1
|
Gadde R, Shah S, Böhlke M, Kim J, Betharia S. N,N'-bis(2-mercaptoethyl)isophthalamide (NBMI) as a novel chelator for Wilson's disease. Free Radic Biol Med 2025; 232:421-436. [PMID: 40032031 DOI: 10.1016/j.freeradbiomed.2025.02.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/15/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
Wilson's Disease (WD) is a rare autosomal recessive disorder caused by mutations in the ATP7B gene. These mutations lead to defective copper (Cu) transport and to accumulation of Cu in tissues, primarily in the liver and brain. Current treatment options such as D-penicillamine, trientine, and zinc salts focus on increasing Cu excretion or reducing Cu absorption, but often cause debilitating side effects. N,N'-bis(2-mercaptoethyl)isophthalamide (NBMI) is a lipophilic thiol-based compound originally developed for environmental decontamination. It has been shown to chelate toxic metals such as mercury, lead, and cadmium. This study was designed to evaluate the efficacy of NBMI to mitigate Cu overload using both in vitro and in vivo models of WD. HepG2 cells with the ATP7B gene knocked down had increased sensitivity to copper sulfate (CuSO4) compared to wild-type (WT) cells, validating the cell model for WD. Pretreatment with NBMI (2.5-50 μM) improved cell viability, reduced Cu-induced oxidative stress, decreased metallothionein levels, mitigated resulting DNA damage, and reduced overall levels of free intracellular Cu. In an established toxic milk mouse (tx-J) model of WD, 1% dietary NBMI effectively lowered hepatic, cerebral, and renal Cu levels. Treatment with 1% NBMI also improved liver function, as evidenced by reduced ALT levels and normalized hepatocyte morphology. Tx-J mice displayed higher liver-to-body weight ratios compared to WT mice, and treatment with 1% NBMI effectively reduced this ratio. While NBMI did not impact the elevated white blood cell counts and low platelet levels characteristic of tx-J mice, it also did not cause any detrimental effects on red blood cell, hemoglobin, and hematocrit levels. This dose of NBMI also restored homeostasis of other dysregulated essential metal ions in tx-J mice. These findings suggest that dietary administration of NBMI effectively chelates excess free Cu, ameliorates WD symptoms and offers a promising alternative to existing chelators.
Collapse
Affiliation(s)
- Rajitha Gadde
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, Boston, MA, USA
| | - Shrey Shah
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, Boston, MA, USA
| | - Mark Böhlke
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, Boston, MA, USA
| | - Jonghan Kim
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, MA, USA
| | - Swati Betharia
- Department of Pharmaceutical Sciences, MCPHS University, School of Pharmacy, Boston, MA, USA.
| |
Collapse
|
2
|
Greco V, Lanza V, Tomasello B, Naletova I, Cairns WRL, Sciuto S, Rizzarelli E. Copper Complexes with New Glycyl-l-histidyl-l-lysine-Hyaluronan Conjugates Show Antioxidant Properties and Osteogenic and Angiogenic Synergistic Effects. Bioconjug Chem 2025; 36:662-675. [PMID: 40123442 DOI: 10.1021/acs.bioconjchem.4c00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
In recent years, hyaluronic acid (HA) and the natural tripeptide glycyl-l-histidyl-l-lysine (GHK), especially its copper(II) complex (GHK-Cu), individually have been shown to exert helpful properties for bone protection and regeneration. However, they are not strong enough to handle oxidative stress, hydrolytic attack, or environmental conditions. Being aware that conjugation chemistry has recently emerged as an appealing approach for generating new molecular entities capable of preserving the molecular integrity of their moieties or delaying their degradation, herein we present the synthesis of conjugates of HA with GHK (GHK-HA), at different loadings of the tripeptide. GHK-HA binds copper(II) ions and potentiates the chemical and biological properties of the two components in in vitro assays. The results highlight copper's role in promoting the expression and release of certain trophic, angiogenic, and osteogenic factors, including brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), as well as bone morphogenetic protein-2 (BMP-2). The protective and regenerative activities of the metal ion are related to the translocation of its intracellular chaperones Copper Chaperone for Superoxide Dismutase (CCS) and Antioxidant-1 (Atox1) to the nucleus where they act as transcription factors.
Collapse
Affiliation(s)
- Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Valeria Lanza
- Institute of Crystallography, National Council of Research (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Irina Naletova
- Institute of Crystallography, National Council of Research (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Warren R L Cairns
- CNR-Institute of Polar Sciences (CNR-ISP), Via Torino 155, 30172 Venice, Italy
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Enrico Rizzarelli
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
- Institute of Crystallography, National Council of Research (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| |
Collapse
|
3
|
Sahoo SS, Manna D. Nanomaterial-Triggered Ferroptosis and Cuproptosis in Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412462. [PMID: 40018870 DOI: 10.1002/smll.202412462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Cancer remains one of the leading causes of the death of individuals globally. Conventional treatment techniques like chemotherapy and radiation often suffer various drawbacks like toxicity and drug resistance. The study of cell death has been predominantly focused on classical forms like apoptosis, but the role of metal ions in governing controlled cell death is a fascinating and less explored area. Metal-mediated controlled cell death is a process where metal triggers cell death via a unique mechanism. Nanomaterial-based strategies have gained attention for their ability to deliver precise therapeutic agents while also triggering Regulated Cell Death (RCD) mechanisms in cancer cells. The recently discovered metal-mediated controlled cell death techniques like cuproptosis and ferroptosis can be used in cancer treatment as they can be used selectively for the treatment of drug-resistant cancer. Nano material-based delivery system can also be used for the precise delivery of the drug to the targeted sites. In this review, we have given some idea about the mechanism of metal-mediated controlled cell death techniques (ferroptosis and cuproptosis) and how we can initiate controlled cell deaths using nanomaterials for cancer treatment.
