulating mitochondrial function and cellular metabolism. These findings offer new insight into a novel Wnt signaling pathway and identify new targets to inhibit WNT4 signaling as potential treatments against ILC cells. Abstract Invasive lobular carcinoma of the breast (ILC) is strongly estrogen-driven and represents a unique context for estrogen receptor (ER) signaling. In ILC, ER controls the expression of the Wnt ligand WNT4, which is critical for endocrine response and anti-estrogen resistance. However, signaling mediated by WNT4 is cell type- and tissue-specific, and has not been explored in ILC. We utilized reverse phase protein array (RPPA) to characterize ER and WNT4-driven signaling in ILC cells and identified that WNT4 mediates downstream mTOR signaling via phosphorylation of S6 Kinase. Additionally, ER and WNT4 control levels of MCL-1, which is associated with regulation of mitochondrial function. In this context, WNT4 knockdown led to decreased ATP production and increased mitochondrial fragmentation. WNT4 regulation of both mTOR signaling and MCL-1 were also observed in anti-estrogen resistant models of ILC. We identified that high WNT4 expression is associated with similar mTOR pathway activation in ILC and serous ovarian cancer tumors, suggesting that WNT4 signaling is active in multiple tumor types. The identified downstream pathways offer insight into WNT4 signaling and represent potential targets to overcome anti-estrogen resistance for patients with ILC.
Collapse
|
21
|
Xiang Y, Fang B, Liu Y, Yan S, Cao D, Mei H, Wang Q, Hu Y, Guo T. SR18292 exerts potent antitumor effects in multiple myeloma via inhibition of oxidative phosphorylation. Life Sci 2020; 256:117971. [PMID: 32553925 DOI: 10.1016/j.lfs.2020.117971] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/29/2020] [Accepted: 06/11/2020] [Indexed: 12/30/2022]
Abstract
AIMS Multiple myeloma (MM) was recently reported to rely on increased oxidative phosphorylation (OXPHOS) for survival, providing a potential opportunity for MM therapy. Herein, we aimed to propose a novel targeted drug for MM treatment, followed by the exploration of reason for OXPHOS enhancement in MM cells. MATERIALS AND METHODS The expression of OXPHOS genes and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) was analyzed using bioinformatics analyses, followed by verification in MM cell lines. The effects of SR18292 on OXPHOS were measured by qRT-PCR, Western blot, transmission electron microscopy, oxygen consumption rate and so on. The proliferation and apoptosis were evaluated by CCK-8, flow cytometry and Western blot. The efficiency and safety of SR18292 were assessed in a mouse model of MM. KEY FINDINGS The OXPHOS genes were generally overexpressed in MM cells, which was associated with poorer prognosis of MM patients. PGC-1α, a transcriptional coactivator, was upregulated in MM cells, and MM patients with higher PGC-1α expression exhibited increased enrichment of the OXPHOS gene set. Treatment with SR18292 (an inhibitor of PGC-1α) significantly impaired the proliferation and survival of MM cells due to OXPHOS metabolism dysfunction, which leads to energy exhaustion and oxidative damage. Besides, SR18292 potently inhibited tumor growth at a well-tolerated dose in MM model mice. SIGNIFICANCE The overexpression of OXPHOS gene set mediated by upregulated PGC-1α provides a structural basis for enhanced OXPHOS in MM cells, and SR18292 (a PGC-1α inhibitor) exerts potent antimyeloma effects, offering a potential tangible avenue for MM therapy.
Collapse
Affiliation(s)
- Yu Xiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bin Fang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yilin Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siqi Yan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Dedong Cao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Huiling Mei
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiuguo Wang
- Pediatrics Department of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Tao Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
22
|
Cytoprotective effects of (E)-N-(2-(3, 5-dimethoxystyryl) phenyl) furan-2-carboxamide (BK3C231) against 4-nitroquinoline 1-oxide-induced damage in CCD-18Co human colon fibroblast cells. PLoS One 2020; 15:e0223344. [PMID: 32365104 PMCID: PMC7197815 DOI: 10.1371/journal.pone.0223344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/15/2020] [Indexed: 12/31/2022] Open
Abstract
Stilbenes are a group of chemicals characterized with the presence of 1,2-diphenylethylene. Previously, our group has demonstrated that synthesized (E)-N-(2-(3, 5-dimethoxystyryl) phenyl) furan-2-carboxamide (BK3C231) possesses potential chemopreventive activity specifically inducing NAD(P)H:quinone oxidoreductase 1 (NQO1) protein expression and activity. In this study, the cytoprotective effects of BK3C231 on cellular DNA and mitochondria were investigated in normal human colon fibroblast, CCD-18Co cells. The cells were pretreated with BK3C231 prior to exposure to the carcinogen 4-nitroquinoline 1-oxide (4NQO). BK3C231 was able to inhibit 4NQO-induced cytotoxicity. Cells treated with 4NQO alone caused high level of DNA and mitochondrial damages. However, pretreatment with BK3C231 protected against these damages by reducing DNA strand breaks and micronucleus formation as well as decreasing losses of mitochondrial membrane potential (ΔΨm) and cardiolipin. Interestingly, our study has demonstrated that nitrosative stress instead of oxidative stress was involved in 4NQO-induced DNA and mitochondrial damages. Inhibition of 4NQO-induced nitrosative stress by BK3C231 was observed through a decrease in nitric oxide (NO) level and an increase in glutathione (GSH) level. These new findings elucidate the cytoprotective potential of BK3C231 in human colon fibroblast CCD-18Co cell model which warrants further investigation into its chemopreventive role.
Collapse
|
23
|
Hayes P, Fergus C, Ghanim M, Cirzi C, Burtnyak L, McGrenaghan CJ, Tuorto F, Nolan DP, Kelly VP. Queuine Micronutrient Deficiency Promotes Warburg Metabolism and Reversal of the Mitochondrial ATP Synthase in Hela Cells. Nutrients 2020; 12:nu12030871. [PMID: 32213952 PMCID: PMC7146442 DOI: 10.3390/nu12030871] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 01/01/2023] Open
Abstract
Queuine is a eukaryotic micronutrient, derived exclusively from eubacteria. It is incorporated into both cytosolic and mitochondrial transfer RNA to generate a queuosine nucleotide at position 34 of the anticodon loop. The transfer RNA of primary tumors has been shown to be hypomodified with respect to queuosine, with decreased levels correlating with disease progression and poor patient survival. Here, we assess the impact of queuine deficiency on mitochondrial bioenergetics and substrate metabolism in HeLa cells. Queuine depletion is shown to promote a Warburg type metabolism, characterized by increased aerobic glycolysis and glutaminolysis, concomitant with increased ammonia and lactate production and elevated levels of lactate dehydrogenase activity but in the absence of significant changes to proliferation. In intact cells, queuine deficiency caused an increased rate of mitochondrial proton leak and a decreased rate of ATP synthesis, correlating with an observed reduction in cellular ATP levels. Data from permeabilized cells demonstrated that the activity of individual complexes of the mitochondrial electron transport chain were not affected by the micronutrient. Notably, in queuine free cells that had been adapted to grow in galactose medium, the re-introduction of glucose permitted the mitochondrial F1FO-ATP synthase to operate in the reverse direction, acting to hyperpolarize the mitochondrial membrane potential; a commonly observed but poorly understood cancer trait. Together, our data suggest that queuosine hypomodification is a deliberate and advantageous adaptation of cancer cells to facilitate the metabolic switch between oxidative phosphorylation and aerobic glycolysis.
