1
|
Chen J, Sun Q, Wang Y, Yin W. Revealing the key role of cuproptosis in osteoporosis via the bioinformatic analysis and experimental validation of cuproptosis-related genes. Mamm Genome 2024; 35:414-431. [PMID: 38904833 DOI: 10.1007/s00335-024-10049-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/14/2024] [Indexed: 06/22/2024]
Abstract
The incidence of osteoporosis has rapidly increased owing to the ageing population. Cuproptosis, a novel mechanism that regulates cell death, may be a new therapeutic approach. However, the relevance of cuproptosis in the immune microenvironment and osteoporosis immunotherapy is still unknown. We intersected the differentially expressed genes from osteoporotic samples with 75 cuproptosis-related genes to identify 16 significantly expressed cuproptosis genes. We further explored the connection between the cuproptosis pattern, immune microenvironment, and immunotherapy. The weighted gene co-expression network analysis algorithm was used to identify cuproptosis phenotype-associated genes, and we used quantitative real-time PCR and immunohistochemistry in mouse femur tissues to verify hub gene (MAP2K2, FDX1, COX19, VEGFA, CDKN2A, and NFE2L2) expression. Six hub genes and 59 cuproptosis phenotype-associated genes involved in immunisation were identified among the osteoporosis and control groups, and the majority of these 59 genes were enriched in the inflammatory response, as well as in signal transducers, Janus kinase, and transcription pathway activators. In addition, two different clusters of cuproptosis were found, and immune infiltration analysis showed that gene Cluster 1 had a greater immune score and immune infiltration level. Further analysis revealed that three key genes (COX19, MAP2K2, and FDX1) were highly correlated with immune cell infiltration, and external experiments validated the association of these three genes with the prognosis of osteoporosis. We used the three key mRNAs COX19, MAP2K2, and FDX1 as a classification model that may systematically elucidate the complex connection between cuproptosis and the immune microenvironment of osteoporosis. New insights into osteoporosis pathogenesis and immunotherapy prospects may be gained from this study.
Collapse
Affiliation(s)
- Jianxing Chen
- Department of Joint Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Qifeng Sun
- Department of Joint Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Yi Wang
- Department of Joint Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Wenzhe Yin
- Department of Joint Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
2
|
Aguilar K, Jakubek P, Zorzano A, Wieckowski MR. Primary mitochondrial diseases: The intertwined pathophysiology of bioenergetic dysregulation, oxidative stress and neuroinflammation. Eur J Clin Invest 2024; 54:e14217. [PMID: 38644687 DOI: 10.1111/eci.14217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/23/2024]
Abstract
OBJECTIVES AND SCOPE Primary mitochondrial diseases (PMDs) are rare genetic disorders resulting from mutations in genes crucial for effective oxidative phosphorylation (OXPHOS) that can affect mitochondrial function. In this review, we examine the bioenergetic alterations and oxidative stress observed in cellular models of primary mitochondrial diseases (PMDs), shedding light on the intricate complexity between mitochondrial dysfunction and cellular pathology. We explore the diverse cellular models utilized to study PMDs, including patient-derived fibroblasts, induced pluripotent stem cells (iPSCs) and cybrids. Moreover, we also emphasize the connection between oxidative stress and neuroinflammation. INSIGHTS The central nervous system (CNS) is particularly vulnerable to mitochondrial dysfunction due to its dependence on aerobic metabolism and the correct functioning of OXPHOS. Similar to other neurodegenerative diseases affecting the CNS, individuals with PMDs exhibit several neuroinflammatory hallmarks alongside neurodegeneration, a pattern also extensively observed in mouse models of mitochondrial diseases. Based on histopathological analysis of postmortem human brain tissue and findings in mouse models of PMDs, we posit that neuroinflammation is not merely a consequence of neurodegeneration but a potential pathogenic mechanism for disease progression that deserves further investigation. This recognition may pave the way for novel therapeutic strategies for this group of devastating diseases that currently lack effective treatments. SUMMARY In summary, this review provides a comprehensive overview of bioenergetic alterations and redox imbalance in cellular models of PMDs while underscoring the significance of neuroinflammation as a potential driver in disease progression.
Collapse
Affiliation(s)
- Kevin Aguilar
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Patrycja Jakubek
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| |
Collapse
|
3
|
Povea-Cabello S, Brischigliaro M, Fernández-Vizarra E. Emerging mechanisms in the redox regulation of mitochondrial cytochrome c oxidase assembly and function. Biochem Soc Trans 2024; 52:873-885. [PMID: 38526156 DOI: 10.1042/bst20231183] [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] [Received: 01/30/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
In eukaryotic cells, mitochondria perform cellular respiration through a series of redox reactions ultimately reducing molecular oxygen to water. The system responsible for this process is the respiratory chain or electron transport system (ETS) composed of complexes I-IV. Due to its function, the ETS is the main source of reactive oxygen species (ROS), generating them on both sides of the mitochondrial inner membrane, i.e. the intermembrane space (IMS) and the matrix. A correct balance between ROS generation and scavenging is important for keeping the cellular redox homeostasis and other important aspects of cellular physiology. However, ROS generated in the mitochondria are important signaling molecules regulating mitochondrial biogenesis and function. The IMS contains a large number of redox sensing proteins, containing specific Cys-rich domains, that are involved in ETS complex biogenesis. The large majority of these proteins function as cytochrome c oxidase (COX) assembly factors, mainly for the handling of copper ions necessary for the formation of the redox reactive catalytic centers. A particular case of ROS-regulated COX assembly factor is COA8, whose intramitochondrial levels are increased by oxidative stress, promoting COX assembly and/or protecting the enzyme from oxidative damage. In this review, we will discuss the current knowledge concerning the role played by ROS in regulating mitochondrial activity and biogenesis, focusing on the COX enzyme and with a special emphasis on the functional role exerted by the redox sensitive Cys residues contained in the COX assembly factors.
Collapse
Affiliation(s)
- Suleva Povea-Cabello
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Michele Brischigliaro
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Erika Fernández-Vizarra
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| |
Collapse
|
4
|
Gao S, Zhang H, Zhang X, Wang J, Bai W, Jiang B. COX19 Is a New Target of MACC1 and Promotes Colorectal Cancer Progression by Regulating Copper Transport in Mitochondria. J Nutr 2024; 154:381-394. [PMID: 38141772 DOI: 10.1016/j.tjnut.2023.12.032] [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] [Received: 10/08/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND Recent studies have demonstrated that copper (Cu) plays an important role in the progression of tumor diseases. Metastasis associated with colon cancer protein 1 (MACC1) promotes the transcription and expression of various tumor-related genes. Cytochrome c oxidase (COX) 19, present in the cytoplasm and intermembrane space of mitochondria, may transport Cu within the mitochondria. However, the mechanism through which MACC1 regulates the Cu homeostasis mediated by COX19 remains unclear. OBJECTIVES The aim of this study was to elucidate the mechanism through which MACC1 initiates the transcription and expression of COX19, and promotes malignant behavior in tumor cells. METHODS Immunohistochemistry, western blotting, and real-time polymerase chain reaction (PCR) analyses were conducted to analyze the expression of MACC1 and COX19 proteins and genes in tumor and normal tissues. RNA-chromatin immunoprecipitation was used to detect the transcriptional initiation of COX19 by MACC1. The effects of MACC1 and COX19 on mitochondrial activity were determined using an ATP assay kit and Cytochrome c Oxidase Assay Kit. A Cell Counting Kit-8 kit was used to detect the effect of high-dose Cu or overexpression of MACC1 and COX19 on tumor cell proliferation. A xenograft mouse model was used to analyze the effect of the COX19 overexpression on the malignant behavior of the tumors. RESULTS Cu enhanced the proliferation, invasion, and migration and inhibited apoptosis of SW480 cells. MACC1 was highly expressed in colorectal cancer tissues and activated the expression of COX19 by binding to its promoter region of COX19. The overexpression of COX19 increased mitochondrial Cu content and enhanced the activity of mitochondrial COX and ATP content, and inhibited apoptosis, promoted tumor growth of mice. CONCLUSIONS Our results indicate that COX19 functions as a target gene of MACC1 and regulates mitochondrial activity and promotes the progression of colorectal cancer. MACC1/COX19 may provide a novel therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Sheng Gao
- Department of Colorectal and Anal Surgery, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, P.R. China.
| | - Hong Zhang
- Medical Department, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, P.R. China.
| | - Xiaodong Zhang
- Department of Colorectal and Anal Surgery, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, P.R. China.
| | - Jian Wang
- Department of General Surgery, Bethune Hospital of Shanxi Province, Taiyuan 030000, P.R. China
| | - Wenqi Bai
- Department of Colorectal and Anal Surgery, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, P.R. China
| | - Bo Jiang
- Department of Colorectal and Anal Surgery, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, P.R. China
| |
Collapse
|
5
|
Mandatori S, Liu Y, Marturia-Navarro J, Hadi M, Henriksen K, Zheng J, Rasmussen LM, Rizza S, Kaestner KH, Issazadeh-Navikas S. PRKAG2.2 is essential for FoxA1 + regulatory T cell differentiation and metabolic rewiring distinct from FoxP3 + regulatory T cells. SCIENCE ADVANCES 2023; 9:eadj8442. [PMID: 38117896 PMCID: PMC10732530 DOI: 10.1126/sciadv.adj8442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/20/2023] [Indexed: 12/22/2023]
Abstract
Forkhead box A1 (FoxA1)+ regulatory T cells (Tregs) exhibit distinct characteristics from FoxP3+ Tregs while equally effective in exerting anti-inflammatory properties. The role of FoxP3+ Tregs in vivo has been challenged, motivating a better understanding of other Tregs in modulating hyperactive immune responses. FoxA1+ Tregs are generated on activation of the transcription factor FoxA1 by interferon-β (IFNβ), an anti-inflammatory cytokine. T cell activation, expansion, and function hinge on metabolic adaptability. We demonstrated that IFNβ promotes a metabolic rearrangement of FoxA1+ Tregs by enhancing oxidative phosphorylation and mitochondria clearance by mitophagy. In response to IFNβ, FoxA1 induces a specific transcription variant of adenosine 5'-monophosphate-activated protein kinase (AMPK) γ2 subunit, PRKAG2.2. This leads to the activation of AMPK signaling, thereby enhancing mitochondrial respiration and mitophagy by ULK1-BNIP3. This IFNβ-FoxA1-PRKAG2.2-BNIP3 axis is pivotal for their suppressive function. The involvement of PRKAG2.2 in FoxA1+ Treg, not FoxP3+ Treg differentiation, underscores the metabolic differences between Treg populations and suggests potential therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Sara Mandatori
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yawei Liu
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joana Marturia-Navarro
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mahdieh Hadi
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Henriksen
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jin Zheng
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Louise Munk Rasmussen
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Salvatore Rizza
- Redox Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Klaus H. Kaestner
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Sneha NP, Dharshini SAP, Taguchi YH, Gromiha MM. Investigating Neuron Degeneration in Huntington's Disease Using RNA-Seq Based Transcriptome Study. Genes (Basel) 2023; 14:1801. [PMID: 37761940 PMCID: PMC10530489 DOI: 10.3390/genes14091801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/02/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused due to a CAG repeat expansion in the huntingtin (HTT) gene. The primary symptoms of HD include motor dysfunction such as chorea, dystonia, and involuntary movements. The primary motor cortex (BA4) is the key brain region responsible for executing motor/movement activities. Investigating patient and control samples from the BA4 region will provide a deeper understanding of the genes responsible for neuron degeneration and help to identify potential markers. Previous studies have focused on overall differential gene expression and associated biological functions. In this study, we illustrate the relationship between variants and differentially expressed genes/transcripts. We identified variants and their associated genes along with the quantification of genes and transcripts. We also predicted the effect of variants on various regulatory activities and found that many variants are regulating gene expression. Variants affecting miRNA and its targets are also highlighted in our study. Co-expression network studies revealed the role of novel genes. Function interaction network analysis unveiled the importance of genes involved in vesicle-mediated transport. From this unified approach, we propose that genes expressed in immune cells are crucial for reducing neuron death in HD.