Collapse
Affiliation(s)
- Suman Sekhar Sahoo
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| | - Debasish Manna
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
4
|
Sun R, Li M, Zhang T, Yang W, Yang L. Effects of Dietary Copper Sources and Levels on Liver Copper Metabolism and the Expression of Transporters in Growing Pigs. Animals (Basel) 2025; 15:526. [PMID: 40003008 PMCID: PMC11851888 DOI: 10.3390/ani15040526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Research on the effects of organic and inorganic Cu sources on metabolic processes and mechanisms in pigs is lacking. This study investigated the effects of different copper (Cu) sources and levels on hepatic Cu metabolism and transporter factors in growing pigs. Sixty healthy piglets (initial body weight 14.00 ± 0.30 kg) were randomly divided into four groups with five replicates of three pigs each. Four diets (AM, AH, BM, and BH) had different Cu sources [Cu sulphate (CuSO4): A and Cu amino acids (Cu-AA): B] and levels [supplemented (120 mg/kg DM): M, supplemented (240 mg/kg DM): H]. The pre-feeding period was 7 days, followed by a 45-day feeding period. Slaughter and sample collection were carried out on the 46th day of the formal feeding period. Significant differences were considered at p < 0.05. The final weight and average daily gain (ADG) of growing pigs in the Cu-AA groups were significantly higher than those in the CuSO4 groups. Serum Cu increased with increasing Cu supplementation on days 20 and 40. Cu concentrations in muscle, liver, and liver subcellular organelles were higher in Cu-AA groups. In the CuSO4 groups, Cu concentrations were higher in kidneys and faeces. In Cu-AA groups, both the Cu concentrations in lysosomes and cytosol were higher, and the activities of cathepsin D (CTSD), β-glucosidase (BGL), and acid phosphatase (ACP) in lysosomes and cytoplasm were higher. Comparisons between groups showed that liver mRNA of copper transporter protein 1 (CTR1), ATPase copper-transporting beta (ATP7B), ceruloplasmin (CP), antioxidant protein 1 (ATOX1), and metallothionein (MT) was lower in the CuSO4 group than in the Cu-AA group, with the best performance at 120 mg/kg Cu. mRNAs for ATPase copper-transporting alpha (ATP7A), cytochrome c oxidase copper chaperone 17 (COX17), and copper chaperone for superoxide dismutase (CCS) showed a decreasing trend in the Cu-AA groups. Cu-AA is better for Cu deposition, enhances the utilisation of Cu, reduces Cu excretion, and promotes the expression of relevant enzymes and transporters in the liver.
Collapse
Affiliation(s)
- Rui Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (R.S.); (M.L.); (T.Z.)
- Ministry of Education Laboratory of Animal Production and Security, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun 130118, China
| | - Meng Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (R.S.); (M.L.); (T.Z.)
- Ministry of Education Laboratory of Animal Production and Security, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun 130118, China
| | - Tianrui Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (R.S.); (M.L.); (T.Z.)
- Ministry of Education Laboratory of Animal Production and Security, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun 130118, China
| | - Wenyan Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (R.S.); (M.L.); (T.Z.)
- Ministry of Education Laboratory of Animal Production and Security, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun 130118, China
| | - Lianyu Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (R.S.); (M.L.); (T.Z.)
- Ministry of Education Laboratory of Animal Production and Security, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun 130118, China
| |
Collapse
|
5
|
Fitisemanu FM, Padilla-Benavides T. Emerging perspectives of copper-mediated transcriptional regulation in mammalian cell development. Metallomics 2024; 16:mfae046. [PMID: 39375833 PMCID: PMC11503025 DOI: 10.1093/mtomcs/mfae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Copper (Cu) is a vital micronutrient necessary for proper development and function of mammalian cells and tissues. Cu mediates the function of redox active enzymes that facilitate metabolic processes and signaling pathways. Cu levels are tightly regulated by a network of Cu-binding transporters, chaperones, and small molecule ligands. Extensive research has focused on the mammalian Cu homeostasis (cuprostasis) network and pathologies, which result from mutations and perturbations. There are roles for Cu-binding proteins as transcription factors (Cu-TFs) and regulators that mediate metal homeostasis through the activation or repression of genes associated with Cu handling. Emerging evidence suggests that Cu and some Cu-TFs may be involved in the regulation of targets related to development-expanding the biological roles of Cu-binding proteins. Cu and Cu-TFs are implicated in embryonic and tissue-specific development alongside the mediation of the cellular response to oxidative stress and hypoxia. Cu-TFs are also involved in the regulation of targets implicated in neurological disorders, providing new biomarkers and therapeutic targets for diseases such as Parkinson's disease, prion disease, and Friedreich's ataxia. This review provides a critical analysis of the current understanding of the role of Cu and cuproproteins in transcriptional regulation.
Collapse
|
6
|
Bizup B, Tzounopoulos T. On the genesis and unique functions of zinc neuromodulation. J Neurophysiol 2024; 132:1241-1254. [PMID: 39196675 PMCID: PMC11495185 DOI: 10.1152/jn.00285.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/30/2024] Open
Abstract
In addition to the essential structural and catalytic functions of zinc, evolution has adopted synaptic zinc as a neuromodulator. In the brain, synaptic zinc is released primarily from glutamatergic neurons, notably in the neocortex, hippocampus, amygdala, and auditory brainstem. In these brain areas, synaptic zinc is essential for neuronal and sensory processing fine-tuning. But what niche does zinc fill in neural signaling that other neuromodulators do not? Here, we discuss the evolutionary history of zinc as a signaling agent and its eventual adoption as an essential neuromodulator in the mammalian brain. We then attempt to describe the unique roles that zinc has carved out of the vast and diverse landscape of neuromodulators.