Collapse
Affiliation(s)
- Patti Hayes
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland; (P.H.); (C.F.); (M.G.); (L.B.); (C.J.M.); (D.P.N.)
| | - Claire Fergus
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland; (P.H.); (C.F.); (M.G.); (L.B.); (C.J.M.); (D.P.N.)
| | - Magda Ghanim
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland; (P.H.); (C.F.); (M.G.); (L.B.); (C.J.M.); (D.P.N.)
| | - Cansu Cirzi
- Division of Epigenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; (C.C.); (F.T.)
- Faculty of Biosciences, University of Heidelberg, 69120 Heidelberg, Germany
| | - Lyubomyr Burtnyak
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland; (P.H.); (C.F.); (M.G.); (L.B.); (C.J.M.); (D.P.N.)
| | - Callum J. McGrenaghan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland; (P.H.); (C.F.); (M.G.); (L.B.); (C.J.M.); (D.P.N.)
| | - Francesca Tuorto
- Division of Epigenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; (C.C.); (F.T.)
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty, Heidelberg University, 68167 Mannheim, Germany
| | - Derek P. Nolan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland; (P.H.); (C.F.); (M.G.); (L.B.); (C.J.M.); (D.P.N.)
| | - Vincent P. Kelly
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 2 Dublin, Ireland; (P.H.); (C.F.); (M.G.); (L.B.); (C.J.M.); (D.P.N.)
- Correspondence: ; Tel.: +353-1-8963507
| |
Collapse
|
24
|
Tian J, Shi R, Xiao P, Liu T, She R, Wu Q, An J, Hao W, Soomro M. Hepatitis E Virus Induces Brain Injury Probably Associated With Mitochondrial Apoptosis. Front Cell Infect Microbiol 2019; 9:433. [PMID: 31921708 PMCID: PMC6932957 DOI: 10.3389/fcimb.2019.00433] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/04/2019] [Indexed: 01/01/2023] Open
Abstract
Hepatitis E virus (HEV) infection has been associated with extrahepatic manifestations, particularly neurological disorders. Although it has been reported that HEV infection induced hepatocyte apoptosis associated with mitochondria injury, activation of mitochondrial apoptotic pathway in the central nervous system during HEV infection was not clearly understood. In this study, the induction of mitochondrial apoptosis-associated proteins and pro-inflammatory cytokines were detected in HEV infected Mongolian gerbil model and primary human brain microvascular endothelial cells (HBMVECs). Mitochondrial exhibited fragments with loss of cristae and matrix in HEV infected brain tissue by transmission electron microscope (TEM). In vitro studies showed that expression of NADPH oxidase 4 (NOX4) was significantly increased in HEV infected HBMVECs (p < 0.05), while ATP5A1 was significantly decreased (p < 0.01). Expressions of pro-apoptotic proteins were further evaluated. Bax was significantly increased in both HEV infected brain tissues and HBMVECs (p < 0.01). In vivo studies showed that caspase-9 and caspase-3 were activated after HEV inoculation (p < 0.01), associated with PCNA overexpression as response to apoptosis. Cytokines were measured to evaluate tissue inflammatory levels. Results showed that the release of TNFα and IL-1β were significantly increased after HEV infection (p < 0.01), which might be attributed to microglia activation characterized by high level of IBA1 expression (p < 0.01). Taken together, these data support that HEV infection induces high levels of pro-inflammatory cytokines, associated with mitochondria-mediated apoptosis. The results provide new insight into mechanisms of extra-hepatic injury of HEV infection, especially in the central nervous system.
Collapse
Affiliation(s)
- Jijing Tian
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ruihan Shi
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China.,Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Peng Xiao
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Tianlong Liu
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ruiping She
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qiaoxing Wu
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Junqing An
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wenzhuo Hao
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - MajidHussain Soomro
- Laboratory of Animal Pathology and Public Health, Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
25
|
Hiyoshi Y, Sato Y, Ichinoe M, Nagashio R, Hagiuda D, Kobayashi M, Kusuhara S, Igawa S, Shiomi K, Goshima N, Murakumo Y, Saegusa M, Satoh Y, Masuda N, Naoki K. Prognostic significance of IMMT expression in surgically-resected lung adenocarcinoma. Thorac Cancer 2019; 10:2142-2151. [PMID: 31583841 PMCID: PMC6825906 DOI: 10.1111/1759-7714.13200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction contributes to many types of human disorders and cancer progression. Inner membrane mitochondrial protein (IMMT) plays an important role in the maintenance of mitochondrial structure and function. The aims of this study were to examine IMMT expression in lung adenocarcinoma and evaluate its correlation with clinicopathological parameters and patient prognosis. METHODS IMMT expression was immunohistochemically studied in 176 consecutive lung adenocarcinoma resection tissues, and its correlations with clinicopathological parameters were evaluated. Kaplan-Meier survival analysis and Cox-proportional hazards models were used to estimate the effect of IMMT expression on survival. RESULTS High-IMMT expression was detected in 84 of 176 (47.7%) lung adenocarcinomas. Levels were significantly correlated with advanced disease stage (stage II and III; P = 0.024), larger tumor size (>3 cm; P = 0.002), intratumoral vascular invasion (P < 0.001), and poorer adenocarcinoma patient prognosis (P = 0.002). Based on 176 patients with adenocarcinoma, multivariate analysis revealed that IMMT expression was an independent predictor of poorer survival (HR, 1.99; 95% confidence interval [CI], 1.06-3.74; P = 0.031). Further, treating A549 cells derived from lung adenocarcinoma, with IMMT siRNA resulted in significantly decreased proliferation. CONCLUSION Here, we first demonstrated that high-IMMT expression is related to some clinicopathological parameters, and that its expression is an independent prognostic predictor of poorer survival in patients with lung adenocarcinoma; further studies are required to clarify the biological function of IMMT in lung adenocarcinoma. However, results suggest that this protein could be a novel prognostic indicator and therapeutic target.
Collapse
Affiliation(s)
- Yasuhiro Hiyoshi
- Department of Respiratory Medicine, School of MedicineKitasato UniversitySagamiharaJapan
| | - Yuichi Sato
- Applied Tumor Pathology, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
| | - Masaaki Ichinoe
- Department of Pathology, School of MedicineKitasato UniversitySagamiharaJapan
| | - Ryo Nagashio
- Applied Tumor Pathology, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
| | - Daisuke Hagiuda
- Applied Tumor Pathology, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
| | - Makoto Kobayashi
- Applied Tumor Pathology, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
| | - Seiichiro Kusuhara
- Department of Respiratory Medicine, School of MedicineKitasato UniversitySagamiharaJapan
| | - Satoshi Igawa
- Department of Respiratory Medicine, School of MedicineKitasato UniversitySagamiharaJapan
| | - Kazu Shiomi
- Department of Thoracic and Cardiovascular Surgery, School of MedicineKitasato UniversitySagamiharaJapan
| | - Naoki Goshima
- National Institute of Advanced Industrial Science and Technology Tokyo Bay Area CenterKoto‐kuJapan
| | - Yoshiki Murakumo
- Department of Pathology, School of MedicineKitasato UniversitySagamiharaJapan
| | - Makoto Saegusa
- Department of Pathology, School of MedicineKitasato UniversitySagamiharaJapan
| | - Yukitoshi Satoh
- Department of Thoracic and Cardiovascular Surgery, School of MedicineKitasato UniversitySagamiharaJapan
| | - Noriyuki Masuda
- Department of Respiratory Medicine, School of MedicineKitasato UniversitySagamiharaJapan
| | - Katsuhiko Naoki
- Department of Respiratory Medicine, School of MedicineKitasato UniversitySagamiharaJapan
| |
Collapse
|
26
|
Viana LR, Tobar N, Busanello ENB, Marques AC, de Oliveira AG, Lima TI, Machado G, Castelucci BG, Ramos CD, Brunetto SQ, Silveira LR, Vercesi AE, Consonni SR, Gomes-Marcondes MCC. Leucine-rich diet induces a shift in tumour metabolism from glycolytic towards oxidative phosphorylation, reducing glucose consumption and metastasis in Walker-256 tumour-bearing rats. Sci Rep 2019; 9:15529. [PMID: 31664147 PMCID: PMC6820796 DOI: 10.1038/s41598-019-52112-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 10/14/2019] [Indexed: 11/09/2022] Open
Abstract
Leucine can stimulate protein synthesis in skeletal muscle, and recent studies have shown an increase in leucine-related mitochondrial biogenesis and oxidative phosphorylation capacity in muscle cells. However, leucine-related effects in tumour tissues are still poorly understood. Thus, we described the effects of leucine in both in vivo and in vitro models of a Walker-256 tumour. Tumour-bearing Wistar rats were randomly distributed into a control group (W; normoprotein diet) and leucine group (LW; leucine-rich diet [normoprotein + 3% leucine]). After 20 days of tumour evolution, the animals underwent 18-fludeoxyglucose positron emission computed tomography (18F-FDG PET-CT) imaging, and after euthanasia, fresh tumour biopsy samples were taken for oxygen consumption rate measurements (Oroboros Oxygraph), electron microscopy analysis and RNA and protein extraction. Our main results from the LW group showed no tumour size change, lower tumour glucose (18F-FDG) uptake, and reduced metastatic sites. Furthermore, leucine stimulated a shift in tumour metabolism from glycolytic towards oxidative phosphorylation, higher mRNA and protein expression of oxidative phosphorylation components, and enhanced mitochondrial density/area even though the leucine-treated tumour had a higher number of apoptotic nuclei with increased oxidative stress. In summary, a leucine-rich diet directed Walker-256 tumour metabolism to a less glycolytic phenotype profile in which these metabolic alterations were associated with a decrease in tumour aggressiveness and reduction in the number of metastatic sites in rats fed a diet supplemented with this branched-chain amino acid.