Collapse
Affiliation(s)
- Nela Pragathi Sneha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (N.P.S.); (S.A.P.D.)
| | - S. Akila Parvathy Dharshini
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (N.P.S.); (S.A.P.D.)
| | - Y.-h. Taguchi
- Department of Physics, Chuo University, Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan;
| | - M. Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (N.P.S.); (S.A.P.D.)
| |
Collapse
|
7
|
Salman AA, Goldring JPD. Expression and copper binding studies of a Plasmodium falciparum protein with Cox19 copper binding motifs. Exp Parasitol 2023:108572. [PMID: 37348640 DOI: 10.1016/j.exppara.2023.108572] [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: 01/26/2023] [Revised: 05/25/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Copper can exist in an oxidized and a reduced form, which enables the metal to play essential roles as a catalytic co-factor in redox reactions in many organisms. Copper confers redox activity to the terminal electron transport chain cytochrome c oxidase protein. Cytochrome c oxidase in yeast obtains copper for the CuB site in the Cox1 subunit from Cox11 in association with Cox19. When copper is chelated in growth medium, Plasmodium falciparum parasite development in infected red blood cells is inhibited and excess copper is toxic for the parasite. The gene of a 26 kDa Plasmodium falciparum PfCox19 protein with two Cx9C Cox19 copper binding motifs, was cloned and expressed as a 66 kDa fusion protein with maltose binding protein and affinity purified (rMBP-PfCox19). rMBP-PfCox19 bound copper measured by: a bicinchoninic acid release assay; an in vivo bacterial host growth inhibition assay; ascorbate oxidation inhibition and differential scanning fluorimetry. The native protein was detected by antibodies raised against rMBP-PfCox19. PfCox19 binds copper and is predicted to associate with PfCox11 in the insertion of copper into the CuB site of Plasmodium cytochrome c oxidase. Characterisation of the proteins involved in Plasmodium spp. copper metabolism will help us understand the role of cytochrome c oxidase and this essential metal in Plasmodium homeostasis.
Collapse
Affiliation(s)
| | - J P Dean Goldring
- Biochemistry, University of KwaZulu-Natal, Pietermaritzburg, 3201, South Africa.
| |
Collapse
|
8
|
Wynne ME, Ogunbona O, Lane AR, Gokhale A, Zlatic SA, Xu C, Wen Z, Duong DM, Rayaprolu S, Ivanova A, Ortlund EA, Dammer EB, Seyfried NT, Roberts BR, Crocker A, Shanbhag V, Petris M, Senoo N, Kandasamy S, Claypool SM, Barrientos A, Wingo A, Wingo TS, Rangaraju S, Levey AI, Werner E, Faundez V. APOE expression and secretion are modulated by mitochondrial dysfunction. eLife 2023; 12:e85779. [PMID: 37171075 PMCID: PMC10231934 DOI: 10.7554/elife.85779] [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: 12/23/2022] [Accepted: 05/11/2023] [Indexed: 05/13/2023] Open
Abstract
Mitochondria influence cellular function through both cell-autonomous and non-cell autonomous mechanisms, such as production of paracrine and endocrine factors. Here, we demonstrate that mitochondrial regulation of the secretome is more extensive than previously appreciated, as both genetic and pharmacological disruption of the electron transport chain caused upregulation of the Alzheimer's disease risk factor apolipoprotein E (APOE) and other secretome components. Indirect disruption of the electron transport chain by gene editing of SLC25A mitochondrial membrane transporters as well as direct genetic and pharmacological disruption of either complexes I, III, or the copper-containing complex IV of the electron transport chain elicited upregulation of APOE transcript, protein, and secretion, up to 49-fold. These APOE phenotypes were robustly expressed in diverse cell types and iPSC-derived human astrocytes as part of an inflammatory gene expression program. Moreover, age- and genotype-dependent decline in brain levels of respiratory complex I preceded an increase in APOE in the 5xFAD mouse model. We propose that mitochondria act as novel upstream regulators of APOE-dependent cellular processes in health and disease.
Collapse
Affiliation(s)
- Meghan E Wynne
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Oluwaseun Ogunbona
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Department of Pathology and Laboratory Medicine, Emory UniversityAtlantaUnited States
| | - Alicia R Lane
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Avanti Gokhale
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | | | - Chongchong Xu
- Department of Psychiatry and Behavioral Sciences, Emory UniversityAtlantaUnited States
| | - Zhexing Wen
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Department of Psychiatry and Behavioral Sciences, Emory UniversityAtlantaUnited States
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Duc M Duong
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Sruti Rayaprolu
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Anna Ivanova
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Eric A Ortlund
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Eric B Dammer
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | | | - Blaine R Roberts
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Amanda Crocker
- Program in Neuroscience, Middlebury CollegeMiddleburyUnited States
| | - Vinit Shanbhag
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Michael Petris
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Nanami Senoo
- Department of Physiology, Johns Hopkins UniversityBaltimoreUnited States
| | | | | | - Antoni Barrientos
- Department of Neurology and Biochemistry & Molecular Biology, University of MiamiMiamiUnited States
| | - Aliza Wingo
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Thomas S Wingo
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Srikant Rangaraju
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Allan I Levey
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Erica Werner
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Victor Faundez
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| |
Collapse
|
9
|
Xie J, Yang Y, Gao Y, He J. Cuproptosis: mechanisms and links with cancers. Mol Cancer 2023; 22:46. [PMID: 36882769 PMCID: PMC9990368 DOI: 10.1186/s12943-023-01732-y] [Citation(s) in RCA: 261] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/25/2023] [Indexed: 03/09/2023] Open
Abstract
Cuproptosis was a copper-dependent and unique kind of cell death that was separate from existing other forms of cell death. The last decade has witnessed a considerable increase in investigations of programmed cell death, and whether copper induced cell death was an independent form of cell death has long been argued until mechanism of cuproptosis has been revealed. After that, increasing number of researchers attempted to identify the relationship between cuproptosis and the process of cancer. Thus, in this review, we systematically detailed the systemic and cellular metabolic processes of copper and the copper-related tumor signaling pathways. Moreover, we not only focus on the discovery process of cuproptosis and its mechanism, but also outline the association between cuproptosis and cancers. Finally, we further highlight the possible therapeutic direction of employing copper ion ionophores with cuproptosis-inducing functions in combination with small molecule drugs for targeted therapy to treat specific cancers.
Collapse
Affiliation(s)
- Jiaming Xie
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yannan Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yibo Gao
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China. .,Laboratory of Translational Medicine, National Cancer Center/National, Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 101399, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,Laboratory of Translational Medicine, National Cancer Center/National, Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 101399, China.
| |
Collapse
|
10
|
Jett KA, Baker ZN, Hossain A, Boulet A, Cobine PA, Ghosh S, Ng P, Yilmaz O, Barreto K, DeCoteau J, Mochoruk K, Ioannou GN, Savard C, Yuan S, Abdalla OH, Lowden C, Kim BE, Cheng HYM, Battersby BJ, Gohil VM, Leary SC. Mitochondrial dysfunction reactivates α-fetoprotein expression that drives copper-dependent immunosuppression in mitochondrial disease models. J Clin Invest 2023; 133:e154684. [PMID: 36301669 PMCID: PMC9797342 DOI: 10.1172/jci154684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/26/2022] [Indexed: 02/04/2023] Open
Abstract
Signaling circuits crucial to systemic physiology are widespread, yet uncovering their molecular underpinnings remains a barrier to understanding the etiology of many metabolic disorders. Here, we identified a copper-linked signaling circuit activated by disruption of mitochondrial function in the murine liver or heart that resulted in atrophy of the spleen and thymus and caused a peripheral white blood cell deficiency. We demonstrated that the leukopenia was caused by α-fetoprotein, which required copper and the cell surface receptor CCR5 to promote white blood cell death. We further showed that α-fetoprotein expression was upregulated in several cell types upon inhibition of oxidative phosphorylation. Collectively, our data argue that α-fetoprotein may be secreted by bioenergetically stressed tissue to suppress the immune system, an effect that may explain the recurrent or chronic infections that are observed in a subset of mitochondrial diseases or in other disorders with secondary mitochondrial dysfunction.
Collapse
Affiliation(s)
- Kimberly A. Jett
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Zakery N. Baker
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Amzad Hossain
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Aren Boulet
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Paul A. Cobine
- Department of Biological Sciences, Auburn University, Auburn, Alabama, USA
| | - Sagnika Ghosh
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Philip Ng
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Orhan Yilmaz
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| | - Kris Barreto
- Department of Laboratory and Pathology Medicine, University of Saskatchewan, Saskatoon, Canada
| | - John DeCoteau
- Department of Laboratory and Pathology Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Karen Mochoruk
- Department of Laboratory and Pathology Medicine, University of Saskatchewan, Saskatoon, Canada
| | - George N. Ioannou
- Division of Gastroenterology
- Research and Development, Veterans Affairs Puget Sound Health Care System and the
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Christopher Savard
- Division of Gastroenterology
- Research and Development, Veterans Affairs Puget Sound Health Care System and the
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Sai Yuan
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
| | - Osama H.M.H. Abdalla
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Christopher Lowden
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Byung-Eun Kim
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | | | - Vishal M. Gohil
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Scot C. Leary
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
11
|
Malcomson T, Repiščák P, Erhardt S, Paterson MJ. Protocols for Understanding the Redox Behavior of Copper-Containing Systems. ACS OMEGA 2022; 7:45057-45066. [PMID: 36530299 PMCID: PMC9753522 DOI: 10.1021/acsomega.2c05484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/09/2022] [Indexed: 06/17/2023]
Abstract
Suitability of single-reference density functional theory (DFT) methods for the calculation of redox potentials of copper-containing macrocycle complexes was confirmed by the use of T 1 diagnostics along with a verification of negligible spin contamination or wave function instability. When examining the effect of improvement in the cc-pVnZ basis set series on calculated redox potentials, the results readily converged at the cc-pVTZ level. The all-electron Def2-TZVPP basis set is shown to be a suitable choice of a basis set for the calculation of redox potentials when utilizing a cc-pVTZ geometry. The best-performing model chemistries are determined to be the M06/polarizable continuum model (PCM); therefore, a scheme for redox potential calculations of copper macrocycles using either M06/cc-pVTZ with PCM solvation is proposed to reliably reproduce experimental trends.
Collapse
Affiliation(s)
- Thomas Malcomson
- Department
of Chemistry, School of Natural Sciences, The University of Manchester, ManchesterM13 9PL, U.K.
| | - Peter Repiščák
- Beatson
Institute for Cancer Research, University
of Glasgow, Garscube Estate Switchback Road, BearsdenG61 1QH, U.K.
| | - Stefan Erhardt
- School
of Applied Sciences, Edinburgh Napier University, EdinburghEH11 4BN, Scotland, U.K.
| | - Martin J. Paterson
- Institute
of Chemical Sciences, School of Engineering and Physical Sciences, Heriot-Watt University, EdinburghEH14 4AS, U.K.
| |
Collapse
|
12
|
Zheng Q, Yang R, Ni X, Yang S, Xiong L, Yan D, Xia L, Yuan J, Wang J, Jiao P, Wu J, Hao Y, Wang J, Guo L, Jiang Z, Wang L, Chen Z, Liu X. Accurate Diagnosis and Survival Prediction of Bladder Cancer Using Deep Learning on Histological Slides. Cancers (Basel) 2022; 14:cancers14235807. [PMID: 36497289 PMCID: PMC9737237 DOI: 10.3390/cancers14235807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
(1) Background: Early diagnosis and treatment are essential to reduce the mortality rate of bladder cancer (BLCA). We aimed to develop deep learning (DL)-based weakly supervised models for the diagnosis of BLCA and prediction of overall survival (OS) in muscle-invasive bladder cancer (MIBC) patients using whole slide digitized histological images (WSIs). (2) Methods: Diagnostic and prognostic models were developed using 926 WSIs of 412 BLCA patients from The Cancer Genome Atlas cohort. We collected 250 WSIs of 150 BLCA patients from the Renmin Hospital of Wuhan University cohort for external validation of the models. Two DL models were developed: a BLCA diagnostic model (named BlcaMIL) and an MIBC prognostic model (named MibcMLP). (3) Results: The BlcaMIL model identified BLCA with accuracy 0.987 in the external validation set, comparable to that of expert uropathologists and outperforming a junior pathologist. The C-index values for the MibcMLP model on the internal and external validation sets were 0.631 and 0.622, respectively. The risk score predicted by MibcMLP was a strong predictor independent of existing clinical or histopathologic indicators, as demonstrated by univariate Cox (HR = 2.390, p < 0.0001) and multivariate Cox (HR = 2.414, p < 0.0001) analyses. The interpretability of DL models can help in the analysis of critical regions associated with tumors to enrich the information obtained from WSIs. Furthermore, the expression of six genes (ANAPC7, MAPKAPK5, COX19, LINC01106, AL161431.1 and MYO16-AS1) was significantly associated with MibcMLP-predicted risk scores, revealing possible potential biological correlations. (4) Conclusions: Our study developed DL models for accurately diagnosing BLCA and predicting OS in MIBC patients, which will help promote the precise pathological diagnosis of BLCA and risk stratification of MIBC to improve clinical treatment decisions.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xinmiao Ni
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Song Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lin Xiong
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Dandan Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lingli Xia
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingsong Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Panpan Jiao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiejun Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jianguo Wang
- Department of Hepatic-Biliary-Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Liantao Guo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhengyu Jiang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (Z.C.); (X.L.)