Collapse
Affiliation(s)
- Brandon Bizup
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
7
|
Chen X, Li K, Xiao Y, Wu W, Lin H, Qing X, Tian S, Liu S, Feng S, Wang B, Shao Z, Peng Y. SP1/CTR1-mediated oxidative stress-induced cuproptosis in intervertebral disc degeneration. Biofactors 2024; 50:1009-1023. [PMID: 38599595 DOI: 10.1002/biof.2052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 11/27/2023] [Indexed: 04/12/2024]
Abstract
Intervertebral disc degeneration (IDD) is an age-related disease and is responsible for low back pain. Oxidative stress-induced cell death plays a fundamental role in IDD pathogenesis. Cuproptosis is a recently discovered form of programmed cell death dependent on copper availability. Whether cuproptosis is involved in IDD progression remains unknown. Herein, we established in vitro and in vivo models to investigate cuproptosis in IDD and the mechanisms by which oxidative stress interacts with copper sensitivity in nucleus pulposus cells (NPCs). We found that ferredoxin-1 (FDX1) content increased in both rat and human degenerated discs. Sublethal oxidative stress on NPCs led to increased FDX1 expression, tricarboxylic acid (TCA) cycle-related proteins lipoylation and aggregation, and cell death in the presence of Cu2+ at physiological concentrations, while FDX1 knockdown inhibited cell death. Since copper homeostasis is involved in copper-induced cytotoxicity, we investigated the role of copper transport-related proteins, including importer (CTR1) and efflux pumps (ATPase transporter, ATP7A, and ATP7B). CTR1 and ATP7A content increased under oxidative stress, and blocking CTR1 reduced oxidative stress/copper-induced TCA-related protein aggregation and cell death. Moreover, oxidative stress promoted the expression of specific protein 1 (SP1) and SP1-mediated CTR1 transcription. SP1 inhibition decreased cell death rates, preserved disc hydration, and alleviated tissue degeneration. This suggests that oxidative stress upregulates FDX1 expression and copper flux through promoting SP1-mediated CTR1 transcription, leading to increased TCA cycle-related protein aggregation and cuproptosis. This study highlights the importance of cuproptosis in IDD progression and provides a promising therapeutic target for IDD treatment.
Collapse
Affiliation(s)
- Xuanzuo Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kanglu Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xiao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuo Tian
- Departments of Anesthesiology and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiqing Feng
- The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baichuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Maiti BK, Moura I, Moura JJG. Molybdenum-Copper Antagonism In Metalloenzymes And Anti-Copper Therapy. Chembiochem 2024; 25:e202300679. [PMID: 38205937 DOI: 10.1002/cbic.202300679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/23/2023] [Accepted: 01/11/2024] [Indexed: 01/12/2024]
Abstract
The connection between 3d (Cu) and 4d (Mo) via the "Mo-S-Cu" unit is called Mo-Cu antagonism. Biology offers case studies of such interactions in metalloproteins such as Mo/Cu-CO Dehydrogenases (Mo/Cu-CODH), and Mo/Cu Orange Protein (Mo/Cu-ORP). The CODH significantly maintains the CO level in the atmosphere below the toxic level by converting it to non-toxic CO2 for respiring organisms. Several models were synthesized to understand the structure-function relationship of these native enzymes. However, this interaction was first observed in ruminants, and they convert molybdate (MoO4 2- ) into tetrathiomolybdate (MoS4 2- ; TTM), reacting with cellular Cu to yield biological unavailable Mo/S/Cu cluster, then developing Cu-deficiency diseases. These findings inspire the use of TTM as a Cu-sequester drug, especially for treating Cu-dependent human diseases such as Wilson diseases (WD) and cancer. It is well known that a balanced Cu homeostasis is essential for a wide range of biological processes, but negative consequence leads to cell toxicity. Therefore, this review aims to connect the Mo-Cu antagonism in metalloproteins and anti-copper therapy.
Collapse
Affiliation(s)
- Biplab K Maiti
- Department of Chemistry, School of sciences, Cluster University of Jammu, Canal Road, Jammu, 180001, India
| | - Isabel Moura
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology (FCT NOVA), Universidade NOVA de Lisboa, Campus, de Caparica, Portugal
| | - José J G Moura
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology (FCT NOVA), Universidade NOVA de Lisboa, Campus, de Caparica, Portugal
| |
Collapse
|
9
|
Sanniyasi E, Gopal RK, Damodharan R, Thirumurugan T, Mahendran V. Bioaccumulation of Titanium in diatom Cyclotella atomus Hust. Biometals 2024; 37:71-86. [PMID: 37566151 DOI: 10.1007/s10534-023-00528-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
Diatomaceous earth or diatomite is a fossil rock deposit of diatoms made up of silica and other minerals. A distinguishing feature of diatoms that placed them in the single class of microalgae Bacillariophyceae, is the frustule, a transparent, hard-shelled cell wall. It's interesting to note that the diatom has specific proteins and enzymes for heavy metal detoxification and can intake and store more heavy metals in its frustule. Consequently, an attempt has been made in this study to determine the bioaccumulation of metals in the frustules of the diatom. Hence, a centric diatom was isolated from the freshwater sample collected from the Adyar River, Chennai, Tamil Nadu. The diameter of the cell was 5-7.5 µm and 20-23 striations with radial arrangement. A single, dark off-center fultoportula and marginal fultoportula on the striae are found in the diatom. Additionally, one rimoportula between two marginal fultoportula distributed on the striae between the costa was also seen. As a result, the isolated diatom was morphologically identified as Cyclotella atomus Hust. Simultaneously, the bioaccumulation study reveals that the Titanium (Ti) was found accumulated in the frustules of the diatom incubated in the Ti-supplemented culture medium based on the scanning electron microscope-energy-dispersive X-ray analysis (SEM-EDAX). Therefore, the biogenic accumulation and fabrication of Titanium frustules in diatom have advantages in enhancing the efficiency of solar cells.