Collapse
Affiliation(s)
- Laís Rosa Viana
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Biology Institute, University of Campinas, Campinas, SP, Brazil
| | - Natália Tobar
- Division of Nuclear Medicine, Department of Radiology, School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | | | - Ana Carolina Marques
- School of Medical Sciences, Department of Clinical Pathology, State University of Campinas, Campinas, SP, Brazil
| | - Andre Gustavo de Oliveira
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Biology Institute, University of Campinas, Campinas, SP, Brazil.,Obesity and Comorbidities Research Centre, Biology Institute, University of Campinas, Campinas, SP, Brazil
| | - Tanes I Lima
- Obesity and Comorbidities Research Centre, Biology Institute, University of Campinas, Campinas, SP, Brazil
| | - Gabrielly Machado
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Biology Institute, University of Campinas, Campinas, SP, Brazil
| | - Bianca Gazieri Castelucci
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology. Biology Institute, University of Campinas, Campinas, SP, Brazil
| | - Celso Dario Ramos
- Division of Nuclear Medicine, Department of Radiology, School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Sérgio Q Brunetto
- Division of Nuclear Medicine, Department of Radiology, School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Leonardo Reis Silveira
- Obesity and Comorbidities Research Centre, Biology Institute, University of Campinas, Campinas, SP, Brazil
| | - Anibal Eugenio Vercesi
- School of Medical Sciences, Department of Clinical Pathology, State University of Campinas, Campinas, SP, Brazil
| | - Sílvio Roberto Consonni
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology. Biology Institute, University of Campinas, Campinas, SP, Brazil
| | - Maria Cristina Cintra Gomes-Marcondes
- Laboratory of Nutrition and Cancer, Department of Structural and Functional Biology, Biology Institute, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
27
|
Moscheni C, Malucelli E, Castiglioni S, Procopio A, De Palma C, Sorrentino A, Sartori P, Locatelli L, Pereiro E, Maier JA, Iotti S. 3D Quantitative and Ultrastructural Analysis of Mitochondria in a Model of Doxorubicin Sensitive and Resistant Human Colon Carcinoma Cells. Cancers (Basel) 2019; 11:cancers11091254. [PMID: 31461915 PMCID: PMC6769783 DOI: 10.3390/cancers11091254] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/27/2022] Open
Abstract
Drug resistance remains a major obstacle in cancer treatment. Because mitochondria mediate metabolic reprogramming in cancer drug resistance, we focused on these organelles in doxorubicin sensitive and resistant colon carcinoma cells. We employed soft X-ray cryo nano-tomography to map three-dimensionally these cells at nanometer-resolution and investigate the correlation between mitochondrial morphology and drug resistance phenotype. We have identified significant structural differences in the morphology of mitochondria in the two strains of cancer cells, as well as lower amounts of Reactive oxygen species (ROS) in resistant than in sensitive cells. We speculate that these features could elicit an impaired mitochondrial communication in resistant cells, thus preventing the formation of the interconnected mitochondrial network as clearly detected in the sensitive cells. In fact, the qualitative and quantitative three-dimensional assessment of the mitochondrial morphology highlights a different structural organization in resistant cells, which reflects a metabolic cellular adaptation functional to survive to the offense exerted by the antineoplastic treatment.
Collapse
Affiliation(s)
- Claudia Moscheni
- Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli Studi di Milano, 20157 Milano, Italy
| | - Emil Malucelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli Studi di Milano, 20157 Milano, Italy.
| | - Alessandra Procopio
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
| | - Clara De Palma
- Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli Studi di Milano, 20157 Milano, Italy
- Unit of Clinical Pharmacology, "Luigi Sacco" University Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy
| | - Andrea Sorrentino
- ALBA Synchrotron Light Facility, Carrer de la Llum 2-26, 08290 Cerdanyola del Vallès, Spain
| | - Patrizia Sartori
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Laura Locatelli
- Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli Studi di Milano, 20157 Milano, Italy
| | - Eva Pereiro
- ALBA Synchrotron Light Facility, Carrer de la Llum 2-26, 08290 Cerdanyola del Vallès, Spain
| | - Jeanette A Maier
- Department of Biomedical and Clinical Sciences "Luigi Sacco", Università degli Studi di Milano, 20157 Milano, Italy
| | - Stefano Iotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy
- National Institute of Biostructures and Biosystems, 00136 Roma, Italy
| |
Collapse
|
28
|
Abstract
Significance: In addition to their classical role in cellular ATP production, mitochondria are of key relevance in various (patho)physiological mechanisms including second messenger signaling, neuro-transduction, immune responses and death induction. Recent Advances: Within cells, mitochondria are motile and display temporal changes in internal and external structure ("mitochondrial dynamics"). During the last decade, substantial empirical and in silico evidence was presented demonstrating that mitochondrial dynamics impacts on mitochondrial function and vice versa. Critical Issues: However, a comprehensive and quantitative understanding of the bidirectional links between mitochondrial external shape, internal structure and function ("morphofunction") is still lacking. The latter particularly hampers our understanding of the functional properties and behavior of individual mitochondrial within single living cells. Future Directions: In this review we discuss the concept of mitochondrial morphofunction in mammalian cells, primarily using experimental evidence obtained within the last decade. The topic is introduced by briefly presenting the central role of mitochondria in cell physiology and the importance of the mitochondrial electron transport chain (ETC) therein. Next, we summarize in detail how mitochondrial (ultra)structure is controlled and discuss empirical evidence regarding the equivalence of mitochondrial (ultra)structure and function. Finally, we provide a brief summary of how mitochondrial morphofunction can be quantified at the level of single cells and mitochondria, how mitochondrial ultrastructure/volume impacts on mitochondrial bioreactions and intramitochondrial protein diffusion, and how mitochondrial morphofunction can be targeted by small molecules.