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (Z.C.); (X.L.)
| |
Collapse
|
13
|
Liu H, Tang T. Pan-cancer genetic analysis of cuproptosis and copper metabolism-related gene set. Front Oncol 2022; 12:952290. [PMID: 36276096 PMCID: PMC9582932 DOI: 10.3389/fonc.2022.952290] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundA recent paper has revealed a novel cell death pathway, cuproptosis, a programmed cell death based on copper. This study aimed to evaluate the pan-cancer genomics and clinical association of cuproptosis and copper metabolism-related cell death genes, including SLC25A3, SLC25A37, SLC31A1, FDX1, DLAT, LIAS, ATP7A, ATP7B, COX17, SCO1, SCO2, COX11, and COX19.MethodsBy mining multi-omics profiling data, we performed a comprehensive and systematic characterization of cuproptosis genes across more than 9,000 samples of over 30 types of cancer.ResultsATP7B and ATP7A were the two most frequently mutated copper cell death genes in cancer. UCEC and SKCM were the two cancer types that have the highest mutation rates while the mutation of LIAS was associated with worse survival of BRCA. Brain cancer was potentially affected by copper cell death because of the difference in copper cell death gene expression among subtypes and stages. On the contrary, KIRC might have a lower cuproptosis activity because of the decrease in copper cell death gene expression. In lung cancer and kidney cancer, most of the cancer–noncancer expression patterns of copper cell death genes were consistent between mRNA and protein levels. Some of the cuproptosis gene expression was associated with the survival of LGG, KIRC, and ACC. The top five expression-copy numbers correlating cancer types were BRCA, OV, LUSC, HNSC, BLCA, and LUAD. Generally, the copy number variations of these genes in KIRC, UCEC, and LGG were associated with survival. The expression of DLAT, LIAS, and ATP7B was negatively correlated with the methylation in most of the cancer types. The copper cell death genes regulating miRNA and pathway regulation networks were constructed. The copper cell death genes were correlated with immune cell infiltration levels of multiple immune cells. These genes were correlated with the sensitivity of cancer cells to multiple drugs.ConclusionCopper cell death genes are potentially involved in many cancer types and can be developed as candidates for cancer diagnosis, prognosis, and therapeutic biomarkers.
Collapse
Affiliation(s)
| | - Tao Tang
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Tao Tang,
| |
Collapse
|
14
|
Tang S, Zhao L, Wu XB, Wang Z, Cai LY, Pan D, Li Y, Zhou Y, Shen Y. Identification of a Novel Cuproptosis-Related Gene Signature for Prognostic Implication in Head and Neck Squamous Carcinomas. Cancers (Basel) 2022; 14:cancers14163986. [PMID: 36010978 PMCID: PMC9406337 DOI: 10.3390/cancers14163986] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 12/08/2022] Open
Abstract
Simple Summary Head and neck squamous carcinoma (HNSC) is a common malignancy that requires novel therapeutic targets. Cuproptosis is an emerging research hotspot. The purpose of this study is to mine the cuproptosis-related genes to find prognosis-related genes. We successfully identified a 24-gene signature for predicting overall survival (OS) in HNSC patients and may expand the range of potential targets for treating HNSC. Abstract Head and neck squamous carcinoma (HNSC) is a frequent and deadly malignancy that is challenging to manage. The existing treatment options have considerable efficacy limitations. Hence, the identification of new therapeutic targets and the development of efficacious treatments are urgent needs. Cuproptosis, a non-apoptotic programmed cell death caused by excess copper, has only very recently been discovered. The present study investigated the prognostic importance of genes involved in cuproptosis through the mRNA expression data and related clinical information of HNSC patients downloaded from public databases. Our results revealed that many cuproptosis-related genes were differentially expressed between normal and HNSC tissues in the TCGA cohort. Moreover, 39 differentially expressed genes were associated with the prognosis of HNSC patients. Then, a 24-gene signature was identified in the TCGA cohort utilizing the LASSO Cox regression model. HNSC expression data used for validation were obtained from the GEO database. Consequently, we divided patients into high- and low-risk groups based on the 24-gene signature. Furthermore, we demonstrated that the high-risk group had a worse prognosis when compared to the low-risk group. Additionally, significant differences were found between the two groups in metabolic pathways, immune microenvironment, etc. In conclusion, we found a cuproptosis-related gene signature that can be used effectively to predict OS in HNSC patients. Thus, targeting cuproptosis might be an alternative and promising strategy for HNSC patients.
Collapse
Affiliation(s)
- Shouyi Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
| | - Li Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
| | - Xing-Bo Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
| | - Zhen Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
| | - Lu-Yao Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
| | - Dan Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
| | - Ying Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
- State Institute of Drug/Medical Device Clinical Trial, West China Hospital of Stomatology, Chengdu 610041, China
- Correspondence: (Y.Z.); (Y.S.)
| | - Yingqiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 640041, China
- Correspondence: (Y.Z.); (Y.S.)
| |
Collapse
|
15
|
Nývltová E, Dietz JV, Seravalli J, Khalimonchuk O, Barrientos A. Coordination of metal center biogenesis in human cytochrome c oxidase. Nat Commun 2022; 13:3615. [PMID: 35750769 PMCID: PMC9232578 DOI: 10.1038/s41467-022-31413-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/16/2022] [Indexed: 01/12/2023] Open
Abstract
Mitochondrial cytochrome c oxidase (CcO) or respiratory chain complex IV is a heme aa3-copper oxygen reductase containing metal centers essential for holo-complex biogenesis and enzymatic function that are assembled by subunit-specific metallochaperones. The enzyme has two copper sites located in the catalytic core subunits. The COX1 subunit harbors the CuB site that tightly associates with heme a3 while the COX2 subunit contains the binuclear CuA site. Here, we report that in human cells the CcO copper chaperones form macromolecular assemblies and cooperate with several twin CX9C proteins to control heme a biosynthesis and coordinate copper transfer sequentially to the CuA and CuB sites. These data on CcO illustrate a mechanism that regulates the biogenesis of macromolecular enzymatic assemblies with several catalytic metal redox centers and prevents the accumulation of cytotoxic reactive assembly intermediates.
Collapse
Affiliation(s)
- Eva Nývltová
- Department of Neurology, University of Miami Miller School of Medicine, 1420NW 9th Ave, Miami, FL, 33136, USA
| | - Jonathan V Dietz
- Department of Biochemistry, University of Nebraska-Lincoln, 1901 Vine St. Beadle Center, Lincoln, NE, 68588, USA
| | - Javier Seravalli
- Nebraska Redox Biology Center, University of Nebraska-Lincoln, 1901 Vine St. Beadle Center, Lincoln, NE, 68588, USA
| | - Oleh Khalimonchuk
- Department of Biochemistry, University of Nebraska-Lincoln, 1901 Vine St. Beadle Center, Lincoln, NE, 68588, USA
- Nebraska Redox Biology Center, University of Nebraska-Lincoln, 1901 Vine St. Beadle Center, Lincoln, NE, 68588, USA
| | - Antoni Barrientos
- Department of Neurology, University of Miami Miller School of Medicine, 1420NW 9th Ave, Miami, FL, 33136, USA.
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1420NW 9th Ave, Miami, FL, 33136, USA.
| |
Collapse
|
16
|
Zhang H, Yan M, Liu T, Wei P, Chai N, Li L, Wang J, Yu X, Lin Y, Qiu B, Zhao Y. Dynamic Mitochondrial Proteome Under Polyamines Treatment in Cardiac Aging. Front Cell Dev Biol 2022; 10:840389. [PMID: 35372351 PMCID: PMC8965055 DOI: 10.3389/fcell.2022.840389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 11/13/2022] Open
Abstract
Age-related alteration of mitochondria causes impaired cardiac function, along with cellular and molecular changes. Polyamines can extend the life span in mice. However, whether polyamines can affect the dynamic mitochondrial proteome, thereby preventing age-related changes in cardiac function and cardiac aging, remains unclear. In this study, we found that spermine (Spm) and spermidine (Spd) injection for 6 weeks could prevent 24-month-old rats heart dysfunction, improve mitochondrial function, and downregulate apoptosis. Using iTRAQ tools, we identify 75 mitochondrial proteins of statistically significant alteration in aging hearts, which mainly participate in important mitochondrial physiological activity, such as metabolism, translation, transport, apoptosis, and oxidative phosphorylation. Moreover, four proteins of differential expression, pyruvate dehydrogenase kinase (PDK4), trifunctional enzyme subunit alpha (HADHA), nicotinamide nucleotide transhydrogenase (NNT), and Annexin6, which were significantly associated with heart aging, were validated by Western blotting. In vitro, we further demonstrated polyamines could retard cardiomyocytes aging through downregulating the expression of PDK4 and thereby inhibiting cell apoptosis. In summary, the distinct mitochondrial proteins identified in this study suggested some candidates involved in the anti-aging of the heart after polyamines treatment, and PDK4 may provide molecular clues for polyamines to inhibit apoptosis and thus retard aging-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Meng Yan
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Ting Liu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
| | - Peiling Wei
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
| | - Nannan Chai
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
- College of Nursing, Medical School of Chifeng University, Chifeng, China
| | - Lingxu Li
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
- Department of Nephrology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Junying Wang
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
- Department of Medical Technology, Beijing Health Vocational College, Beijing, China
| | - Xue Yu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
| | - Yan Lin
- Department of Pathophysiology, Qiqihar Medical University, Qiqihar, China
| | - Bintao Qiu
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yajun Zhao
- Department of Pathophysiology, Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Harbin, China
- *Correspondence: Yajun Zhao,
| |
Collapse
|
17
|
Wijesinghe TP, Dharmasivam M, Dai CC, Richardson DR. Innovative therapies for neuroblastoma: The surprisingly potent role of iron chelation in up-regulating metastasis and tumor suppressors and down-regulating the key oncogene, N-myc. Pharmacol Res 2021; 173:105889. [PMID: 34536548 DOI: 10.1016/j.phrs.2021.105889] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022]
Abstract
Iron is an indispensable requirement for essential biological processes in cancer cells. Due to the greater proliferation of neoplastic cells, their demand for iron is considerably higher relative to normal cells, making them highly susceptible to iron depletion. Understanding this sensitive relationship led to research exploring the effect of iron chelation therapy for cancer treatment. The classical iron-binding ligand, desferrioxamine (DFO), has demonstrated effective anti-proliferative activity against many cancer-types, particularly neuroblastoma tumors, and has the surprising activity of down-regulating the potent oncogene, N-myc, which is a major oncogenic driver in neuroblastoma. Even more significant is the ability of DFO to simultaneously up-regulate the potent metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), which plays a plethora of roles in suppressing a variety of oncogenic signaling pathways. However, DFO suffers the disadvantage of demonstrating poor membrane permeability and short plasma half-life, requiring administration by prolonged subcutaneous or intravenous infusions. Considering this, the specifically designed di-2-pyridylketone thiosemicarbazone (DpT) series of metal-binding ligands was developed in our laboratory. The lead agent from the first generation DpT series, di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT), showed exceptional anti-cancer properties compared to DFO. However, it exhibited cardiotoxicity in mouse models at higher dosages. Therefore, a second generation of agents was developed with the lead compound being di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) that progressed to Phase I clinical trials. Importantly, DpC showed better anti-proliferative activity than Dp44mT and no cardiotoxicity, demonstrating effective anti-cancer activity against neuroblastoma tumors in vivo.
Collapse
Affiliation(s)
- Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Charles C Dai
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
18
|
Geldon S, Fernández-Vizarra E, Tokatlidis K. Redox-Mediated Regulation of Mitochondrial Biogenesis, Dynamics, and Respiratory Chain Assembly in Yeast and Human Cells. Front Cell Dev Biol 2021; 9:720656. [PMID: 34557489 PMCID: PMC8452992 DOI: 10.3389/fcell.2021.720656] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022] Open
Abstract
Mitochondria are double-membrane organelles that contain their own genome, the mitochondrial DNA (mtDNA), and reminiscent of its endosymbiotic origin. Mitochondria are responsible for cellular respiration via the function of the electron oxidative phosphorylation system (OXPHOS), located in the mitochondrial inner membrane and composed of the four electron transport chain (ETC) enzymes (complexes I-IV), and the ATP synthase (complex V). Even though the mtDNA encodes essential OXPHOS components, the large majority of the structural subunits and additional biogenetical factors (more than seventy proteins) are encoded in the nucleus and translated in the cytoplasm. To incorporate these proteins and the rest of the mitochondrial proteome, mitochondria have evolved varied, and sophisticated import machineries that specifically target proteins to the different compartments defined by the two membranes. The intermembrane space (IMS) contains a high number of cysteine-rich proteins, which are mostly imported via the MIA40 oxidative folding system, dependent on the reduction, and oxidation of key Cys residues. Several of these proteins are structural components or assembly factors necessary for the correct maturation and function of the ETC complexes. Interestingly, many of these proteins are involved in the metalation of the active redox centers of complex IV, the terminal oxidase of the mitochondrial ETC. Due to their function in oxygen reduction, mitochondria are the main generators of reactive oxygen species (ROS), on both sides of the inner membrane, i.e., in the matrix and the IMS. ROS generation is important due to their role as signaling molecules, but an excessive production is detrimental due to unwanted oxidation reactions that impact on the function of different types of biomolecules contained in mitochondria. Therefore, the maintenance of the redox balance in the IMS is essential for mitochondrial function. In this review, we will discuss the role that redox regulation plays in the maintenance of IMS homeostasis as well as how mitochondrial ROS generation may be a key regulatory factor for ETC biogenesis, especially for complex IV.