Collapse
Affiliation(s)
- Elumalai Sanniyasi
- Department of Biotechnology, University of Madras, Guindy Campus, Chennai, 600 025, India.
| | - Rajesh Kanna Gopal
- Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 600077, India
| | - Rajesh Damodharan
- Department of Biotechnology, University of Madras, Guindy Campus, Chennai, 600 025, India
| | - Tarani Thirumurugan
- Department of Biotechnology, Rajalakshmi Engineering College (Autonomous), Thandalam, Chennai, 602 105, India
| | - Vishali Mahendran
- Department of Biotechnology, Rajalakshmi Engineering College (Autonomous), Thandalam, Chennai, 602 105, India
| |
Collapse
|
10
|
Lin YZ, Liu WH, Wu YP, Cai H, Zheng QS, Wei Y, Xu N, Xue XY. Revealing the potential of solute carrier family 31 (copper transporters), member 1: Insights into its role in bladder cancer progression and therapeutic implications. Int J Immunopathol Pharmacol 2024; 38:3946320241240706. [PMID: 38712735 PMCID: PMC11080779 DOI: 10.1177/03946320241240706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/26/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction: Bladder cancer represents a significant public health concern with diverse genetic alterations influencing disease onset, progression, and therapy response. In this study, we explore the multifaceted role of Solute Carrier Family 31 Member 1 (SLC31A1) in bladder cancer, a pivotal gene involved in copper homeostasis. Methods: Our research involved analyzing the SLC31A1 gene expression via RT-qPCR, promoter methylation via targeted bisulfite sequencing, and mutational status via Next Generation Sequencing (NGS) using the clinical samples sourced by the local bladder cancer patients. Later on, The Cancer Genome Atlas (TCGA) datasets were utilized for validation purposes. Moreover, prognostic significance, gene enrichment terms, and therapeutic drugs of SLC31A1 were also explored using KM Plotter, DAVID, and DrugBank databases. Results: We observed that SLC31A1 was significantly up-regulated at both the mRNA and protein levels in bladder cancer tissue samples, suggesting its potential involvement in bladder cancer development and progression. Furthermore, our investigation into the methylation status revealed that SLC31A1 was significantly hypomethylated in bladder cancer tissues, which may contribute to its overexpression. The ROC analysis of the SLC31A1 gene indicated promising diagnostic potential, emphasizing its relevance in distinguishing bladder cancer patients from normal individuals. However, it is crucial to consider other factors such as cancer stage, metastasis, and recurrence for a more accurate evaluation in the clinical context. Interestingly, mutational analysis of SLC31A1 demonstrated only benign mutations, indicating their unknown role in the SLC31A1 disruption. In addition to its diagnostic value, high SLC31A1 expression was associated with poorer overall survival (OS) in bladder cancer patients, shedding light on its prognostic relevance. Gene enrichment analysis indicated that SLC31A1 could influence metabolic and copper-related processes, further underscoring its role in bladder cancer. Lastly, we explored the DrugBank database to identify potential therapeutic agents capable of reducing SLC31A1 expression. Our findings unveiled six important drugs with the potential to target SLC31A1 as a treatment strategy. Conclusion: Our comprehensive investigation highlights SLC31A1 as a promising biomarker for bladder cancer development, progression, and therapy.
Collapse
Affiliation(s)
- Yun-Zhi Lin
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wei-hui Liu
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yu-Peng Wu
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hai Cai
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qing-Shui Zheng
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yong Wei
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ning Xu
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xue-Yi Xue
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Mi J, Luo J, Zeng H, Zhang H, Jamil M, Abdel-Maksoud MA, Zakri AM, Alfuraydi AA, Zhang N, Xiao M. Elucidating cuproptosis-related gene SLC31A1 diagnostic and prognostic values in cancer. Am J Transl Res 2023; 15:6026-6041. [PMID: 37969191 PMCID: PMC10641336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/28/2023] [Indexed: 11/17/2023]
Abstract
OBJECTIVES Cancer remains a global health challenge, necessitating the identification of novel biomarkers and therapeutic targets. Cuproptosis, a recently recognized form of cell death linked to copper metabolism, presents a promising avenue for anticancer strategies. We investigated the clinical significance of SLC31A1, a key regulator of cuproptosis, in multiple cancer types, aiming to elucidate its potential as a diagnostic biomarker, prognostic, indicator and therapeutic target. METHODS We conducted a pan-cancer analysis through TIMER2.0, evaluating SLC31A1 expression across multiple cancer types. Survival analysis was performed using KM plotter. Expression validation was carried out using UALCAN and Human Protein Atlas (HPA) databases. Methylation analysis was conducted with the help of ULACAN and OncoDB. Mutational analysis was performed using cBioPortal database. Immune infiltration analysis via the TIMER2.0 and gene enrichment analysis via the Metascape were performed to gain insights into the potential mechanisms underlying SLC31A1's role in cancer. Finally, Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was employed to confirm SLC31A1 expression in clinical samples. RESULTS Out of analyzed cancer, SLC31A1 exhibited significant up-regulation and correlation with worse overall survival (OS) across Breast Cancer (BRCA), Cervical Squamous Cell Carcinoma (CESC), Head and Neck Squamous Cell Carcinoma (HNSC), and Esophageal Carcinoma (ESCA). Mutational and promoter methylation analyses further revealed that hypomethylation is the major cause of SLC31A1 overexpression among BRCA, CESC, HNSC, and ESCA. Immune infiltration analysis showed significant associations between SLC31A1 expression and the presence of CD8+ T cells, CD4+ T cells, and macrophages in the tumor microenvironment. Gene enrichment analysis provided valuable insights into potential molecular pathways in context to BRCA, CESC, HNSC, and ESCA. Furthermore, when SLC31A1 was analyzed using clinical samples through RT-qPCR, this gene showed promising diagnostic potential, reflected by high Area Under the Curve (AUC) values. CONCLUSION Our pan-cancer study highlights the up-regulation of SLC31A1 and its correlation with worse OS in BRCA, CESC, HNSC, and ESCA. In sum, outcomes of this study showed that SLC31A1 could be a potential biomarker and novel therapeutic target of BRCA, CESC, HNSC, and ESCA.