Collapse
Affiliation(s)
- Elianne P. Bulthuis
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Merel J.W. Adjobo-Hermans
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Peter H.G.M. Willems
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Werner J.H. Koopman
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
- Address correspondence to: Dr. Werner J.H. Koopman, Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, P.O. Box 9101, Nijmegen NL-6500 HB, The Netherlands
| |
Collapse
|
29
|
Myoferlin Contributes to the Metastatic Phenotype of Pancreatic Cancer Cells by Enhancing Their Migratory Capacity through the Control of Oxidative Phosphorylation. Cancers (Basel) 2019; 11:cancers11060853. [PMID: 31248212 PMCID: PMC6628295 DOI: 10.3390/cancers11060853] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/13/2019] [Accepted: 06/16/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies with an overall survival of 5% and is the second cause of death by cancer, mainly linked to its high metastatic aggressiveness. Accordingly, understanding the mechanisms sustaining the PDAC metastatic phenotype remains a priority. In this study, we generated and used a murine in vivo model to select clones from the human Panc-1 PDAC cell line that exhibit a high propensity to seed and metastasize into the liver. We showed that myoferlin, a protein previously reported to be overexpressed in PDAC, is significantly involved in the migratory abilities of the selected cells. We first report that highly metastatic Panc-1 clones expressed a significantly higher myoferlin level than the corresponding low metastatic ones. Using scratch wound and Boyden’s chamber assays, we show that cells expressing a high myoferlin level have higher migratory potential than cells characterized by a low myoferlin abundance. Moreover, we demonstrate that myoferlin silencing leads to a migration decrease associated with a reduction of mitochondrial respiration. Since mitochondrial oxidative phosphorylation has been shown to be implicated in the tumor progression and dissemination, our data identify myoferlin as a valid potential therapeutic target in PDAC.
Collapse
|
30
|
Aung L, Li YZ, Yu H, Chen X, Yu Z, Gao J, Li P. Mitochondrial protein 18 is a positive apoptotic regulator in cardiomyocytes under oxidative stress. Clin Sci (Lond) 2019; 133:1067-1084. [DOI: 10.1042/cs20190125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Abstract
Accumulation of reactive oxygen species is a common phenomenon in cardiac stress conditions, for instance, coronary artery disease, aging-related cardiovascular abnormalities, and exposure to cardiac stressors such as hydrogen peroxide (H2O2). Mitochondrial protein 18 (Mtp18) is a novel mitochondrial inner membrane protein, shown to involve in the regulation of mitochondrial dynamics. Although Mtp18 is abundant in cardiac muscles, its role in cardiac apoptosis remains elusive. The present study aimed to detect the role of Mtp18 in H2O2-induced mitochondrial fission and apoptosis in cardiomyocytes. We studied the effect of Mtp18 in cardiomyocytes by modulating its expression with lentiviral construct of Mtp18-shRNA and Mtp18 c-DNA, respectively. We then analyzed mitochondrial morphological dynamics with MitoTracker Red staining; apoptosis with terminal deoxynucleotidyl transferase-mediated dUTP nick-end-labeling (TUNEL) and cell death detection assays; and protein expression with immunoblotting. Here, we observed that Mtp18 could regulate oxidative stress- mediated mitochondrial fission and apoptosis in cardiac myocytes. Mechanistically, we found that Mtp8 induced mitochondrial fission and apoptosis by enhancing dynamin-related protein 1 (Drp1) accumulation. Conversely, knockdown of Mtp18 interfered with Drp1-associated mitochondrial fission and subsequent activation of apoptosis in both HL-1 cells and primary cardiomyocytes. However, overexpression of Mtp18 alone was not sufficient to execute apoptosis when Drp1 was minimally expressed, suggesting that Mtp18 and Drp1 are interdependent in apoptotic cascade. Together, these data highlight the role of Mtp18 in cardiac apoptosis and provide a novel therapeutic insight to minimize cardiomyocyte loss via targetting mitochondrial dynamics.
Collapse
Affiliation(s)
- Lynn H.H. Aung
- Center for Molecular Genetics, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yu-Zhen Li
- Department of Pathophysiology, Institute of Basic Medical Science, PLA General Hospital, Beijing 100853, China
| | - Hua Yu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao 266000, China
| | - Xiatian Chen
- Center for Molecular Genetics, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Zhongjie Yu
- Center for Molecular Genetics, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Jinning Gao
- Center for Molecular Genetics, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
31
|
Abstract
Mitochondria are ubiquitous and multi-functional organelles involved in diverse metabolic processes, namely energy production and biomolecule synthesis. The intracellular mitochondrial morphology and distribution change dynamically, which reflect the metabolic state of a given cell type. A dramatic change of the mitochondrial dynamics has been observed in early development that led to further investigations on the relationship between mitochondria and the process of development. A significant developmental process to focus on, in this review, is a differentiation of neural progenitor cells into neurons. Information on how mitochondria- regulated cellular energetics is linked to neuronal development will be discussed, followed by functions of mitochondria and associated diseases in neuronal development. Lastly, the potential use of mitochondrial features in analyzing various neurodevelopmental diseases will be addressed. [BMB Reports 2018; 51(11): 549-556].
Collapse
Affiliation(s)
- Geurim Son
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| | - Jinju Han
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| |
Collapse
|
32
|
Reactive oxygen species and cancer: A complex interaction. Cancer Lett 2019; 452:132-143. [PMID: 30905813 DOI: 10.1016/j.canlet.2019.03.020] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/21/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022]
Abstract
Elevated levels of Reactive Oxygen Species (ROS), increased antioxidant ability and the maintenance of redox homeostasis can cumulatively contribute to tumor progression and metastasis. The sources and the role of ROS in a heterogeneous tumor microenvironment can vary at different stages of tumor: initiation, development, and progression, thus making it a complex subject. In this review, we have summarized the sources of ROS generation in cancer cells, its role in the tumor microenvironment, the possible functions of ROS and its important scavenger systems in tumor progression with special emphasis on solid tumors.
Collapse
|
33
|
Li Y, Tran Q, Shrestha R, Piao L, Park S, Park J, Park J. LETM1 is required for mitochondrial homeostasis and cellular viability (Review). Mol Med Rep 2019; 19:3367-3375. [PMID: 30896806 PMCID: PMC6471456 DOI: 10.3892/mmr.2019.10041] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/12/2019] [Indexed: 12/13/2022] Open
Abstract
Leucine zipper/EF-hand-containing transmembrane protein 1 (LETM1) has been identified as the gene responsible for Wolf-Hirschhorn syndrome (WHS), which is characterized by intellectual disability, epilepsy, growth delay and craniofacial dysgenesis. LETM1 is a mitochondrial inner membrane protein that encodes a homolog of the yeast protein Mdm38, which is involved in mitochondrial morphology. In the present review, the importance of LETM1 in WHS and its role within the mitochondrion was explored. LETM1 governs the mitochondrion ion channel and is involved in mitochondrial respiration. Recent studies have reported that LETM1 acts as a mitochondrial Ca2+/H+ antiporter. LETM1 has also been identified as a K+/H+ exchanger, and serves a role in Mg2+ homeostasis. The function of LETM1 in mitochondria regulation is regulated by its binding partners, carboxyl-terminal modulator protein and mitochondrial ribosomal protein L36. Therefore, we describe the remarkable role of LETM1 in mitochondrial network physiology and its function in mitochondrion-mediated cell death. In the context of these findings, we suggest that the participation of LETM1 in tumorigenesis through the alteration of cancer metabolism should be investigated. This review provides a comprehensive description of LETM1 function, which is required for mitochondrial homeostasis and cellular viability.