Collapse
Affiliation(s)
| | - Erika Fernández-Vizarra
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kostas Tokatlidis
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
19
|
Welchen E, Canal MV, Gras DE, Gonzalez DH. Cross-talk between mitochondrial function, growth, and stress signalling pathways in plants. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:4102-4118. [PMID: 33369668 DOI: 10.1093/jxb/eraa608] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/22/2020] [Indexed: 05/16/2023]
Abstract
Plant mitochondria harbour complex metabolic routes that are interconnected with those of other cell compartments, and changes in mitochondrial function remotely influence processes in different parts of the cell. This implies the existence of signals that convey information about mitochondrial function to the rest of the cell. Increasing evidence indicates that metabolic and redox signals are important for this process, but changes in ion fluxes, protein relocalization, and physical contacts with other organelles are probably also involved. Besides possible direct effects of these signalling molecules on cellular functions, changes in mitochondrial physiology also affect the activity of different signalling pathways that modulate plant growth and stress responses. As a consequence, mitochondria influence the responses to internal and external factors that modify the activity of these pathways and associated biological processes. Acting through the activity of hormonal signalling pathways, mitochondria may also exert remote control over distant organs or plant tissues. In addition, an intimate cross-talk of mitochondria with energy signalling pathways, such as those represented by TARGET OF RAPAMYCIN and SUCROSE NON-FERMENTING1-RELATED PROTEIN KINASE 1, can be envisaged. This review discusses available evidence on the role of mitochondria in shaping plant growth and stress responses through various signalling pathways.
Collapse
Affiliation(s)
- Elina Welchen
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina
| | - María Victoria Canal
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina
| | - Diana E Gras
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina
| | - Daniel H Gonzalez
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina
| |
Collapse
|
20
|
Gladyck S, Aras S, Hüttemann M, Grossman LI. Regulation of COX Assembly and Function by Twin CX 9C Proteins-Implications for Human Disease. Cells 2021; 10:197. [PMID: 33498264 PMCID: PMC7909247 DOI: 10.3390/cells10020197] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/29/2022] Open
Abstract
Oxidative phosphorylation is a tightly regulated process in mammals that takes place in and across the inner mitochondrial membrane and consists of the electron transport chain and ATP synthase. Complex IV, or cytochrome c oxidase (COX), is the terminal enzyme of the electron transport chain, responsible for accepting electrons from cytochrome c, pumping protons to contribute to the gradient utilized by ATP synthase to produce ATP, and reducing oxygen to water. As such, COX is tightly regulated through numerous mechanisms including protein-protein interactions. The twin CX9C family of proteins has recently been shown to be involved in COX regulation by assisting with complex assembly, biogenesis, and activity. The twin CX9C motif allows for the import of these proteins into the intermembrane space of the mitochondria using the redox import machinery of Mia40/CHCHD4. Studies have shown that knockdown of the proteins discussed in this review results in decreased or completely deficient aerobic respiration in experimental models ranging from yeast to human cells, as the proteins are conserved across species. This article highlights and discusses the importance of COX regulation by twin CX9C proteins in the mitochondria via COX assembly and control of its activity through protein-protein interactions, which is further modulated by cell signaling pathways. Interestingly, select members of the CX9C protein family, including MNRR1 and CHCHD10, show a novel feature in that they not only localize to the mitochondria but also to the nucleus, where they mediate oxygen- and stress-induced transcriptional regulation, opening a new view of mitochondrial-nuclear crosstalk and its involvement in human disease.
Collapse
Affiliation(s)
- Stephanie Gladyck
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.G.); (S.A.); (M.H.)
| | - Siddhesh Aras
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.G.); (S.A.); (M.H.)
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland and Detroit, MI 48201, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.G.); (S.A.); (M.H.)
| | - Lawrence I. Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.G.); (S.A.); (M.H.)
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland and Detroit, MI 48201, USA
| |
Collapse
|
21
|
Quartey MO, Nyarko JNK, Maley JM, Barnes JR, Bolanos MAC, Heistad RM, Knudsen KJ, Pennington PR, Buttigieg J, De Carvalho CE, Leary SC, Parsons MP, Mousseau DD. The Aβ(1-38) peptide is a negative regulator of the Aβ(1-42) peptide implicated in Alzheimer disease progression. Sci Rep 2021; 11:431. [PMID: 33432101 PMCID: PMC7801637 DOI: 10.1038/s41598-020-80164-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
The pool of β-Amyloid (Aβ) length variants detected in preclinical and clinical Alzheimer disease (AD) samples suggests a diversity of roles for Aβ peptides. We examined how a naturally occurring variant, e.g. Aβ(1-38), interacts with the AD-related variant, Aβ(1-42), and the predominant physiological variant, Aβ(1-40). Atomic force microscopy, Thioflavin T fluorescence, circular dichroism, dynamic light scattering, and surface plasmon resonance reveal that Aβ(1-38) interacts differently with Aβ(1-40) and Aβ(1-42) and, in general, Aβ(1-38) interferes with the conversion of Aβ(1-42) to a β-sheet-rich aggregate. Functionally, Aβ(1-38) reverses the negative impact of Aβ(1-42) on long-term potentiation in acute hippocampal slices and on membrane conductance in primary neurons, and mitigates an Aβ(1-42) phenotype in Caenorhabditis elegans. Aβ(1-38) also reverses any loss of MTT conversion induced by Aβ(1-40) and Aβ(1-42) in HT-22 hippocampal neurons and APOE ε4-positive human fibroblasts, although the combination of Aβ(1-38) and Aβ(1-42) inhibits MTT conversion in APOE ε4-negative fibroblasts. A greater ratio of soluble Aβ(1-42)/Aβ(1-38) [and Aβ(1-42)/Aβ(1-40)] in autopsied brain extracts correlates with an earlier age-at-death in males (but not females) with a diagnosis of AD. These results suggest that Aβ(1-38) is capable of physically counteracting, potentially in a sex-dependent manner, the neuropathological effects of the AD-relevant Aβ(1-42).
Collapse
Affiliation(s)
- Maa O Quartey
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Jennifer N K Nyarko
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Jason M Maley
- Saskatchewan Structural Sciences Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jocelyn R Barnes
- Division of BioMedical Sciences (Neurosciences), Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Ryan M Heistad
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Kaeli J Knudsen
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Paul R Pennington
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada
| | - Josef Buttigieg
- Department of Biology, University of Regina, Regina, SK, Canada
| | | | - Scot C Leary
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Matthew P Parsons
- Division of BioMedical Sciences (Neurosciences), Memorial University of Newfoundland, St. John's, NL, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, GB41 HSB, 107 Wiggins Rd., Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
22
|
Ekim Kocabey A, Rödel G, Gey U. The antioxidant function of Sco proteins depends on a critical surface-exposed residue. Biochim Biophys Acta Gen Subj 2020; 1865:129781. [PMID: 33171213 DOI: 10.1016/j.bbagen.2020.129781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/08/2020] [Accepted: 11/02/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Besides their role in copper metabolism, Sco proteins from different organisms have been shown to play a defensive role against oxidative stress. In the present study, we set out to identify crucial amino acid residues for the antioxidant activity. METHODS Native and mutated Sco proteins from human, Arabidopsis thaliana and the yeast Kluyveromyces lactis were expressed in the model organism Saccharomyces cerevisiae. The oxidative stress resistance of the respective transformants was determined by growth and lipid peroxidation assays. RESULTS A functionally important site, located 15 amino acids downstream of the well-conserved copper binding CxxxC motif, was identified. Mutational analysis revealed that a positive charge at this position has a detrimental effect on the antioxidant capacity. Bioinformatic analysis predicts that this site is surface-exposed, and according to Co-IP data it is required for binding of proteins that are connected to known antioxidant pathways. CONCLUSION This study shows that the antioxidant capacity of eukaryotic Sco proteins is conserved and depends on the presence of functional site(s) rather than the extent of overall sequence homology. GENERAL SIGNIFICANCE These findings provide an insight into the conserved functional sites of eukaryotic Sco proteins that are crucial for combating oxidative stress. This capacity is probably not due to an enzymatic activity but rather is indirectly mediated by interaction with other proteins.
Collapse
Affiliation(s)
| | - Gerhard Rödel
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
| | - Uta Gey
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
23
|
Cobine PA, Moore SA, Leary SC. Getting out what you put in: Copper in mitochondria and its impacts on human disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118867. [PMID: 32979421 DOI: 10.1016/j.bbamcr.2020.118867] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/22/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Mitochondria accumulate copper in their matrix for the eventual maturation of the cuproenzymes cytochrome c oxidase and superoxide dismutase. Transport into the matrix is achieved by mitochondrial carrier family (MCF) proteins. The major copper transporting MCF described to date in yeast is Pic2, which imports the metal ion into the matrix. Pic2 is one of ~30 MCFs that move numerous metabolites, nucleotides and co-factors across the inner membrane for use in the matrix. Genetic and biochemical experiments showed that Pic2 is required for cytochrome c oxidase activity under copper stress, and that it is capable of transporting ionic and complexed forms of copper. The Pic2 ortholog SLC25A3, one of 53 mammalian MCFs, functions as both a copper and a phosphate transporter. Depletion of SLC25A3 results in decreased accumulation of copper in the matrix, a cytochrome c oxidase defect and a modulation of cytosolic superoxide dismutase abundance. The regulatory roles for copper and cuproproteins resident to the mitochondrion continue to expand beyond the organelle. Mitochondrial copper chaperones have been linked to the modulation of cellular copper uptake and export and the facilitation of inter-organ communication. Recently, a role for matrix copper has also been proposed in a novel cell death pathway termed cuproptosis. This review will detail our understanding of the maturation of mitochondrial copper enzymes, the roles of mitochondrial signals in regulating cellular copper content, the proposed mechanisms of copper transport into the organelle and explore the evolutionary origins of copper homeostasis pathways.
Collapse
Affiliation(s)
- Paul A Cobine
- Department of Biological Sciences, Auburn University, Auburn, AL, USA.
| | - Stanley A Moore
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Scot C Leary
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
24
|
Santos G, Borges JMP, Avila-Rodriguez M, Gaíno SB, Barreto GE, Rúbio ÉP, Aguiar RM, Galembeck E, Bromochenkel CB, de Oliveira DM. Copper and Neurotoxicity in Autism Spectrum Disorder. Curr Pharm Des 2019; 25:4747-4754. [PMID: 31845627 DOI: 10.2174/1381612825666191217091939] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/08/2019] [Indexed: 12/23/2022]
Abstract
Free radicals (FR) act on living organisms and present unpaired electrons in the molecular orbitals of oxygen or nitrogen species. They are classified as redox reactions and account for a wide range of processes in biological systems. Genetic and environmental factors may alter the levels of FR in the cell, leading to deleterious consequences such as membrane lipid peroxidation, protein nitration, enzyme, carbohydrate and DNA damage, ultimately resulting in premature aging and a pro-inflammatory microenvironment as observed in Alzheimer's disease (AD) and autism spectrum disorder (ASD). O2 radical ability to act as a Lewis base and to form a complex with metal transition such as iron and copper (Lewis acids) leads to biomolecules oxidation at physiological pH, thus increasing the possibility of injury and oxidative damage in biological tissues. In this review, we discuss the role of metals, like copper, and the amyloid precursor protein (APP) derivative (s-APP-alpha) as an antioxidant and a possible adjuvant in the treatment of some autistic spectrum disorder symptoms (ASD).