Collapse
Affiliation(s)
- Jiaoping Mi
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-sen UniversityGuangzhou 510080, Guangdong, PR China
- Department of Otolaryngology Head and Neck Surgery, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai 519000, Guangdong, PR China
| | - Juncong Luo
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai 519000, Guangdong, PR China
| | - Huanwen Zeng
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai 519000, Guangdong, PR China
| | - Hongyu Zhang
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai 519000, Guangdong, PR China
| | - Muhammad Jamil
- PARC Arid Zone Research CenterDera Ismail Khan 29050, Pakistan
| | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Adel M Zakri
- Department of Plant Production, College of Food and Agricultural Sciences, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Akram A Alfuraydi
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ning Zhang
- Internal Medicine Oncology, Minhang Brunch Fudan University Shanghai Cancer CenterShanghai 200240, PR China
| | - Mei Xiao
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai 519000, Guangdong, PR China
| |
Collapse
|
12
|
Jiang Z, Sha G, Zhang W, Zhang Z, Liu T, Wang D, Tang D. The huge potential of targeting copper status in the treatment of colorectal cancer. Clin Transl Oncol 2023; 25:1977-1990. [PMID: 36781599 DOI: 10.1007/s12094-023-03107-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/28/2023] [Indexed: 02/15/2023]
Abstract
Colorectal cancer (CRC) commonly leads to cancer deaths and is often diagnosed at advanced stages. It also faces difficulties due to the poor results of conventional treatments such as surgery, chemotherapy, and radiotherapy. Copper is a mineral nutrient whose intrinsic properties have a two-way effect on the production and treatment of cancer. Copper's redox properties allow it to be used in developing anti-cancer drugs, while its potential toxicity leads to oxidative stress and even cancer. Copper status is closely related to colorectal tumors' proliferation and metastasis. The study of the mechanisms of copper homeostasis, cuproplasia, and cuproptosis due to altered copper status plays a crucial role in developing anticancer drugs. Therefore, targeting alteration of copper status becomes a potential option for treating colorectal cancer. This review summarizes the mechanisms by which altered copper status causes CRC progression and emphasizes the potential of regulating copper status in treating CRC.
Collapse
Affiliation(s)
- Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Zhilin Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Tian Liu
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225000, People's Republic of China.
| |
Collapse
|
13
|
Liao M, Li C, Hu C, Ding J. Copper-binding proteins genes set predicting the overall survival and immune infiltration in hepatocellular carcinoma by bioinformatic analysis. Biochem Biophys Rep 2023; 34:101466. [PMID: 37125079 PMCID: PMC10130086 DOI: 10.1016/j.bbrep.2023.101466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/19/2023] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
Abnormal Copper (Cu) accumulation shared a close association with hepatocellular carcinoma (HCC), but the regulatory role of Copper-binding proteins in HCC remains largely unknown. The aim of study was to identify the potential regulatory role of Cu-binding proteins, including copper homeostasis maintainer and the downstream effectors of Cu, in the progression of HCC. We conducted a comprehensive bioinformatic analysis of Cu-binding proteins in HCC using data from TCGA and ICGC database. Univariate cox regression analysis was conducted, and four prognostic Cu-binding proteins was identified to be differentially expressed between the normal liver tissues and HCC tissues. In addition, the Cu-binding proteins-based predictive signature (CuPscore) model was generated using the least absolute shrinkage and selection operator (LASSO) cox regression model. Here, we identified the crucial prognostic value of CuPscore in HCC. The pathological stage and CuPscore were independent risk factors for the prognosis of HCC patients. Pathological stage and CuPscore-based nomogram model exhibited great performance in predicting the prognosis of HCC patients. We also observed that the CuPscore shared a close association with several immunomodulatory molecules and the proportion of several tumor infiltrating immune cells, suggesting a potential value of CuPscore in predicting the response to immunotherapy in HCC. Our results demonstrated the prognostic value of Cu-binding proteins and its correlation with immune microenvironment in HCC, providing a therapeutic basis for the precision medicine strategy through targeting Cu-binding proteins in HCC.
Collapse
Affiliation(s)
- Manyu Liao
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Cong Li
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Clinical Center for Liver Cancer, Capital Medical University, Beijing, 100069, China
- Department of Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, 100069, China
- Corresponding author. Department of General Surgery, Beijing Youan Hospital, Capital Medical University, 100069, No. 8, West Toutiao, Outside You'anmen, Fengtai District, Beijing, China.
| | - Caixia Hu
- Center of Oncology and Minimally Invasive Intervention, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Jing Ding
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Clinical Center for Liver Cancer, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
14
|
Zhong CC, Zhao T, Hogstrand C, Song CC, Zito E, Tan XY, Xu YC, Song YF, Wei XL, Luo Z. Copper induces liver lipotoxicity disease by up-regulating Nrf2 expression via the activation of MTF-1 and inhibition of SP1/Fyn pathway. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166752. [PMID: 37182554 DOI: 10.1016/j.bbadis.2023.166752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 05/16/2023]
Abstract
Excessive copper (Cu) intake leads to hepatic lipotoxicity disease, which has adverse effects on health, but the underlying mechanism is unclear. We found that Cu increased lipotoxicity by promoting Nrf2 recruitment to the ARE site in the promoters of five lipogenic genes (g6pd, 6pgd, me, icdh and pparγ). We also found that Cu affected the Nrf2 expression via different pathways: metal regulatory transcription factor 1 (MTF-1) mediated the Cu-induced Nrf2 transcriptional activation; Cu also enhanced the expression of Nrf2 by inhibiting the SP1 expression, which was achieved by inhibiting the negative regulator Fyn of Nrf2. These promoted the enrichment of Nrf2 in the nucleus and ultimately affected lipotoxicity. Thus, for the first time, we elucidated that Cu induced liver lipotoxicity disease by up-regulating Nrf2 expression via the MTF-1 activation and the inhibition of SP1/Fyn pathway. Our study elucidates the Cu-associated obesity and NAFLD for fish and possibly humans.