Collapse
Affiliation(s)
- Yuwen Li
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Quangdon Tran
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Robin Shrestha
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Longzhen Piao
- Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Sungjin Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jisoo Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
34
|
Alavi MV. Targeted OMA1 therapies for cancer. Int J Cancer 2019; 145:2330-2341. [PMID: 30714136 DOI: 10.1002/ijc.32177] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/20/2019] [Accepted: 01/23/2019] [Indexed: 12/12/2022]
Abstract
The mitochondrial inner membrane proteins OMA1 and OPA1 belong to the BAX/BAK1-dependent apoptotic signaling pathway, which can be regulated by tumor protein p53 and the prohibitins PHB and PHB2 in the context of neoplastic disease. For the most part these proteins have been studied separate from each other. Here, I argue that the OMA1 mechanism of action represents the missing link between p53 and cytochrome c release. The mitochondrial fusion protein OPA1 is cleaved by OMA1 in a stress-dependent manner generating S-OPA1. Excessive S-OPA1 can facilitate outer membrane permeabilization upon BAX/BAK1 activation through its membrane shaping properties. p53 helps outer membrane permeabilization in a 2-step process. First, cytosolic p53 activates BAX/BAK1 at the mitochondrial surface. Then, in a second step, p53 binds to prohibitin thereby releasing the restraint on OMA1. This activates OMA1, which cleaves OPA1 and promotes cytochrome c release. Clearly, OMA1 and OPA1 are not root causes for cancer. Yet many cancer cells rely on this pathway for survival, which can explain why loss of p53 function promotes tumor growth and confers resistance to chemotherapies.
Collapse
|
35
|
Meng L, Xin G, Li B, Li D, Sun X, Yan T, Li L, Shi L, Cao S, Meng X. Anthocyanins Extracted from Aronia melanocarpa Protect SH-SY5Y Cells against Amyloid-beta (1-42)-Induced Apoptosis by Regulating Ca 2+ Homeostasis and Inhibiting Mitochondrial Dysfunction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:12967-12977. [PMID: 30415542 DOI: 10.1021/acs.jafc.8b05404] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We investigated the cytoprotective effects of anthocyanins in Aronia melanocarpa against apoptosis induced by Aβ1-42, a key mediator of AD pathophysiology. We measured intracellular calcium with a colorimetric kit, cellular apoptosis with DAPI, intracellular ROS with the fluorescent marker 2,3-dimethoxy-1,4-naphthoquinone, mitochondrial membrane potential with JC-1, and ATP with a colorimetric kit. Gene transcription and protein expression levels of calmodulin, cytochrome c, caspase-9, cleaved caspase-3, Bcl-2, and Bax were analyzed by RT-PCR and Western blotting. The results showed that pretreatment with anthocyanins significantly inhibited Aβ1-42-induced apoptosis, decreased intracellular calcium and ROS, and increased ATP and mitochondrial membrane potential. RT-PCR and Western blotting revealed that anthocyanins upregulated the gene transcription and protein expression of calmodulin and Bcl-2 and downregulated those of cytochrome c, caspase-9, cleaved caspase-3, and Bax. A. melanocarpa anthocyanins protected SH-SY5Y cells against Aβ1-42-induced apoptosis by regulating Ca2+ homeostasis and apoptosis-related genes and inhibiting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Lingshuai Meng
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Guang Xin
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Bin Li
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Dongnan Li
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Xiyun Sun
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Tingcai Yan
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Li Li
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Lin Shi
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| | - Sen Cao
- School of Food and Pharmaceutical Engineering , Guiyang College , Guiyang , Guizhou 550000 , P. R. China
| | - Xianjun Meng
- College of Food Science , Shenyang Agricultural University , Shenyang , Liaoning 110866 , P. R. China
| |
Collapse
|
36
|
Son G, Han J. Roles of mitochondria in neuronal development. BMB Rep 2018; 51:549-556. [PMID: 30269744 PMCID: PMC6283025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Indexed: 04/06/2024] Open
Abstract
Mitochondria are ubiquitous and multi-functional organelles involved in diverse metabolic processes, namely energy production and biomolecule synthesis. The intracellular mitochondrial morphology and distribution change dynamically, which reflect the metabolic state of a given cell type. A dramatic change of the mitochondrial dynamics has been observed in early development that led to further investigations on the relationship between mitochondria and the process of development. A significant developmental process to focus on, in this review, is a differentiation of neural progenitor cells into neurons. Information on how mitochondria- regulated cellular energetics is linked to neuronal development will be discussed, followed by functions of mitochondria and associated diseases in neuronal development. Lastly, the potential use of mitochondrial features in analyzing various neurodevelopmental diseases will be addressed. [BMB Reports 2018; 51(11): 549-556].
Collapse
Affiliation(s)
- Geurim Son
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| | - Jinju Han
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141,
Korea
| |
Collapse
|
37
|
De R, Sarkar S, Mazumder S, Debsharma S, Siddiqui AA, Saha SJ, Banerjee C, Nag S, Saha D, Pramanik S, Bandyopadhyay U. Macrophage migration inhibitory factor regulates mitochondrial dynamics and cell growth of human cancer cell lines through CD74-NF-κB signaling. J Biol Chem 2018; 293:19740-19760. [PMID: 30366984 DOI: 10.1074/jbc.ra118.003935] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 09/25/2018] [Indexed: 12/13/2022] Open
Abstract
The indispensable role of macrophage migration inhibitory factor (MIF) in cancer cell proliferation is unambiguous, although which specific roles the cytokine plays to block apoptosis by preserving cell growth is still obscure. Using different cancer cell lines (AGS, HepG2, HCT116, and HeLa), here we report that the silencing of MIF severely deregulated mitochondrial structural dynamics by shifting the balance toward excess fission, besides inducing apoptosis with increasing sub-G0 cells. Furthermore, enhanced mitochondrial Bax translocation along with cytochrome c release, down-regulation of Bcl-xL, and Bcl-2 as well as up-regulation of Bad, Bax, and p53 indicated the activation of a mitochondrial pathway of apoptosis upon MIF silencing. The data also indicate a concerted down-regulation of Opa1 and Mfn1 along with a significant elevation of Drp1, cumulatively causing mitochondrial fragmentation upon MIF silencing. Up-regulation of Drp1 was found to be further coupled with fissogenic serine 616 phosphorylation and serine 637 dephosphorylation, thus ensuring enhanced mitochondrial translocation. Interestingly, MIF silencing was found to be associated with decreased NF-κB activation. In fact, NF-κB knockdown in turn increased mitochondrial fission and cell death. In addition, the silencing of CD74, the cognate receptor of MIF, remarkably increased mitochondrial fragmentation in addition to preventing cell proliferation, inducing mitochondrial depolarization, and increasing apoptotic cell death. This indicates the active operation of a MIF-regulated CD74-NF-κB signaling axis for maintaining mitochondrial stability and cell growth. Thus, we propose that MIF, through CD74, constitutively activates NF-κB to control mitochondrial dynamics and stability for promoting carcinogenesis via averting apoptosis.