Collapse
Affiliation(s)
- Gesivaldo Santos
- Department of Biological Science, State University of Southwestern of Bahia, Bahia, Brazil
| | - Julita M P Borges
- Department of Science and Technology, State University of Southwestern of Bahia, Bahia, Brazil
| | | | | | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Érika P Rúbio
- Department of Science and Technology, State University of Southwestern of Bahia, Bahia, Brazil
| | - Rosane M Aguiar
- Department of Science and Technology, State University of Southwestern of Bahia, Bahia, Brazil
| | - Eduardo Galembeck
- Institute of Biology, State University of Campinas-São Paulo, São Paulo, Brazil
| | | | - Djalma M de Oliveira
- Department of Science and Technology, State University of Southwestern of Bahia, Bahia, Brazil
| |
Collapse
|
25
|
Garcia L, Mansilla N, Ocampos N, Pagani MA, Welchen E, Gonzalez DH. The mitochondrial copper chaperone COX19 influences copper and iron homeostasis in arabidopsis. PLANT MOLECULAR BIOLOGY 2019; 99:621-638. [PMID: 30778722 DOI: 10.1007/s11103-019-00840-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/09/2019] [Indexed: 05/24/2023]
Abstract
The mitochondrial metallochaperone COX19 influences iron and copper responses highlighting a role of mitochondria in modulating metal homeostasis in Arabidopsis. The mitochondrial copper chaperone COX19 participates in the biogenesis of cytochrome c oxidase (COX) in yeast and humans. In this work, we studied the function of COX19 in Arabidopsis thaliana, using plants with either decreased or increased COX19 levels. A fusion of COX19 to the red fluorescent protein localized to mitochondria in vivo, suggesting that Arabidopsis COX19 is a mitochondrial protein. Silencing of COX19 using an artificial miRNA did not cause changes in COX activity levels or respiration in plants grown under standard conditions. These amiCOX19 plants, however, showed decreased expression of the low-copper responsive miRNA gene MIR398b and an induction of the miR398 target CSD1 relative to wild-type plants. Plants with increased COX19 levels, instead, showed induction of MIR398b and other low-copper responsive genes. In addition, global transcriptional changes in rosettes of amiCOX19 plants resembled those observed under iron deficiency. Phenotypic analysis indicated that the roots of amiCOX19 plants show altered growth responses to copper excess and iron deficiency. COX activity levels and COX-dependent respiration were lower in amiCOX19 plants than in wild-type plants under iron deficiency conditions, suggesting that COX19 function is particularly important for COX assembly under iron deficiency. The results indicate that the mitochondrial copper chaperone COX19 has a role in regulating copper and iron homeostasis and responses in plants.
Collapse
Affiliation(s)
- Lucila Garcia
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Centro Científico Tecnológico CONICET Santa Fe, Universidad Nacional del Litoral, Colectora Ruta Nac. Nº 168 km 0, Paraje el Pozo s/n, 3000, Santa Fe, Argentina
- Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Ocampo y Esmeralda s/n, 2000, Rosario, Argentina
| | - Natanael Mansilla
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Centro Científico Tecnológico CONICET Santa Fe, Universidad Nacional del Litoral, Colectora Ruta Nac. Nº 168 km 0, Paraje el Pozo s/n, 3000, Santa Fe, Argentina
| | - Natacha Ocampos
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Centro Científico Tecnológico CONICET Santa Fe, Universidad Nacional del Litoral, Colectora Ruta Nac. Nº 168 km 0, Paraje el Pozo s/n, 3000, Santa Fe, Argentina
| | - María A Pagani
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Suipacha 570, 2000, Rosario, Argentina
| | - Elina Welchen
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Centro Científico Tecnológico CONICET Santa Fe, Universidad Nacional del Litoral, Colectora Ruta Nac. Nº 168 km 0, Paraje el Pozo s/n, 3000, Santa Fe, Argentina
| | - Daniel H Gonzalez
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Centro Científico Tecnológico CONICET Santa Fe, Universidad Nacional del Litoral, Colectora Ruta Nac. Nº 168 km 0, Paraje el Pozo s/n, 3000, Santa Fe, Argentina.
| |
Collapse
|
26
|
Yamada Y, Prosser RA. Copper in the suprachiasmatic circadian clock: A possible link between multiple circadian oscillators. Eur J Neurosci 2018; 51:47-70. [PMID: 30269387 DOI: 10.1111/ejn.14181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/07/2023]
Abstract
The mammalian circadian clock in the suprachiasmatic nucleus (SCN) is very robust, able to coordinate our daily physiological and behavioral rhythms with exquisite accuracy. Simultaneously, the SCN clock is highly sensitive to environmental timing cues such as the solar cycle. This duality of resiliency and sensitivity may be sustained in part by a complex intertwining of three cellular oscillators: transcription/translation, metabolic/redox, and membrane excitability. We suggest here that one of the links connecting these oscillators may be forged from copper (Cu). Cellular Cu levels are highly regulated in the brain and peripherally, and Cu affects cellular metabolism, redox state, cell signaling, and transcription. We have shown that both Cu chelation and application induce nighttime phase shifts of the SCN clock in vitro and that these treatments affect glutamate, N-methyl-D-aspartate receptor, and associated signaling processes differently. More recently we found that Cu induces mitogen-activated protein kinase-dependent phase shifts, while the mechanisms by which Cu removal induces phase shifts remain unclear. Lastly, we have found that two Cu transporters are expressed in the SCN, and that one of these transporters (ATP7A) exhibits a day/night rhythm. Our results suggest that Cu homeostasis is tightly regulated in the SCN, and that changes in Cu levels may serve as a time cue for the circadian clock. We discuss these findings in light of the existing literature and current models of multiple coupled circadian oscillators in the SCN.
Collapse
Affiliation(s)
- Yukihiro Yamada
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| | - Rebecca A Prosser
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|
27
|
Baker ZN, Jett K, Boulet A, Hossain A, Cobine PA, Kim BE, El Zawily AM, Lee L, Tibbits GF, Petris MJ, Leary SC. The mitochondrial metallochaperone SCO1 maintains CTR1 at the plasma membrane to preserve copper homeostasis in the murine heart. Hum Mol Genet 2018; 26:4617-4628. [PMID: 28973536 PMCID: PMC5886179 DOI: 10.1093/hmg/ddx344] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/31/2017] [Indexed: 11/14/2022] Open
Abstract
SCO1 is a ubiquitously expressed, mitochondrial protein with essential roles in cytochrome c oxidase (COX) assembly and the regulation of copper homeostasis. SCO1 patients present with severe forms of early onset disease, and ultimately succumb from liver, heart or brain failure. However, the inherent susceptibility of these tissues to SCO1 mutations and the clinical heterogeneity observed across SCO1 pedigrees remain poorly understood phenomena. To further address this issue, we generated Sco1hrt/hrt and Sco1stm/stm mice in which Sco1 was specifically deleted in heart and striated muscle, respectively. Lethality was observed in both models due to a combined COX and copper deficiency that resulted in a dilated cardiomyopathy. Left ventricular dilation and loss of heart function was preceded by a temporal decrease in COX activity and copper levels in the longer-lived Sco1stm/stm mice. Interestingly, the reduction in copper content of Sco1stm/stm cardiomyocytes was due to the mislocalisation of CTR1, the high affinity transporter that imports copper into the cell. CTR1 was similarly mislocalized to the cytosol in the heart of knockin mice carrying a homozygous G115S substitution in Sco1, which in humans causes a hypertrophic cardiomyopathy. Our current findings in the heart are in marked contrast to our prior observations in the liver, where Sco1 deletion results in a near complete absence of CTR1 protein. These data collectively argue that mutations perturbing SCO1 function have tissue-specific consequences for the machinery that ultimately governs copper homeostasis, and further establish the importance of aberrant mitochondrial signaling to the etiology of copper handling disorders.
Collapse
Affiliation(s)
- Zakery N Baker
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Kimberly Jett
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Aren Boulet
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Amzad Hossain
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Paul A Cobine
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Byung-Eun Kim
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Amr M El Zawily
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Ling Lee
- Department of Cardiovascular Sciences, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Glen F Tibbits
- Department of Cardiovascular Sciences, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Michael J Petris
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Scot C Leary
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
28
|
Lang J, Zhao Q, He Y, Yu X. Bone turnover markers and novel biomarkers in lung cancer bone metastases. Biomarkers 2018; 23:518-526. [PMID: 29683727 DOI: 10.1080/1354750x.2018.1463566] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT Lung cancer still remains the leading cause of cancer-related mortality worldwide. Bone is one of preferred metastatic sites for lung cancer cells. So far, both accurate diagnosis and effective treatment of lung cancer bone metastases are difficult. OBJECTIVE This review aimed to evaluate roles of bone turnover markers (BTMs), microRNAs (miRNAs), dickkopf1 (DKK1) and insulin like growth factor binding protein 3 (IGFBP-3) in lung cancer bone metastases. METHODS We searched articles about these four biomarkers in lung cancer bone metastases mainly in PubMed. RESULT The levels of bone specific alkaline phosphatase (BALP), cross-linked carboxy-terminal telopeptide of type-I collagen (ICTP) and N-terminal telopeptides of type-I collagen (NTX) were reported to be significantly increased in lung cancer patients with bone metastases. ALP, NTX and bone sialoprotein were thought to be associated with prognosis of lung cancer patients with bone metastases. MiRNA-335, miRNA-33a, miRNA-21, DKK1 and IGFBP-3 were revealed to be novel biomarkers in lung cancer bone metastases. DISCUSSION AND CONCLUSION Current researches have revealed that BTMs, miRNAs, DKK1 and IGFBP-3 may be useful in diagnosis, prognosis evaluation or treatment of lung cancer bone metastases. More studies about these biomarkers in lung cancer bone metastases are needed.
Collapse
Affiliation(s)
- Jiangli Lang
- a Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism , State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , China
| | - Qian Zhao
- b Department of General practice , West China Hospital, Sichuan University , Chengdu , China
| | - Yuedong He
- c Department of Gynecology , West China Second University Hospital, Sichuan University , Chengdu , China
| | - Xijie Yu
- a Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism , State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , China
| |
Collapse
|
29
|
Bhattacharjee A, Chakraborty K, Shukla A. Cellular copper homeostasis: current concepts on its interplay with glutathione homeostasis and its implication in physiology and human diseases. Metallomics 2018; 9:1376-1388. [PMID: 28675215 DOI: 10.1039/c7mt00066a] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Copper is a trace element essential for almost all living organisms. But the level of intracellular copper needs to be tightly regulated. Dysregulation of cellular copper homeostasis leading to various diseases demonstrates the importance of this tight regulation. Copper homeostasis is regulated not only within the cell but also within individual intracellular compartments. Inactivation of export machinery results in excess copper being redistributed into various intracellular organelles. Recent evidence suggests the involvement of glutathione in playing an important role in regulating copper entry and intracellular copper homeostasis. Therefore interplay of both homeostases might play an important role within the cell. Similar to copper, glutathione balance is tightly regulated within individual cellular compartments. This review explores the existing literature on the role of glutathione in regulating cellular copper homeostasis. On the one hand, interplay of glutathione and copper homeostasis performs an important role in normal physiological processes, for example neuronal differentiation. On the other hand, perturbation of the interplay might play a key role in the pathogenesis of copper homeostasis disorders.
Collapse
|
30
|
Mansilla N, Racca S, Gras DE, Gonzalez DH, Welchen E. The Complexity of Mitochondrial Complex IV: An Update of Cytochrome c Oxidase Biogenesis in Plants. Int J Mol Sci 2018; 19:ijms19030662. [PMID: 29495437 PMCID: PMC5877523 DOI: 10.3390/ijms19030662] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial respiration is an energy producing process that involves the coordinated action of several protein complexes embedded in the inner membrane to finally produce ATP. Complex IV or Cytochrome c Oxidase (COX) is the last electron acceptor of the respiratory chain, involved in the reduction of O2 to H2O. COX is a multimeric complex formed by multiple structural subunits encoded in two different genomes, prosthetic groups (heme a and heme a3), and metallic centers (CuA and CuB). Tens of accessory proteins are required for mitochondrial RNA processing, synthesis and delivery of prosthetic groups and metallic centers, and for the final assembly of subunits to build a functional complex. In this review, we perform a comparative analysis of COX composition and biogenesis factors in yeast, mammals and plants. We also describe possible external and internal factors controlling the expression of structural proteins and assembly factors at the transcriptional and post-translational levels, and the effect of deficiencies in different steps of COX biogenesis to infer the role of COX in different aspects of plant development. We conclude that COX assembly in plants has conserved and specific features, probably due to the incorporation of a different set of subunits during evolution.