Collapse
Affiliation(s)
- Chong-Chao Zhong
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Tao Zhao
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Christer Hogstrand
- Diabetes and Nutritional Sciences Division, School of Medicine, King's College London, London, UK
| | - Chang-Chun Song
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Xiao-Ying Tan
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Yi-Chuang Xu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Yu-Feng Song
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Xiao-Lei Wei
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Zhi Luo
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, Shandong Province, China.
| |
Collapse
|
15
|
Santiago-Díaz P, Rivero A, Rico M, González González A, González-Dávila M, Santana-Casiano M. Copper toxicity leads to accumulation of free amino acids and polyphenols in Phaeodactylum tricornutum diatoms. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:51261-51270. [PMID: 36809613 PMCID: PMC10104907 DOI: 10.1007/s11356-023-25939-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/10/2023] [Indexed: 04/16/2023]
Abstract
This work is focused on the effect of lethal and sub-lethal copper (Cu) concentrations on the free amino acid and polyphenol production by the marine diatom Phaeodactylum tricornutum (P. tricornutum) after 12, 18, and 21 days of exposure. The concentrations of 10 amino acids (arginine, aspartic acid, glutamic acid, histidine, lysine, methionine, proline, valine, isoleucine, and phenylalanine) and 10 polyphenols (gallic acid, protocatechuic acid, p-coumaric acid, ferulic acid, catechin, vanillic acid, epicatechin syringic acid, rutin, and gentisic acid) were measured by RP-HPLC. Under lethal doses of Cu, free amino acids reached levels significantly higher than those in the control cells (up to 21.9 times higher), where histidine and methionine showed the highest increases (up to 37.4 and 65.8 times higher, respectively). The total phenolic content also increased up to 11.3 and 5.59 times higher compared to the reference cells, showing gallic acid the highest increase (45.8 times greater). The antioxidant activities of cells exposed to Cu were also enhanced with increasing doses of Cu(II). They were evaluated by 2,2-diphenyl-1-picrylhydrazyl (DPPH) free radical scavenging ability (RSA), cupric ion reducing antioxidant capacity (CUPRAC), and ferric reducing antioxidant power (FRAP) assays. Malonaldehyde (MDA) exhibited the same tendency: cells grown at the highest lethal Cu concentration yielded the highest MDA level. These findings reflect the involvement of amino acids and polyphenols in protective mechanisms to overcome the toxicity of copper in marine microalgae.
Collapse
Affiliation(s)
- Paula Santiago-Díaz
- Departamento de Química, Facultad de Ciencias del Mar, Universidad de Las Palmas de Gran Canaria, Campus de Tafira, 35017, Las Palmas de Gran Canaria, Spain
- Instituto de Oceanografía Y Cambio Global (IOCAG), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Argimiro Rivero
- Departamento de Química, Facultad de Ciencias del Mar, Universidad de Las Palmas de Gran Canaria, Campus de Tafira, 35017, Las Palmas de Gran Canaria, Spain
- Instituto de Oceanografía Y Cambio Global (IOCAG), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Milagros Rico
- Departamento de Química, Facultad de Ciencias del Mar, Universidad de Las Palmas de Gran Canaria, Campus de Tafira, 35017, Las Palmas de Gran Canaria, Spain.
- Instituto de Oceanografía Y Cambio Global (IOCAG), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| | - Aridane González González
- Departamento de Química, Facultad de Ciencias del Mar, Universidad de Las Palmas de Gran Canaria, Campus de Tafira, 35017, Las Palmas de Gran Canaria, Spain
- Instituto de Oceanografía Y Cambio Global (IOCAG), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Melchor González-Dávila
- Departamento de Química, Facultad de Ciencias del Mar, Universidad de Las Palmas de Gran Canaria, Campus de Tafira, 35017, Las Palmas de Gran Canaria, Spain
- Instituto de Oceanografía Y Cambio Global (IOCAG), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Magdalena Santana-Casiano
- Departamento de Química, Facultad de Ciencias del Mar, Universidad de Las Palmas de Gran Canaria, Campus de Tafira, 35017, Las Palmas de Gran Canaria, Spain
- Instituto de Oceanografía Y Cambio Global (IOCAG), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
16
|
Squitti R, Catalli C, Gigante L, Marianetti M, Rosari M, Mariani S, Bucossi S, Mastromoro G, Ventriglia M, Simonelli I, Tondolo V, Singh P, Kumar A, Pal A, Rongioletti M. Non-Ceruloplasmin Copper Identifies a Subtype of Alzheimer’s Disease (CuAD): Characterization of the Cognitive Profile and Case of a CuAD Patient Carrying an RGS7 Stop-Loss Variant. Int J Mol Sci 2023; 24:ijms24076377. [PMID: 37047347 PMCID: PMC10094789 DOI: 10.3390/ijms24076377] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Alzheimer’s disease (AD) is a type of dementia whose cause is incompletely defined. Copper (Cu) involvement in AD etiology was confirmed by a meta-analysis on about 6000 participants, showing that Cu levels were decreased in AD brain specimens, while Cu and non-bound ceruloplasmin Cu (non-Cp Cu) levels were increased in serum/plasma samples. Non-Cp Cu was advocated as a stratification add-on biomarker of a Cu subtype of AD (CuAD subtype). To further circumstantiate this concept, we evaluated non-Cp Cu reliability in classifying subtypes of AD based on the characterization of the cognitive profile. The stratification of the AD patients into normal AD (non-Cp Cu ≤ 1.6 µmol/L) and CuAD (non-Cp Cu > 1.6 µmol/L) showed a significant difference in executive function outcomes, even though patients did not differ in disease duration and severity. Among the Cu-AD patients, a 76-year-old woman showed significantly abnormal levels in the Cu panel and underwent whole exome sequencing. The CuAD patient was detected with possessing the homozygous (c.1486T > C; p.(Ter496Argext*19) stop-loss variant in the RGS7 gene (MIM*602517), which encodes for Regulator of G Protein Signaling 7. Non-Cp Cu as an add-on test in the AD diagnostic pathway can provide relevant information about the underlying pathological processes in subtypes of AD and suggest specific therapeutic options.