Collapse
Affiliation(s)
- Rudranil De
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Souvik Sarkar
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Somnath Mazumder
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Subhashis Debsharma
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Asim Azhar Siddiqui
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Shubhra Jyoti Saha
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Chinmoy Banerjee
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Shiladitya Nag
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Debanjan Saha
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Saikat Pramanik
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| | - Uday Bandyopadhyay
- From the Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, West Bengal, India
| |
Collapse
|
38
|
Spindola DG, Hinsberger A, Antunes VMDS, Michelin LFG, Bincoletto C, Oliveira CR. In vitro cytotoxicity of chemical preservatives on human fibroblast cells. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000100031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
| | | | | | | | | | - Carlos Rocha Oliveira
- Universidade Anhembi Morumbi, Brazil; Instituto de Osmologia e Óleos Essenciais, Brasil; Universidade Federal de São Paulo, Brazil
| |
Collapse
|
39
|
Myoferlin controls mitochondrial structure and activity in pancreatic ductal adenocarcinoma, and affects tumor aggressiveness. Oncogene 2018; 37:4398-4412. [PMID: 29720728 PMCID: PMC6085282 DOI: 10.1038/s41388-018-0287-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/08/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related death. Therapeutic options remain very limited and are based on classical chemotherapies. Energy metabolism reprogramming appears as an emerging hallmark of cancer and is considered a therapeutic target with considerable potential. Myoferlin, a ferlin family member protein overexpressed in PDAC, is involved in plasma membrane biology and has a tumor-promoting function. In the continuity of our previous studies, we investigated the role of myoferlin in the context of energy metabolism in PDAC. We used selected PDAC tumor samples and PDAC cell lines together with small interfering RNA technology to study the role of myoferlin in energetic metabolism. In PDAC patients, we showed that myoferlin expression is negatively correlated with overall survival and with glycolytic activity evaluated by 18F-deoxyglucose positron emission tomography. We found out that myoferlin is more abundant in lipogenic pancreatic cancer cell lines and is required to maintain a branched mitochondrial structure and a high oxidative phosphorylation activity. The observed mitochondrial fission induced by myoferlin depletion led to a decrease of cell proliferation, ATP production, and autophagy induction, thus indicating an essential role of myoferlin for PDAC cell fitness. The metabolic phenotype switch generated by myoferlin silencing could open up a new perspective in the development of therapeutic strategies, especially in the context of energy metabolism.
Collapse
|
40
|
Mukherjee A, Patra U, Bhowmick R, Chawla-Sarkar M. Rotaviral nonstructural protein 4 triggers dynamin-related protein 1-dependent mitochondrial fragmentation during infection. Cell Microbiol 2018; 20:e12831. [PMID: 29444369 DOI: 10.1111/cmi.12831] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/06/2018] [Accepted: 02/06/2018] [Indexed: 12/16/2022]
Abstract
Dynamic equilibrium between mitochondrial fission and mitochondrial fusion serves as an important quality control system within cells ensuring cellular vitality and homeostasis. Viruses often target mitochondrial dynamics as a part of their obligatory cellular reprogramming. The present study was undertaken to assess the status and regulation of mitochondrial dynamics during rotavirus infection. Distinct fragmentation of mitochondrial syncytia was observed during late hours of RV (SA11, Wa, A5-13) infection. RV nonstructural protein 4 (NSP4) was identified as the viral trigger for disrupted mitochondrial morphology. Severance of mitochondrial interconnections was found to be a dynamin-related protein 1 (Drp1)-dependent process resulting synergistically from augmented mitochondrial fission and attenuated mitochondrial fusion. Cyclin-dependent kinase 1 was subsequently identified as the cellular kinase responsible for fission-active Ser616 phosphorylation of Drp1. In addition to its positive role in mitochondrial fission, Drp1 also resulted in mitochondrial translocation of E3-ubiquitin ligase Parkin leading to degradation of mitochondrial fusion protein Mitofusin 1. Interestingly, RV-NSP4 was found to interact with and be involved in recruiting fission-active pool of Serine 616 phosphoDrp1 (Ser616 pDrp1) to mitochondria independent of accessory adaptors Mitochondrial fission factor and Fission protein 1 (Fis1). Inhibition of either Drp1 or Ser616 pDrp1 resulted in significant decrease in RV-NSP4-induced intrinsic apoptotic pathway. Overall, this study underscores an efficient strategy utilised by RV to couple apoptosis to mitochondrial fission facilitating dissemination of viral progeny.
Collapse
Affiliation(s)
- Arpita Mukherjee
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Upayan Patra
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Rahul Bhowmick
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Mamta Chawla-Sarkar
- Division of Virology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
41
|
Kim YY, Yun SH, Yun J. Downregulation of Drp1, a fission regulator, is associated with human lung and colon cancers. Acta Biochim Biophys Sin (Shanghai) 2018; 50:209-215. [PMID: 29329364 DOI: 10.1093/abbs/gmx137] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 11/30/2017] [Indexed: 12/25/2022] Open
Abstract
Dynamin-related protein 1 (Drp1), a dynamin-related GTPase, is a key regulator of mitochondrial fission. Although recent studies have shown that Drp1 plays important roles in various important cellular processes, such as maintaining proper mitochondrial function, apoptosis and necrosis, the potential involvement of Drp1 in cancer development has not been fully addressed. To explore the role of Drp1 in cancer, we examined Drp1 levels in various human cancer tissues. Tissue array analysis showed that the level of Drp1 was decreased significantly in malignant colon and lung cancer tissues, whereas no change in Drp1 was observed in breast and prostate tumors. Pairwise comparisons of cancer tissue and adjacent normal tissue from colon and lung cancer patients further confirmed decreases in Drp1 expression of 75% in colon cancer patients and 78% in lung cancer patients. Moreover, Drp1 levels were decreased further with advanced grade in both colon and lung cancers, suggesting that loss of Drp1 is associated with the progression of human lung and colon cancer. Consistent with this observation, knockdown of Drp1 increased cellular migration activity in human lung cancer cells and tumor formation in a xenograft tumor model. Taken together, these results suggest that the loss of Drp1 expression could contribute to the development of human lung and colon cancers.
Collapse
Affiliation(s)
- Young Yeon Kim
- Department of Biochemistry College of Medicine, Dong-A University, Busan 49201, Repuplic of Korea
- Peripheral Neuropathy Research Center, College of Medicine, Dong-A University, Busan 49201, Republic of Korea
| | - Seong-Hoon Yun
- Department of Biochemistry College of Medicine, Dong-A University, Busan 49201, Repuplic of Korea
- Peripheral Neuropathy Research Center, College of Medicine, Dong-A University, Busan 49201, Republic of Korea
| | - Jeanho Yun
- Department of Biochemistry College of Medicine, Dong-A University, Busan 49201, Repuplic of Korea
- Peripheral Neuropathy Research Center, College of Medicine, Dong-A University, Busan 49201, Republic of Korea
| |
Collapse
|
42
|
Regulation of Tumor Progression by Programmed Necrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3537471. [PMID: 29636841 PMCID: PMC5831895 DOI: 10.1155/2018/3537471] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022]
Abstract
Rapidly growing malignant tumors frequently encounter hypoxia and nutrient (e.g., glucose) deprivation, which occurs because of insufficient blood supply. This results in necrotic cell death in the core region of solid tumors. Necrotic cells release their cellular cytoplasmic contents into the extracellular space, such as high mobility group box 1 (HMGB1), which is a nonhistone nuclear protein, but acts as a proinflammatory and tumor-promoting cytokine when released by necrotic cells. These released molecules recruit immune and inflammatory cells, which exert tumor-promoting activity by inducing angiogenesis, proliferation, and invasion. Development of a necrotic core in cancer patients is also associated with poor prognosis. Conventionally, necrosis has been thought of as an unregulated process, unlike programmed cell death processes like apoptosis and autophagy. Recently, necrosis has been recognized as a programmed cell death, encompassing processes such as oncosis, necroptosis, and others. Metabolic stress-induced necrosis and its regulatory mechanisms have been poorly investigated until recently. Snail and Dlx-2, EMT-inducing transcription factors, are responsible for metabolic stress-induced necrosis in tumors. Snail and Dlx-2 contribute to tumor progression by promoting necrosis and inducing EMT and oncogenic metabolism. Oncogenic metabolism has been shown to play a role(s) in initiating necrosis. Here, we discuss the molecular mechanisms underlying metabolic stress-induced programmed necrosis that promote tumor progression and aggressiveness.