Collapse
Affiliation(s)
- Natanael Mansilla
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina.
| | - Sofia Racca
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina.
| | - Diana E Gras
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina.
| | - Daniel H Gonzalez
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina.
| | - Elina Welchen
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, 3000 Santa Fe, Argentina.
| |
Collapse
|
31
|
Boulet A, Vest KE, Maynard MK, Gammon MG, Russell AC, Mathews AT, Cole SE, Zhu X, Phillips CB, Kwong JQ, Dodani SC, Leary SC, Cobine PA. The mammalian phosphate carrier SLC25A3 is a mitochondrial copper transporter required for cytochrome c oxidase biogenesis. J Biol Chem 2017; 293:1887-1896. [PMID: 29237729 DOI: 10.1074/jbc.ra117.000265] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/09/2017] [Indexed: 01/01/2023] Open
Abstract
Copper is required for the activity of cytochrome c oxidase (COX), the terminal electron-accepting complex of the mitochondrial respiratory chain. The likely source of copper used for COX biogenesis is a labile pool found in the mitochondrial matrix. In mammals, the proteins that transport copper across the inner mitochondrial membrane remain unknown. We previously reported that the mitochondrial carrier family protein Pic2 in budding yeast is a copper importer. The closest Pic2 ortholog in mammalian cells is the mitochondrial phosphate carrier SLC25A3. Here, to investigate whether SLC25A3 also transports copper, we manipulated its expression in several murine and human cell lines. SLC25A3 knockdown or deletion consistently resulted in an isolated COX deficiency in these cells, and copper addition to the culture medium suppressed these biochemical defects. Consistent with a conserved role for SLC25A3 in copper transport, its heterologous expression in yeast complemented copper-specific defects observed upon deletion of PIC2 Additionally, assays in Lactococcus lactis and in reconstituted liposomes directly demonstrated that SLC25A3 functions as a copper transporter. Taken together, these data indicate that SLC25A3 can transport copper both in vitro and in vivo.
Collapse
Affiliation(s)
- Aren Boulet
- From the Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan 7N 5E5, Canada
| | - Katherine E Vest
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - Margaret K Maynard
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - Micah G Gammon
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | | | - Alexander T Mathews
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - Shelbie E Cole
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - Xinyu Zhu
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - Casey B Phillips
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - Jennifer Q Kwong
- Department of Pediatrics, Emory University, Atlanta, Georgia 30322, and
| | - Sheel C Dodani
- the Department of Chemistry and Biochemistry, University of Texas at Dallas, Dallas, Texas 75080
| | - Scot C Leary
- From the Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan 7N 5E5, Canada
| | - Paul A Cobine
- the Department of Biological Sciences, Auburn University, Auburn, Alabama 36849,
| |
Collapse
|
32
|
Abstract
All known eukaryotes require copper for their development and survival. The essentiality of copper reflects its widespread use as a co-factor in conserved enzymes that catalyze biochemical reactions critical to energy production, free radical detoxification, collagen deposition, neurotransmitter biosynthesis and iron homeostasis. However, the prioritized use of copper poses an organism with a considerable challenge because, in its unbound form, copper can potentiate free radical production and displace iron-sulphur clusters to disrupt protein function. Protective mechanisms therefore evolved to mitigate this challenge and tightly regulate the acquisition, trafficking and storage of copper such that the metal ion is rarely found in its free form in the cell. Findings by a number of groups over the last ten years emphasize that this regulatory framework forms the foundation of a system that is capable of monitoring copper status and reprioritizing copper usage at both the cellular and systemic levels of organization. While the identification of relevant molecular mechanisms and signaling pathways has proven to be difficult and remains a barrier to our full understanding of the regulation of copper homeostasis, mounting evidence points to the mitochondrion as a pivotal hub in this regard in both healthy and diseased states. Here, we review our current understanding of copper handling pathways contained within the organelle and consider plausible mechanisms that may serve to functionally couple their activity to that of other cellular copper handling machinery to maintain copper homeostasis.
Collapse
Affiliation(s)
- Zakery N. Baker
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK Canada S7N 5E5
| | - Paul A. Cobine
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849, USA
| | - Scot C. Leary
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK Canada S7N 5E5
| |
Collapse
|
33
|
Comstra HS, McArthy J, Rudin-Rush S, Hartwig C, Gokhale A, Zlatic SA, Blackburn JB, Werner E, Petris M, D'Souza P, Panuwet P, Barr DB, Lupashin V, Vrailas-Mortimer A, Faundez V. The interactome of the copper transporter ATP7A belongs to a network of neurodevelopmental and neurodegeneration factors. eLife 2017; 6. [PMID: 28355134 PMCID: PMC5400511 DOI: 10.7554/elife.24722] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/28/2017] [Indexed: 02/04/2023] Open
Abstract
Genetic and environmental factors, such as metals, interact to determine neurological traits. We reasoned that interactomes of molecules handling metals in neurons should include novel metal homeostasis pathways. We focused on copper and its transporter ATP7A because ATP7A null mutations cause neurodegeneration. We performed ATP7A immunoaffinity chromatography and identified 541 proteins co-isolating with ATP7A. The ATP7A interactome concentrated gene products implicated in neurodegeneration and neurodevelopmental disorders, including subunits of the Golgi-localized conserved oligomeric Golgi (COG) complex. COG null cells possess altered content and subcellular localization of ATP7A and CTR1 (SLC31A1), the transporter required for copper uptake, as well as decreased total cellular copper, and impaired copper-dependent metabolic responses. Changes in the expression of ATP7A and COG subunits in Drosophila neurons altered synapse development in larvae and copper-induced mortality of adult flies. We conclude that the ATP7A interactome encompasses a novel COG-dependent mechanism to specify neuronal development and survival.
Collapse
Affiliation(s)
- Heather S Comstra
- Departments of Cell Biology, Emory University, Atlanta, United States
| | - Jacob McArthy
- School of Biological Sciences, Illinois State University, Normal, United States
| | | | - Cortnie Hartwig
- Department of Chemistry, Agnes Scott College, Decatur, Georgia
| | - Avanti Gokhale
- Departments of Cell Biology, Emory University, Atlanta, United States
| | | | - Jessica B Blackburn
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Erica Werner
- Department of Biochemistry, Emory University, Atlanta, United States
| | - Michael Petris
- Department of Biochemistry, University of Missouri, Columbia, United States
| | - Priya D'Souza
- Rollins School of Public Health, Emory University, Atlanta, United States
| | - Parinya Panuwet
- Rollins School of Public Health, Emory University, Atlanta, United States
| | - Dana Boyd Barr
- Rollins School of Public Health, Emory University, Atlanta, United States
| | - Vladimir Lupashin
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, United States
| | | | - Victor Faundez
- Departments of Cell Biology, Emory University, Atlanta, United States
| |
Collapse
|
34
|
Öhrvik H, Aaseth J, Horn N. Orchestration of dynamic copper navigation – new and missing pieces. Metallomics 2017; 9:1204-1229. [DOI: 10.1039/c7mt00010c] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A general principle in all cells in the body is that an essential metal – here copper – is taken up at the plasma membrane, directed through cellular compartments for use in specific enzymes and pathways, stored in specific scavenging molecules if in surplus, and finally expelled from the cells.
Collapse
Affiliation(s)
- Helena Öhrvik
- Medical Biochemistry and Microbiology
- Uppsala University
- Sweden
| | - Jan Aaseth
- Innlandet Hospital Trust and Inland Norway University of Applied Sciences
- Norway
| | | |
Collapse
|
35
|
Abstract
Copper (Cu) is indispensible for growth and development of human organisms. It is required for such fundamental and ubiquitous processes as respiration and protection against reactive oxygen species. Cu also enables catalytic activity of enzymes that critically contribute to the functional identity of many cells and tissues. Pigmentation, production of norepinephrine by the adrenal gland, the key steps in the formation of connective tissue, neuroendocrine signaling, wound healing - all these processes require Cu and depend on Cu entering the secretory pathway. To reach the Cu-dependent enzymes in a lumen of the trans-Golgi network and various vesicular compartments, Cu undertakes a complex journey crossing the extracellular and intracellular membranes and staying firmly on course while traveling in a cytosol. The proteins that assist Cu in this journey by mediating its entry, distribution, and export, have been identified. The accumulating data also indicate that the current model of cellular Cu homeostasis is still a "skeleton" that has to be fleshed out with many new details. This review summarizes recent data on the mechanisms responsible for Cu transfer to the secretory pathway. The emerging new concepts and gaps in our knowledge are discussed.
Collapse
Affiliation(s)
- Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, 725 N. Wolfe street, Baltimore, MD 21205, USA.
| |
Collapse
|
36
|
Systems-level effects of ectopic galectin-7 reconstitution in cervical cancer and its microenvironment. BMC Cancer 2016; 16:680. [PMID: 27558259 PMCID: PMC4997669 DOI: 10.1186/s12885-016-2700-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
Background Galectin-7 (Gal-7) is negatively regulated in cervical cancer, and appears to be a link between the apoptotic response triggered by cancer and the anti-tumoral activity of the immune system. Our understanding of how cervical cancer cells and their molecular networks adapt in response to the expression of Gal-7 remains limited. Methods Meta-analysis of Gal-7 expression was conducted in three cervical cancer cohort studies and TCGA. In silico prediction and bisulfite sequencing were performed to inquire epigenetic alterations. To study the effect of Gal-7 on cervical cancer, we ectopically re-expressed it in the HeLa and SiHa cervical cancer cell lines, and analyzed their transcriptome and SILAC-based proteome. We also examined the tumor and microenvironment host cell transcriptomes after xenotransplantation into immunocompromised mice. Differences between samples were assessed with the Kruskall-Wallis, Dunn’s Multiple Comparison and T tests. Kaplan–Meier and log-rank tests were used to determine overall survival. Results Gal-7 was constantly downregulated in our meta-analysis (p < 0.0001). Tumors with combined high Gal-7 and low galectin-1 expression (p = 0.0001) presented significantly better prognoses (p = 0.005). In silico and bisulfite sequencing assays showed de novo methylation in the Gal-7 promoter and first intron. Cells re-expressing Gal-7 showed a high apoptosis ratio (p < 0.05) and their xenografts displayed strong growth retardation (p < 0.001). Multiple gene modules and transcriptional regulators were modulated in response to Gal-7 reconstitution, both in cervical cancer cells and their microenvironments (FDR < 0.05 %). Most of these genes and modules were associated with tissue morphogenesis, metabolism, transport, chemokine activity, and immune response. These functional modules could exert the same effects in vitro and in vivo, even despite different compositions between HeLa and SiHa samples. Conclusions Gal-7 re-expression affects the regulation of molecular networks in cervical cancer that are involved in diverse cancer hallmarks, such as metabolism, growth control, invasion and evasion of apoptosis. The effect of Gal-7 extends to the microenvironment, where networks involved in its configuration and in immune surveillance are particularly affected. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2700-8) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
The Role of Copper Chaperone Atox1 in Coupling Redox Homeostasis to Intracellular Copper Distribution. Antioxidants (Basel) 2016; 5:antiox5030025. [PMID: 27472369 PMCID: PMC5039574 DOI: 10.3390/antiox5030025] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/13/2016] [Accepted: 07/22/2016] [Indexed: 01/15/2023] Open
Abstract
Human antioxidant protein 1 (Atox1) is a small cytosolic protein with an essential role in copper homeostasis. Atox1 functions as a copper carrier facilitating copper transfer to the secretory pathway. This process is required for activation of copper dependent enzymes involved in neurotransmitter biosynthesis, iron efflux, neovascularization, wound healing, and regulation of blood pressure. Recently, new cellular roles for Atox1 have emerged. Changing levels of Atox1 were shown to modulate response to cancer therapies, contribute to inflammatory response, and protect cells against various oxidative stresses. It has also become apparent that the activity of Atox1 is tightly linked to the cellular redox status. In this review, we summarize biochemical information related to a dual role of Atox1 as a copper chaperone and an antioxidant. We discuss how these two activities could be linked and contribute to establishing the intracellular copper balance and functional identity of cells during differentiation.
Collapse
|
38
|
Dela Cruz R, Jeong MY, Winge DR. Cox1 mutation abrogates need for Cox23 in cytochrome c oxidase biogenesis. MICROBIAL CELL 2016; 3:275-284. [PMID: 28357365 PMCID: PMC5354592 DOI: 10.15698/mic2016.07.511] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cox23 is a known conserved assembly factor for cytochrome c
oxidase, although its role in cytochrome c oxidase (CcO)
biogenesis remains unresolved. To gain additional insights into its role, we
isolated spontaneous suppressors of the respiratory growth defect in
cox23∆ yeast cells. We recovered independent colonies that
propagated on glycerol/lactate medium for cox23∆ cells at 37°C.
We mapped these mutations to the mitochondrial genome and specifically to
COX1 yielding an I101F substitution. The
I101F Cox1 allele is a gain-of-function mutation enabling yeast
to respire in the absence of Cox23. CcO subunit steady-state levels were
restored with the I101F Cox1 suppressor mutation and oxygen
consumption and CcO activity were likewise restored. Cells harboring the
mitochondrial genome encoding I101F Cox1 were used to delete genes
for other CcO assembly factors to test the specificity of the Cox1 mutation as a
suppressor of cox23∆ cells. The Cox1 mutant allele fails to
support respiratory growth in yeast lacking Cox17, Cox19, Coa1, Coa2, Cox14 or
Shy1, demonstrating its specific suppressor activity for cox23∆
cells.