Collapse
Affiliation(s)
- Rosanna Squitti
- Department of Laboratory Science, Research and Development Division, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
- Correspondence: rosanna.squitti.fw.@fbf-isola.it or
| | - Claudio Catalli
- Osakidetza Basque Health Service, Department of Genetics, Cruces University Hospital, 48903 Barakaldo, Spain
- Neuromuscular Disorders Research Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Laura Gigante
- Eurofins Genoma Group, Molecular Genetics Laboratory, 00138 Rome, Italy
| | - Massimo Marianetti
- Experimental Alzheimer Center, Fatebenefratelli Roman Province, 00189 Rome, Italy
| | - Mattia Rosari
- Experimental Alzheimer Center, Fatebenefratelli Roman Province, 00189 Rome, Italy
| | - Stefania Mariani
- Department of Laboratory Science, Research and Development Division, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
| | - Serena Bucossi
- Department of Laboratory Science, Research and Development Division, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
| | - Gioia Mastromoro
- Department of Laboratory Science, Research and Development Division, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
| | - Mariacarla Ventriglia
- Department of Laboratory Science, Research and Development Division, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
| | - Ilaria Simonelli
- Department of Laboratory Science, Research and Development Division, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
| | - Vincenzo Tondolo
- Digestive and Colorectal Surgery, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
- Digestive Surgery Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Parminder Singh
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh 160025, India
| | - Ashok Kumar
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh 160025, India
| | - Amit Pal
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Kalyani 741245, India
| | - Mauro Rongioletti
- Department of Laboratory Science, Research and Development Division, Fatebenefratelli Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
| |
Collapse
|
17
|
Kong FS, Ren CY, Jia R, Zhou Y, Chen JH, Ma Y. Systematic pan-cancer analysis identifies SLC31A1 as a biomarker in multiple tumor types. BMC Med Genomics 2023; 16:61. [PMID: 36973786 PMCID: PMC10041742 DOI: 10.1186/s12920-023-01489-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Solute Carrier Family 31 Member 1 (SLC31A1) has recently been identified as a cuproptosis-regulatory gene. Recent studies have indicated that SLC31A1 may play a role in colorectal and lung cancer tumorigenesis. However, the role of SLC31A1 and its cuproptosis-regulatory functions in multiple tumor types remains to be further elucidated. METHODS Online websites and datasets such as HPA, TIMER2, GEPIA, OncoVar, and cProSite were used to extract data on SLC31A1 in multiple cancers. DAVID and BioGRID were used to conduct functional analysis and construct the protein-protein interaction (PPI) network, respectively. The protein expression data of SLC31A1 was obtained from the cProSite database. RESULTS The Cancer Genome Atlas (TCGA) datasets showed increased SLC31A1 expression in tumor tissues compared with non-tumor tissues in most tumor types. In patients with tumor types including adrenocortical carcinoma, low-grade glioma, or mesothelioma, higher SLC31A1 expression was associated with shorter overall survival and disease-free survival. S105Y was the most prevalent point mutation in SLC31A1 in TCGA pan-cancer datasets. Moreover, SLC31A1 expression was positively correlated with the infiltration of immune cells such as macrophages and neutrophils in tumor tissues in several tumor types. Functional enrichment analysis showed that SLC31A1 co-expressed genes were involved in protein binding, integral components of the membrane, metabolic pathways, protein processing, and endoplasmic reticulum. Copper Chaperone For Superoxide Dismutase, Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha and Solute Carrier Family 31 Member 2 were copper homeostasis-regulated genes shown in the PPI network, and their expression was positively correlated with SLC31A1. Analysis showed there was a correlation between SLC31A1 protein and mRNA in various tumors. CONCLUSIONS These findings demonstrated that SLC31A1 is associated with multiple tumor types and disease prognosis. SLC31A1 may be a potential key biomarker and therapeutic target in cancers.
Collapse
Affiliation(s)
- Fan-Sheng Kong
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, China
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chun-Yan Ren
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Ruofan Jia
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, China
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuan Zhou
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, China
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Jian-Huan Chen
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China.
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China.
| | - Yaping Ma
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, China.
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
18
|
Wu M, Wang C, Ke L, Chen D, Qin Y, Han J. Correlation between copper speciation and transport pathway in Caco-2 cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:1895-1900. [PMID: 36287610 DOI: 10.1002/jsfa.12292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/11/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Previous studies have demonstrated that, in contrast to the properties of food-derived copper, water-derived copper exerts neurotoxic effects and exhibits different speciation during digestion. The cellular uptake efficiencies of different speciation of copper are distinct. However, it is unclear whether these different speciation share the same transport pathway in intestinal epithelial cells. In the present study, the intracellular accumulation of copper derived from copper ion and copper complex solutions was investigated in Caco-2 cells. RESULTS The cellular accumulation of copper derived from copper ions was higher than that of copper derived from the copper complex. Treatment with carboplatin and Ag+ , which are copper transporter receptor 1 (Ctr1, LC31A1) inhibitors, did not inhibit copper accumulation in Caco-2 cells, but inhibited copper accumulation in HepG2 cells. Zinc ion significantly decreased the intracellular copper content from 114 ± 7 μg g-1 protein to 88 ± 4 μg g-1 protein in the copper ion-treated Caco-2 cells, but not in the copper complex-treated Caco-2 cells (84.6 ± 14 μg g-1 protein versus 87.7 ± 20 μg g-1 protein, P > 0.05). Additionally, copper accumulation in Caco-2 and HepG2 cells significantly differed depending on different solvents (Hanks' balanced salt solution and NaNO3 , P < 0.05). CONCLUSION These results indicate that the intracellular accumulation of copper derived from copper ion and copper complex is mediated by distinct copper transport pathways. Copper speciation may be an important factor that affects copper absorption and toxicity. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Min Wu
- Hangzhou Vocational and Technical College, Ecology and Health Institute, Hangzhou, China
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Cong Wang
- Hangzhou Vocational and Technical College, Ecology and Health Institute, Hangzhou, China
| | - Leqin Ke
- Hangzhou Vocational and Technical College, Ecology and Health Institute, Hangzhou, China
| | - Dewen Chen
- Hangzhou Vocational and Technical College, Ecology and Health Institute, Hangzhou, China
| | - Yumei Qin
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Jianzhong Han
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| |
Collapse
|
19
|
Camponeschi F, Banci L. Metal trafficking in the cell: Combining atomic resolution with cellular dimension. FEBS Lett 2023; 597:122-133. [PMID: 36285633 DOI: 10.1002/1873-3468.14524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 01/14/2023]
Abstract
Metals are widely present in biological systems as simple ions or complex cofactors, and are involved in a variety of processes essential for life. Their transport inside cells and insertion into the binding sites of the proteins that need metals to function occur through complex and selective pathways involving dedicated multiprotein machineries specifically and transiently interacting with each other, often sharing the coordination of metal ions and/or cofactors. The understanding of these machineries requires integrated approaches, ranging from bioinformatics to experimental investigations, possibly in the cellular context. In this review, we report two case studies where the use of integrated in vitro and in cellulo approaches is necessary to clarify at atomic resolution essential aspects of metal trafficking in cells.