Collapse
|
43
|
D'Alessandro A, El Kasmi KC, Plecitá-Hlavatá L, Ježek P, Li M, Zhang H, Gupte SA, Stenmark KR. Hallmarks of Pulmonary Hypertension: Mesenchymal and Inflammatory Cell Metabolic Reprogramming. Antioxid Redox Signal 2018; 28. [PMID: 28637353 PMCID: PMC5737722 DOI: 10.1089/ars.2017.7217] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE The molecular events that promote the development of pulmonary hypertension (PH) are complex and incompletely understood. The complex interplay between the pulmonary vasculature and its immediate microenvironment involving cells of immune system (i.e., macrophages) promotes a persistent inflammatory state, pathological angiogenesis, and fibrosis that are driven by metabolic reprogramming of mesenchymal and immune cells. Recent Advancements: Consistent with previous findings in the field of cancer metabolism, increased glycolytic rates, incomplete glucose and glutamine oxidation to support anabolism and anaplerosis, altered lipid synthesis/oxidation ratios, increased one-carbon metabolism, and activation of the pentose phosphate pathway to support nucleoside synthesis are but some of the key metabolic signatures of vascular cells in PH. In addition, metabolic reprogramming of macrophages is observed in PH and is characterized by distinct features, such as the induction of specific activation or polarization states that enable their participation in the vascular remodeling process. CRITICAL ISSUES Accumulation of reducing equivalents, such as NAD(P)H in PH cells, also contributes to their altered phenotype both directly and indirectly by regulating the activity of the transcriptional co-repressor C-terminal-binding protein 1 to control the proliferative/inflammatory gene expression in resident and immune cells. Further, similar to the role of anomalous metabolism in mitochondria in cancer, in PH short-term hypoxia-dependent and long-term hypoxia-independent alterations of mitochondrial activity, in the absence of genetic mutation of key mitochondrial enzymes, have been observed and explored as potential therapeutic targets. FUTURE DIRECTIONS For the foreseeable future, short- and long-term metabolic reprogramming will become a candidate druggable target in the treatment of PH. Antioxid. Redox Signal. 28, 230-250.
Collapse
Affiliation(s)
- Angelo D'Alessandro
- 1 Department of Biochemistry and Molecular Genetics, University of Colorado - Denver , Colorado
| | - Karim C El Kasmi
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado.,3 Department of Pediatric Gastroenterology, University of Colorado - Denver , Colorado
| | - Lydie Plecitá-Hlavatá
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Ježek
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Min Li
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Hui Zhang
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Sachin A Gupte
- 5 Department of Pharmacology, School of Medicine, New York Medical College , Valhalla, New York
| | - Kurt R Stenmark
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| |
Collapse
|
44
|
Clanton R, Gonzalez A, Shankar S, Akabani G. Rapid synthesis of 125I integrated gold nanoparticles for use in combined neoplasm imaging and targeted radionuclide therapy. Appl Radiat Isot 2018; 131:49-57. [DOI: 10.1016/j.apradiso.2017.10.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 04/27/2017] [Accepted: 10/13/2017] [Indexed: 01/08/2023]
|
45
|
Lee Y, Cho W, Sung J, Kim E, Park SB. Monochromophoric Design Strategy for Tetrazine-Based Colorful Bioorthogonal Probes with a Single Fluorescent Core Skeleton. J Am Chem Soc 2017; 140:974-983. [DOI: 10.1021/jacs.7b10433] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Youngjun Lee
- CRI Center for Chemical
Proteomics, Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Wansang Cho
- CRI Center for Chemical
Proteomics, Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - June Sung
- Department
of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Eunha Kim
- Department
of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Seung Bum Park
- CRI Center for Chemical
Proteomics, Department of Chemistry, Seoul National University, Seoul 08826, Korea
- Department
of Biophysics and Chemical Biology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
46
|
Järviaho T, Hurme-Niiranen A, Soini HK, Niinimäki R, Möttönen M, Savolainen ER, Hinttala R, Harila-Saari A, Uusimaa J. Novel non-neutral mitochondrial DNA mutations found in childhood acute lymphoblastic leukemia. Clin Genet 2017; 93:275-285. [PMID: 28708239 DOI: 10.1111/cge.13100] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/29/2017] [Accepted: 07/09/2017] [Indexed: 12/18/2022]
Abstract
Mitochondria produce adenosine triphosphate (ATP) for energy requirements via the mitochondrial oxidative phosphorylation (OXPHOS) system. One of the hallmarks of cancer is the energy shift toward glycolysis. Low OXPHOS activity and increased glycolysis are associated with aggressive types of cancer. Mitochondria have their own genome (mitochondrial DNA [mtDNA]) encoding for 13 essential subunits of the OXPHOS enzyme complexes. We studied mtDNA in childhood acute lymphoblastic leukemia (ALL) to detect potential pathogenic mutations in OXPHOS complexes. The whole mtDNA from blood and bone marrow samples at diagnosis and follow-up from 36 ALL patients were analyzed. Novel or previously described pathogenic mtDNA mutations were identified in 8 out of 36 patients. Six out of these 8 patients had died from ALL. Five out of 36 patients had an identified poor prognosis genetic marker, and 4 of these patients had mtDNA mutations. Missense or nonsense mtDNA mutations were detected in the genes encoding subunits of OXPHOS complexes, as follows: MT-ND1, MT-ND2, MT-ND4L and MT-ND6 of complex I; MT-CO3 of complex IV; and MT-ATP6 and MT-ATP8 of complex V. We discovered mtDNA mutations in childhood ALL supporting the hypothesis that non-neutral variants in mtDNA affecting the OXPHOS function may be related to leukemic clones.
Collapse
Affiliation(s)
- T Järviaho
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - A Hurme-Niiranen
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - H K Soini
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - R Niinimäki
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - M Möttönen
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - E-R Savolainen
- Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.,NordLab Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.,Department of Clinical Chemistry, University of Oulu, Oulu, Finland
| | - R Hinttala
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland
| | - A Harila-Saari
- Department of Women's and Children's Health, Karolinska University Hospital Solna, Stockholm, Sweden
| | - J Uusimaa
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.,Biocenter Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
47
|
Proietti S, Cucina A, Minini M, Bizzarri M. Melatonin, mitochondria, and the cancer cell. Cell Mol Life Sci 2017; 74:4015-4025. [PMID: 28785807 PMCID: PMC11107593 DOI: 10.1007/s00018-017-2612-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/03/2017] [Indexed: 12/12/2022]
Abstract
The long-recognized fact that oxidative stress within mitochondria is a hallmark of mitochondrial dysfunction has stimulated the development of mitochondria-targeted antioxidant therapies. Melatonin should be included among the pharmacological agents able to modulate mitochondrial functions in cancer, given that a number of relevant melatonin-dependent effects are triggered by targeting mitochondrial functions. Indeed, melatonin may modulate the mitochondrial respiratory chain, thus antagonizing the cancer highly glycolytic bioenergetic pathway of cancer cells. Modulation of the mitochondrial respiratory chain, together with Ca2+ release and mitochondrial apoptotic effectors, may enhance the spontaneous or drug-induced apoptotic processes. Given that melatonin may efficiently counteract the Warburg effect while stimulating mitochondrial differentiation and mitochondrial-based apoptosis, it is argued that the pineal neurohormone could represent a promising new perspective in cancer treatment strategy.