Collapse
Affiliation(s)
- Richard Dela Cruz
- University of Utah Health Sciences Center, Departments of Medicine and Biochemistry, Salt Lake City, Utah 84132, USA. ; Present address: Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Mi-Young Jeong
- University of Utah Health Sciences Center, Departments of Medicine and Biochemistry, Salt Lake City, Utah 84132, USA
| | - Dennis R Winge
- University of Utah Health Sciences Center, Departments of Medicine and Biochemistry, Salt Lake City, Utah 84132, USA
| |
Collapse
|
39
|
Bhattacharjee A, Yang H, Duffy M, Robinson E, Conrad-Antoville A, Lu YW, Capps T, Braiterman L, Wolfgang M, Murphy MP, Yi L, Kaler SG, Lutsenko S, Ralle M. The Activity of Menkes Disease Protein ATP7A Is Essential for Redox Balance in Mitochondria. J Biol Chem 2016; 291:16644-58. [PMID: 27226607 PMCID: PMC4974379 DOI: 10.1074/jbc.m116.727248] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Indexed: 11/13/2022] Open
Abstract
Copper-transporting ATPase ATP7A is essential for mammalian copper homeostasis. Loss of ATP7A activity is associated with fatal Menkes disease and various other pathologies. In cells, ATP7A inactivation disrupts copper transport from the cytosol into the secretory pathway. Using fibroblasts from Menkes disease patients and mouse 3T3-L1 cells with a CRISPR/Cas9-inactivated ATP7A, we demonstrate that ATP7A dysfunction is also damaging to mitochondrial redox balance. In these cells, copper accumulates in nuclei, cytosol, and mitochondria, causing distinct changes in their redox environment. Quantitative imaging of live cells using GRX1-roGFP2 and HyPer sensors reveals highest glutathione oxidation and elevation of H2O2 in mitochondria, whereas the redox environment of nuclei and the cytosol is much less affected. Decreasing the H2O2 levels in mitochondria with MitoQ does not prevent glutathione oxidation; i.e. elevated copper and not H2O2 is a primary cause of glutathione oxidation. Redox misbalance does not significantly affect mitochondrion morphology or the activity of respiratory complex IV but markedly increases cell sensitivity to even mild glutathione depletion, resulting in loss of cell viability. Thus, ATP7A activity protects mitochondria from excessive copper entry, which is deleterious to redox buffers. Mitochondrial redox misbalance could significantly contribute to pathologies associated with ATP7A inactivation in tissues with paradoxical accumulation of copper (i.e. renal epithelia).
Collapse
Affiliation(s)
| | | | - Megan Duffy
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | - Emily Robinson
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | - Arianrhod Conrad-Antoville
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | | | - Tony Capps
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | | | - Michael Wolfgang
- Cell Biology Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Michael P Murphy
- the Medical Research Council Mitochondrial Biology Unit, Cambridge CB2 0XY, United Kingdom, and
| | - Ling Yi
- the Section on Translational Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Stephen G Kaler
- the Section on Translational Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | | | - Martina Ralle
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239,
| |
Collapse
|
40
|
Pastorkova Z, Skarda J, Andel J. The role of microRNA in metastatic processes of non-small cell lung carcinoma. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2016; 160:343-57. [PMID: 27108604 DOI: 10.5507/bp.2016.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 04/08/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND MicroRNAs are small non-coding one-stranded RNA molecules that play an important role in the post-transcriptional regulation of genes. Bioinformatic predictions indicate that each miRNA can regulate hundreds of target genes. MicroRNA expression can be associated with various cellular processes leading to the metastasis of malignant tumours including non-small cell lung carcinoma. This review summarizes current knowledge on the role of microRNAs in NSCLC metastasis to the brain and lymph nodes. METHODS A search of the NCBI/PubMed database for publications on expression levels and the mechanisms of microRNA action in NSCLC metastasis. RESULTS AND CONCLUSION Dysregulation of microRNAs in NSCLC can be associated with brain and lymph node metastasis. There are differences in microRNA expression profiling between NSCLC with and without metastases but it is currently not possible to reliably predict the site of metastasis in NSCLC. Based on data from RNAmicroarrays, bioinformatics analysis is able to predict the target genes of highlighted microRNAs, providing us with complex information about cancer cell features such as enhanced proliferation, migration and invasion. Such microRNAs may then be knocked-down using siRNAs or substituted with miRNA mimics. RNA microarray profiling may thus be a useful tool to select up- or down-regulated microRNAs. A number of authors suggest that microRNAs could serve as biomarkers and therapeutic targets in the treatment of NSCLC metastasis.
Collapse
Affiliation(s)
- Zuzana Pastorkova
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Jozef Skarda
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Jozef Andel
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| |
Collapse
|
41
|
Kovářová N, Pecina P, Nůsková H, Vrbacký M, Zeviani M, Mráček T, Viscomi C, Houštěk J. Tissue- and species-specific differences in cytochrome c oxidase assembly induced by SURF1 defects. Biochim Biophys Acta Mol Basis Dis 2016; 1862:705-715. [PMID: 26804654 PMCID: PMC4793088 DOI: 10.1016/j.bbadis.2016.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 12/08/2015] [Accepted: 01/08/2016] [Indexed: 12/17/2022]
Abstract
Mitochondrial protein SURF1 is a specific assembly factor of cytochrome c oxidase (COX), but its function is poorly understood. SURF1 gene mutations cause a severe COX deficiency manifesting as the Leigh syndrome in humans, whereas in mice SURF1−/− knockout leads only to a mild COX defect. We used SURF1−/− mouse model for detailed analysis of disturbed COX assembly and COX ability to incorporate into respiratory supercomplexes (SCs) in different tissues and fibroblasts. Furthermore, we compared fibroblasts from SURF1−/− mouse and SURF1 patients to reveal interspecies differences in kinetics of COX biogenesis using 2D electrophoresis, immunodetection, arrest of mitochondrial proteosynthesis and pulse-chase metabolic labeling. The crucial differences observed are an accumulation of abundant COX1 assembly intermediates, low content of COX monomer and preferential recruitment of COX into I–III2–IVn SCs in SURF1 patient fibroblasts, whereas SURF1−/− mouse fibroblasts were characterized by low content of COX1 assembly intermediates and milder decrease in COX monomer, which appeared more stable. This pattern was even less pronounced in SURF1−/− mouse liver and brain. Both the control and SURF1−/− mice revealed only negligible formation of the I–III2–IVn SCs and marked tissue differences in the contents of COX dimer and III2–IV SCs, also less noticeable in liver and brain than in heart and muscle. Our studies support the view that COX assembly is much more dependent on SURF1 in humans than in mice. We also demonstrate markedly lower ability of mouse COX to form I–III2–IVn supercomplexes, pointing to tissue-specific and species-specific differences in COX biogenesis. In SURF1 −/− mouse the decrease of COX amount and activity was tissue/cell specific. Assembly kinetics proceeded to the level of stable COX monomer in SURF1 −/− mouse. COX assembly intermediates were faster degraded/depleted in time in SURF1 −/− mouse. COX was preferentially recruited in supercomplex I–III2–IV1 in SURF1 patient cells. Newly synthesized COX monomer was unstable and rapidly degraded in SURF1 patient.
Collapse
Affiliation(s)
- Nikola Kovářová
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague, Czech Republic
| | - Petr Pecina
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague, Czech Republic
| | - Hana Nůsková
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague, Czech Republic
| | - Marek Vrbacký
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague, Czech Republic
| | - Massimo Zeviani
- Molecular Neurogenetics Unit, Instituto Neurologico "C. Besta", via Temolo 4, 20126 Milan, Italy; MRC-Mitochondrial Biology Unit, Wellcome Trust MRC Bldg, Addenbrookes Hospital Hills Rd, Cambridge CB2 0XY, UK
| | - Tomáš Mráček
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague, Czech Republic
| | - Carlo Viscomi
- MRC-Mitochondrial Biology Unit, Wellcome Trust MRC Bldg, Addenbrookes Hospital Hills Rd, Cambridge CB2 0XY, UK
| | - Josef Houštěk
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, Prague, Czech Republic.
| |
Collapse
|
42
|
Cao Z, Zhang Y, Sun T, Zhang S, Yu W, Zhu J. Homocysteine induces cardiac hypertrophy by up-regulating ATP7a expression. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:12829-12836. [PMID: 26722473 PMCID: PMC4680418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/26/2015] [Indexed: 06/05/2023]
Abstract
AIMS The aim of the study is to investigate the molecular mechanism by which homocysteine (Hcy) induces cardiac hypertrophy. METHODS Primary cardiomyocytes were obtained from baby Sprague-Dawley rats within 3 days after birth. Flow cytometry was used to measure cell sizes. Quantitative real-time polymerase chain reaction was performed to measure the expression of β-myosin heavy chain and atrial natriuretic peptide genes. Western blotting assay was employed to determine ATP7a protein expression. Cytochrome C oxidase (COX) activity test was used to evaluate the activity of COX. Atomic absorption spectroscopy was performed to determine copper content. siRNAs were used to target-silence the expression of ATP7a. RESULTS Hcy induced cardiac hypertrophy and increased the expression of cardiac hypertrophy-related genes. ATP7a was a key factor in cardiac hypertrophy induced by Hcy. Reduced ATP7a expression inhibited cardiac hypertrophy induced by Hcy. Elevated ATP7a expression induced by Hcy inhibited COX activity. Enhanced ATP7a expression inhibited COX activity by lowering intracellular copper content. CONCLUSIONS Hcy elevates ATP7a protein expression, reduces copper content, and lowers COX activity, finally leading to cardiac hypertrophy.
Collapse
Affiliation(s)
- Zhanwei Cao
- Department of Cardiology, General Hospital of Pingmei Shenma Medical GroupPingdingshan 467000, P.R. China
| | - Yanzhou Zhang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450000, P.R. China
| | - Tongwen Sun
- Critical Care Medical Department, First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450000, P.R. China
| | - Shuguang Zhang
- Critical Care Medical Department, First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450000, P.R. China
| | - Weiya Yu
- Department of Cardiology, General Hospital of Pingmei Shenma Medical GroupPingdingshan 467000, P.R. China
| | - Jie Zhu
- Department of Intervention, Pingdingshan City First People’s HospitalPingdingshan 467000, P.R. China
| |
Collapse
|
43
|
Liddell JR. Targeting mitochondrial metal dyshomeostasis for the treatment of neurodegeneration. Neurodegener Dis Manag 2015; 5:345-64. [DOI: 10.2217/nmt.15.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial impairment and metal dyshomeostasis are suggested to be associated with many neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and Friedreich's ataxia. Treatments aimed at restoring metal homeostasis are highly effective in models of these diseases, and clinical trials hold promise. However, in general, the effect of these treatments on mitochondrial metal homeostasis is unclear, and the contribution of mitochondrial metal dyshomeostasis to disease pathogenesis requires further investigation. This review describes the role of metals in mitochondria in health, how mitochondrial metals are disrupted in neurodegenerative diseases, and potential therapeutics aimed at restoring mitochondrial metal homeostasis and function.
Collapse
Affiliation(s)
- Jeffrey R Liddell
- Department of Pathology, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
44
|
Stroud DA, Maher MJ, Lindau C, Vögtle FN, Frazier AE, Surgenor E, Mountford H, Singh AP, Bonas M, Oeljeklaus S, Warscheid B, Meisinger C, Thorburn DR, Ryan MT. COA6 is a mitochondrial complex IV assembly factor critical for biogenesis of mtDNA-encoded COX2. Hum Mol Genet 2015; 24:5404-15. [PMID: 26160915 DOI: 10.1093/hmg/ddv265] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/06/2015] [Indexed: 01/29/2023] Open
Abstract
Biogenesis of complex IV of the mitochondrial respiratory chain requires assembly factors for subunit maturation, co-factor attachment and stabilization of intermediate assemblies. A pathogenic mutation in COA6, leading to substitution of a conserved tryptophan for a cysteine residue, results in a loss of complex IV activity and cardiomyopathy. Here, we demonstrate that the complex IV defect correlates with a severe loss in complex IV assembly in patient heart but not fibroblasts. Complete loss of COA6 activity using gene editing in HEK293T cells resulted in a profound growth defect due to complex IV deficiency, caused by impaired biogenesis of the copper-bound mitochondrial DNA-encoded subunit COX2 and subsequent accumulation of complex IV assembly intermediates. We show that the pathogenic mutation in COA6 does not affect its import into mitochondria but impairs its maturation and stability. Furthermore, we show that COA6 has the capacity to bind copper and can associate with newly translated COX2 and the mitochondrial copper chaperone SCO1. Our data reveal that COA6 is intricately involved in the copper-dependent biogenesis of COX2.