Collapse
Affiliation(s)
- Sara Proietti
- Department of Surgery, "Pietro Valdoni", Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy
| | - Alessandra Cucina
- Department of Surgery, "Pietro Valdoni", Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy
| | - Mirko Minini
- Department of Surgery, "Pietro Valdoni", Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
48
|
Lee KM, Giltnane JM, Balko JM, Schwarz LJ, Guerrero-Zotano AL, Hutchinson KE, Nixon MJ, Estrada MV, Sánchez V, Sanders ME, Lee T, Gómez H, Lluch A, Pérez-Fidalgo JA, Wolf MM, Andrejeva G, Rathmell JC, Fesik SW, Arteaga CL. MYC and MCL1 Cooperatively Promote Chemotherapy-Resistant Breast Cancer Stem Cells via Regulation of Mitochondrial Oxidative Phosphorylation. Cell Metab 2017; 26:633-647.e7. [PMID: 28978427 PMCID: PMC5650077 DOI: 10.1016/j.cmet.2017.09.009] [Citation(s) in RCA: 454] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/06/2017] [Accepted: 09/15/2017] [Indexed: 12/21/2022]
Abstract
Most patients with advanced triple-negative breast cancer (TNBC) develop drug resistance. MYC and MCL1 are frequently co-amplified in drug-resistant TNBC after neoadjuvant chemotherapy. Herein, we demonstrate that MYC and MCL1 cooperate in the maintenance of chemotherapy-resistant cancer stem cells (CSCs) in TNBC. MYC and MCL1 increased mitochondrial oxidative phosphorylation (mtOXPHOS) and the generation of reactive oxygen species (ROS), processes involved in maintenance of CSCs. A mutant of MCL1 that cannot localize in mitochondria reduced mtOXPHOS, ROS levels, and drug-resistant CSCs without affecting the anti-apoptotic function of MCL1. Increased levels of ROS, a by-product of activated mtOXPHOS, led to the accumulation of HIF-1α. Pharmacological inhibition of HIF-1α attenuated CSC enrichment and tumor initiation in vivo. These data suggest that (1) MYC and MCL1 confer resistance to chemotherapy by expanding CSCs via mtOXPHOS and (2) targeting mitochondrial respiration and HIF-1α may reverse chemotherapy resistance in TNBC.
Collapse
Affiliation(s)
- Kyung-Min Lee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jennifer M Giltnane
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Luis J Schwarz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | - Mellissa J Nixon
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mónica V Estrada
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Violeta Sánchez
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Melinda E Sanders
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Taekyu Lee
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Henry Gómez
- Instituto Nacional de Enfermedades Neoplásicas, 15038 Lima, Perú
| | - Ana Lluch
- Hospital Clínico Universitario, Biomedical Research Institute INCLIVA, Universidad de Valencia, 46010 Valencia, Spain
| | - J Alejandro Pérez-Fidalgo
- Hospital Clínico Universitario, Biomedical Research Institute INCLIVA, Universidad de Valencia, 46010 Valencia, Spain
| | - Melissa Magdalene Wolf
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gabriela Andrejeva
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Stephen W Fesik
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Carlos L Arteaga
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
49
|
Aung LH, Li R, Prabhakar BS, Maker AV, Li P. Mitochondrial protein 18 (MTP18) plays a pro-apoptotic role in chemotherapy-induced gastric cancer cell apoptosis. Oncotarget 2017; 8:56582-56597. [PMID: 28915614 PMCID: PMC5593585 DOI: 10.18632/oncotarget.17508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023] Open
Abstract
One of the severe limitations of chemotherapy is the development of drug resistance. However, the mechanisms underlying chemotherapy resistance remain to be elucidated. Mitochondrial mediated apoptosis is a form of cell death induced by chemotherapy. Several chemotherapeutic agents have been shown to induce mitochondrial fission, and finally activate the apoptosis cascade in various cancer cells. Here, we report that the mitochondrial membrane protein 18 (MTP18) induced mitochondrial fragmentation in gastric cancer cells under doxorubicin (DOX) exposure. Upon over-expression of MTP18, a sub-cytotoxic dose of DOX could sensitize a significant number of cells to undergo mitochondrial fission and subsequent apoptosis. These findings suggest that MTP18 can enhance the sensitivity of gastric cancer cells to DOX. Mechanistically, we found that MTP18 enriched dynamic-related protein 1 (DRP1) accumulation in mitochondria and it was responsible for mediating DOX-induced signaling required for mitochondrial fission. Intriguingly, MTP18 expression was downregulated during DOX treatment. Thus, down-regulation of MTP18 expression could be one of the resistance factors interfering with DOX-induced apoptosis in gastric cancer cells.
Collapse
Affiliation(s)
- Lynn H.H. Aung
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ruibei Li
- School of Professional Studies, Northwestern University, Chicago, IL, USA
| | - Bellur S. Prabhakar
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ajay V. Maker
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, Creticos Cancer Center, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Peifeng Li
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
50
|
Beckervordersandforth R, Ebert B, Schäffner I, Moss J, Fiebig C, Shin J, Moore DL, Ghosh L, Trinchero MF, Stockburger C, Friedland K, Steib K, von Wittgenstein J, Keiner S, Redecker C, Hölter SM, Xiang W, Wurst W, Jagasia R, Schinder AF, Ming GL, Toni N, Jessberger S, Song H, Lie DC. Role of Mitochondrial Metabolism in the Control of Early Lineage Progression and Aging Phenotypes in Adult Hippocampal Neurogenesis. Neuron 2017; 93:560-573.e6. [PMID: 28111078 DOI: 10.1016/j.neuron.2016.12.017] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 08/06/2016] [Accepted: 11/23/2016] [Indexed: 12/20/2022]
Abstract
Precise regulation of cellular metabolism is hypothesized to constitute a vital component of the developmental sequence underlying the life-long generation of hippocampal neurons from quiescent neural stem cells (NSCs). The identity of stage-specific metabolic programs and their impact on adult neurogenesis are largely unknown. We show that the adult hippocampal neurogenic lineage is critically dependent on the mitochondrial electron transport chain and oxidative phosphorylation machinery at the stage of the fast proliferating intermediate progenitor cell. Perturbation of mitochondrial complex function by ablation of the mitochondrial transcription factor A (Tfam) reproduces multiple hallmarks of aging in hippocampal neurogenesis, whereas pharmacological enhancement of mitochondrial function ameliorates age-associated neurogenesis defects. Together with the finding of age-associated alterations in mitochondrial function and morphology in NSCs, these data link mitochondrial complex function to efficient lineage progression of adult NSCs and identify mitochondrial function as a potential target to ameliorate neurogenesis-defects in the aging hippocampus.
Collapse
Affiliation(s)
- Ruth Beckervordersandforth
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Birgit Ebert
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Iris Schäffner
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jonathan Moss
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Christian Fiebig
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jaehoon Shin
- Institute for Cell Engineering, Department of Neurology, The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Darcie L Moore
- Brain Research Institute, Faculty of Medicine and Science, University of Zurich, 8057 Zurich, Switzerland
| | - Laboni Ghosh
- Brain Research Institute, Faculty of Medicine and Science, University of Zurich, 8057 Zurich, Switzerland
| | - Mariela F Trinchero
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA, CONICET), C1405BWE Buenos Aires, Argentina
| | - Carola Stockburger
- Molecular and Clinical Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kristina Friedland
- Molecular and Clinical Pharmacy, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kathrin Steib
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Julia von Wittgenstein
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Silke Keiner
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Christoph Redecker
- Hans Berger Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Sabine M Hölter
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Wei Xiang
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany
| | - Ravi Jagasia
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich-Neuherberg, Germany; F. Hoffmann-La Roche Ltd, CNS Discovery; Pharma Research and Early Development, 4070 Basel, Switzerland
| | - Alejandro F Schinder
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA, CONICET), C1405BWE Buenos Aires, Argentina
| | - Guo-Li Ming
- Institute for Cell Engineering, Department of Neurology, The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicolas Toni
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Sebastian Jessberger
- Brain Research Institute, Faculty of Medicine and Science, University of Zurich, 8057 Zurich, Switzerland
| | - Hongjun Song
- Institute for Cell Engineering, Department of Neurology, The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - D Chichung Lie
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|