Collapse
Affiliation(s)
- David A Stroud
- Department of Biochemistry and Molecular Biology, Monash University, Clayton 3800, Melbourne, Australia
| | - Megan J Maher
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Melbourne, Australia
| | - Caroline Lindau
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Melbourne, Australia
| | - F-Nora Vögtle
- Institut für Biochemie und Molekularbiologie, ZBMZ, University of Freiburg, 79104 Freiburg, Germany
| | - Ann E Frazier
- Murdoch Childrens Research Institute and Victorian Clinical Genetics Services, Royal Children's Hospital, University of Melbourne and Department of Pediatrics, University of Melbourne, 3052 Melbourne, Australia and
| | - Elliot Surgenor
- Department of Biochemistry and Molecular Biology, Monash University, Clayton 3800, Melbourne, Australia
| | - Hayley Mountford
- Murdoch Childrens Research Institute and Victorian Clinical Genetics Services, Royal Children's Hospital, University of Melbourne and Department of Pediatrics, University of Melbourne, 3052 Melbourne, Australia and
| | - Abeer P Singh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Melbourne, Australia
| | - Matteo Bonas
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Melbourne, Australia
| | - Silke Oeljeklaus
- Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology and BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - Bettina Warscheid
- Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology and BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - Chris Meisinger
- Institut für Biochemie und Molekularbiologie, ZBMZ, University of Freiburg, 79104 Freiburg, Germany, Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology and BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - David R Thorburn
- Institut für Biochemie und Molekularbiologie, ZBMZ, University of Freiburg, 79104 Freiburg, Germany
| | - Michael T Ryan
- Department of Biochemistry and Molecular Biology, Monash University, Clayton 3800, Melbourne, Australia,
| |
Collapse
|
45
|
Guo Q, Zhang H, Zhang L, He Y, Weng S, Dong Z, Wang J, Zhang P, Nao R. MicroRNA-21 regulates non-small cell lung cancer cell proliferation by affecting cell apoptosis via COX-19. Int J Clin Exp Med 2015; 8:8835-8841. [PMID: 26309536 PMCID: PMC4538100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 04/23/2015] [Indexed: 06/04/2023]
Abstract
AIMS This study is to investigate the regulatory effect of microRNA-21 (miR-21) on bone metastasis of non-small cell lung cancer (NSCLC). METHODS In this study, 18 patients were diagnosed with vertebral column metastasis of NSCLC. MiR-21 or small interfering RNAs were transfected into H2170 cells using Lipofectamine 2000. Real-time PCR was performed to detect miR-21 expression. Western blotting was used to measure the expression of COX-19 protein. Enzymatic activity tests were performed to measure the activity of cytochrome C oxidase. Flow cytometry was used to monitor changes in cell apoptotic rate. MTT assay was used to determine the capability of cell proliferation. RESULTS Bone metastasis of NSCLC enhanced the levels of miR-21 in NSCLC patients. Proliferation capability of cells with high expression of miR-21 was greater than that of cells with the inhibition of miR-21 expression. High expression of miR-21 promoted cell proliferation by inhibiting cell apoptosis. COX-19 was a key factor in the inhibition of apoptosis by miR-21. Inhibition of COX-19 expression reduced cell proliferation by enhancing cell apoptosis. CONCLUSIONS This study demonstrates that inhibition of miRNA-21 suppresses NSCLC cell proliferation by promoting cell apoptosis via the decrease of COX-19 expression.
Collapse
Affiliation(s)
- Qi Guo
- Orthopedics Department I, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Hui Zhang
- Orthopedics Department I, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Ling Zhang
- Orthopedics Department II, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Yang He
- Orthopedics Department I, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Shaobo Weng
- Department of Urology, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Ziping Dong
- Department of Emergency, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Jianmin Wang
- Orthopedics Department I, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Pingfang Zhang
- Orthopedics Department I, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| | - Ribu Nao
- Department of Emergency, General Hospital of North China Petroleum AdministrationRenqiu 062552, P.R. China
| |
Collapse
|
46
|
Bode M, Woellhaf MW, Bohnert M, van der Laan M, Sommer F, Jung M, Zimmermann R, Schroda M, Herrmann JM. Redox-regulated dynamic interplay between Cox19 and the copper-binding protein Cox11 in the intermembrane space of mitochondria facilitates biogenesis of cytochrome c oxidase. Mol Biol Cell 2015; 26:2385-401. [PMID: 25926683 PMCID: PMC4571295 DOI: 10.1091/mbc.e14-11-1526] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 04/24/2015] [Indexed: 01/02/2023] Open
Abstract
Members of the twin Cx9C protein family constitute the largest group of proteins in the intermembrane space (IMS) of mitochondria. Despite their conserved nature and their essential role in the biogenesis of the respiratory chain, the molecular function of twin Cx9C proteins is largely unknown. We performed a SILAC-based quantitative proteomic analysis to identify interaction partners of the conserved twin Cx9C protein Cox19. We found that Cox19 interacts in a dynamic manner with Cox11, a copper transfer protein that facilitates metalation of the Cu(B) center of subunit 1 of cytochrome c oxidase. The interaction with Cox11 is critical for the stable accumulation of Cox19 in mitochondria. Cox19 consists of a helical hairpin structure that forms a hydrophobic surface characterized by two highly conserved tyrosine-leucine dipeptides. These residues are essential for Cox19 function and its specific binding to a cysteine-containing sequence in Cox11. Our observations suggest that an oxidative modification of this cysteine residue of Cox11 stimulates Cox19 binding, pointing to a redox-regulated interplay of Cox19 and Cox11 that is critical for copper transfer in the IMS and thus for biogenesis of cytochrome c oxidase.
Collapse
Affiliation(s)
- Manuela Bode
- Cell Biology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Michael W Woellhaf
- Cell Biology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Maria Bohnert
- Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, 79104 Freiburg, Germany
| | - Martin van der Laan
- Institute of Biochemistry and Molecular Biology, ZBMZ, University of Freiburg, 79104 Freiburg, Germany BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Frederik Sommer
- Molecular Biotechnology and Systems Biology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Martin Jung
- Medical Biochemistry and Molecular Biology, Saarland University, 66424 Homburg, Germany
| | - Richard Zimmermann
- Medical Biochemistry and Molecular Biology, Saarland University, 66424 Homburg, Germany
| | - Michael Schroda
- Molecular Biotechnology and Systems Biology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | | |
Collapse
|
47
|
Cotruvo JA, Aron AT, Ramos-Torres KM, Chang CJ. Synthetic fluorescent probes for studying copper in biological systems. Chem Soc Rev 2015; 44:4400-14. [PMID: 25692243 DOI: 10.1039/c4cs00346b] [Citation(s) in RCA: 375] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The potent redox activity of copper is required for sustaining life. Mismanagement of its cellular pools, however, can result in oxidative stress and damage connected to aging, neurodegenerative diseases, and metabolic disorders. Therefore, copper homeostasis is tightly regulated by cells and tissues. Whereas copper and other transition metal ions are commonly thought of as static cofactors buried within protein active sites, emerging data points to the presence of additional loosely bound, labile pools that can participate in dynamic signalling pathways. Against this backdrop, we review advances in sensing labile copper pools and understanding their functions using synthetic fluorescent indicators. Following brief introductions to cellular copper homeostasis and considerations in sensor design, we survey available fluorescent copper probes and evaluate their properties in the context of their utility as effective biological screening tools. We emphasize the need for combined chemical and biological evaluation of these reagents, as well as the value of complementing probe data with other techniques for characterizing the different pools of metal ions in biological systems. This holistic approach will maximize the exciting opportunities for these and related chemical technologies in the study and discovery of novel biology of metals.
Collapse
Affiliation(s)
- Joseph A Cotruvo
- Department of Chemistry, University of California, Berkeley, CA 94720, USA.
| | | | | | | |
Collapse
|
48
|
Hlynialuk CJ, Ling B, Baker ZN, Cobine PA, Yu LD, Boulet A, Wai T, Hossain A, El Zawily AM, McFie PJ, Stone SJ, Diaz F, Moraes CT, Viswanathan D, Petris MJ, Leary SC. The Mitochondrial Metallochaperone SCO1 Is Required to Sustain Expression of the High-Affinity Copper Transporter CTR1 and Preserve Copper Homeostasis. Cell Rep 2015; 10:933-943. [PMID: 25683716 DOI: 10.1016/j.celrep.2015.01.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 12/15/2014] [Accepted: 01/08/2015] [Indexed: 02/03/2023] Open
Abstract
Human SCO1 fulfills essential roles in cytochrome c oxidase (COX) assembly and the regulation of copper (Cu) homeostasis, yet it remains unclear why pathogenic mutations in this gene cause such clinically heterogeneous forms of disease. Here, we establish a Sco1 mouse model of human disease and show that ablation of Sco1 expression in the liver is lethal owing to severe COX and Cu deficiencies. We further demonstrate that the Cu deficiency is explained by a functional connection between SCO1 and CTR1, the high-affinity transporter that imports Cu into the cell. CTR1 is rapidly degraded in the absence of SCO1 protein, and we show that its levels are restored in Sco1-/- mouse embryonic fibroblasts upon inhibition of the proteasome. These data suggest that mitochondrial signaling through SCO1 provides a post-translational mechanism to regulate CTR1-dependent Cu import into the cell, and they further underpin the importance of mitochondria in cellular Cu homeostasis.
Collapse
Affiliation(s)
| | - Binbing Ling
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Zakery N Baker
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Paul A Cobine
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Lisa D Yu
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Aren Boulet
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Timothy Wai
- Institute for Genetics, University of Cologne, 50931 Cologne, Germany
| | - Amzad Hossain
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Amr M El Zawily
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Faculty of Science, Damanhour University, Damanhour 22516, Egypt
| | - Pamela J McFie
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Scot J Stone
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Francisca Diaz
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carlos T Moraes
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Deepa Viswanathan
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Michael J Petris
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Scot C Leary
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
49
|
Kalderon B, Pines O. Protein folding as a driving force for dual protein targeting in eukaryotes. Front Mol Biosci 2014; 1:23. [PMID: 25988164 PMCID: PMC4428415 DOI: 10.3389/fmolb.2014.00023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/28/2014] [Indexed: 01/19/2023] Open
Abstract
It is well documented that in eukaryotic cells molecules of one protein can be located in several subcellular locations, a phenomenon termed dual targeting, dual localization, or dual distribution. The differently localized identical or nearly identical proteins are termed “echoforms.” Our conventional definition of dual targeted proteins refers to situations in which one of the echoforms is translocated through/into a membrane. Thus, dual targeted proteins are recognized by at least one organelle's receptors and translocation machineries within the lipid bilayer. In this review we attempt to evaluate mechanisms and situations in which protein folding is the major determinant of dual targeting and of the relative distribution levels of echoforms in the subcellular compartments of the eukaryotic cell. We show that the decisive folding step can occur prior, during or after translocation through the bilayer of a biological membrane. This phenomenon involves folding catalysts in the cell such as chaperones, proteases and modification enzymes, and targeting processes such as signal recognition, translocation through membranes, trapping, retrotranslocation and reverse translocation.
Collapse
Affiliation(s)
- Bella Kalderon
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Institute for Medical Research Israel-Canada, Hebrew University of Jerusalem Jerusalem, Israel
| | - Ophry Pines
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Institute for Medical Research Israel-Canada, Hebrew University of Jerusalem Jerusalem, Israel ; CREATE-NUS-HUJ Cellular and Molecular Mechanisms of Inflammation Program, National University of Singapore Singapore, Singapore
| |
Collapse
|
50
|
Garcia L, Welchen E, Gonzalez DH. Mitochondria and copper homeostasis in plants. Mitochondrion 2014; 19 Pt B:269-74. [PMID: 24582977 DOI: 10.1016/j.mito.2014.02.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 11/17/2022]
Abstract
Copper (Cu) and other transition metals are essential for living organisms but also toxic when present in excess. To cope with this apparent paradox, organisms have developed sophisticated mechanisms to acquire, transport and store these metals. Particularly, plant mitochondria require Cu for the assembly and function of cytochrome c oxidase (COX), the terminal enzyme of the respiratory chain. COX assembly is a complex process that requires the action of multiple factors, many of them involved in the delivery and insertion of Cu into the enzyme. In this review, we summarize what is known about the processes involved in Cu delivery to mitochondria and how these processes impact in Cu homeostasis at the cellular level. We also discuss evidence indicating that metallochaperones involved in COX assembly play additional roles in signaling pathways related to changes in Cu and redox homeostasis and the response of plants to stress. We propose that cysteine-rich proteins present in the mitochondrial intermembrane space are excellent candidates as sensors of these changes and transducers of signals originated in the organelle to the rest of the cell.
Collapse
Affiliation(s)
- Lucila Garcia
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CC 242 Paraje El Pozo, 3000 Santa Fe, Argentina
| | - Elina Welchen
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CC 242 Paraje El Pozo, 3000 Santa Fe, Argentina
| | - Daniel H Gonzalez
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CC 242 Paraje El Pozo, 3000 Santa Fe, Argentina.
| |
Collapse
|