Collapse
Affiliation(s)
- Francesca Camponeschi
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy
| | - Lucia Banci
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy.,Department of Chemistry, University of Florence, Italy
| |
Collapse
|
20
|
Liu H, Kong Y, Liang X, Liu Z, Guo X, Yang B, Yin T, He H, Gou J, Zhang Y, Tang X. The treatment of hepatocellular carcinoma with SP94 modified asymmetrical bilayer lipid-encapsulated Cu(DDC) 2 nanoparticles facilitating Cu accumulation in the tumor. Expert Opin Drug Deliv 2023; 20:145-158. [PMID: 36462209 DOI: 10.1080/17425247.2023.2155631] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
BACKGROUND Copper diethyldithiocarbamate (Cu(DDC)2) has been demonstrated to possess excellent antitumor activity. However, the extremely poor water solubility of Cu(DDC)2 bring difficulty for its formulation research. In this study, we aim to develop a novel nanocarrier for Cu(DDC)2 delivery to overcome this obstacle and enhance antitumor activity. METHODS The SP94 modified asymmetrical bilayer lipid-encapsulated Cu(DDC)2 nanoparticles (DCDP) was established by combining the method of inverse microemulsion aggregation and thin-film dispersion. In vitro cellular assays and in vivo tumor-xenograft experiments were conducted to evaluate the tumor chemotherapeutic effect of DCDP. And the vital role of copper ions played in DSF or DDC (DSF/DDC)-based cancer chemotherapy was also explored. RESULTS DCDP with an encapsulation efficiency (EE%) of 74.0% were successfully prepared. SP94 modification facilitated cellular intake for DCDP, and promoted apoptosis to repress tumor cell proliferation (IC50, 200 nM). And DCDP effectively inhibited tumor growth with a high tumor inhibition rate of 74.84%. Furthermore, Cu(DDC)2 was found to facilitate the copper ion accumulation in tumor tissues, which is beneficial to therapy with high potency. CONCLUSION DCDP exhibited high-efficient tumor chemotherapeutic efficacy and provided a novel strategy for investigating the anticancer mechanism of Cu(DDC)2.
Collapse
Affiliation(s)
- Hao Liu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, China
| | - Yihan Kong
- Technology Research & Development Centre, Tianjin Pharmaceutical Research Institute Co., Ltd, Tianjin, China
| | - Xue Liang
- R&D & Innovation Committee, CSPC Pharmaceutical Group Limited, Shijiazhuang, China
| | - Zixu Liu
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, China
| | - Xueting Guo
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, China
| | - Bing Yang
- Department of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, China
| | - Tian Yin
- Department of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Haibing He
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, China
| | - Jingxin Gou
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, China
| | - Yu Zhang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, China
| | - Xing Tang
- Department of Pharmaceutics Science, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
21
|
Li C, Wu Y, Li H, Wang H, Liu JX. Lipid-related metabolism during zebrafish embryogenesis under unbalanced copper homeostasis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:1571-1586. [PMID: 36161547 DOI: 10.1007/s10695-022-01127-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/17/2022] [Indexed: 05/13/2023]
Abstract
Copper (Cu) is an essential trace element, playing an important role in lipid metabolism, and its transporters ATP7A and ATP7B, as Cu-transporting P-type ATPases, are involved in maintaining the Cu homeostasis in cells. Numerous studies in mammals have shown that Cu homeostasis and lipid metabolism are closely related, but studies on the link between the effects of excess Cu, ATP7A, and ATP7B on lipid metabolism during vertebrate embryogenesis are scarce. In this study, zebrafish disease models with Cu overload and ATP7A and ATP7B inactivation, respectively, were used to study the lipid metabolism-related differentially expressed genes (DEGs) which were enriched in the models. The dynamic and spatiotemporal expressions of the DEGs in WTs, atp7a-/-, and atp7b-/- mutants with or without Cu stress were unveiled in this study and they mostly distributed in brain at 24 hpf then in liver and intestine at 96 hpf, suggesting their potential roles in lipid and glycogen metabolism to apply energy for normal development in zebrafish. Meanwhile, the correlation analysis for the DEGs among the three groups unveiled that most of the DEGs were involved in the glyceride metabolism pathway. This is the first report to establish the relationship between atp7a and atp7b with Cu-stimulated intestinal and liver lipid metabolism during fish embryogenesis, and this study will provide a theoretical basis for fish embryonic development and lipid metabolism disorders under unbalanced copper homeostasis.
Collapse
Affiliation(s)
- ChangShun Li
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - You Wu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - HaoTian Li
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hai Wang
- Wuhan Zhihuiyuan Environmental Protection Technology, Co., Ltd, Wuhan, 430070, China
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
22
|
Schoeberl A, Gutmann M, Theiner S, Corte-Rodríguez M, Braun G, Vician P, Berger W, Koellensperger G. The copper transporter CTR1 and cisplatin accumulation at the single-cell level by LA-ICP-TOFMS. Front Mol Biosci 2022; 9:1055356. [PMID: 36518851 PMCID: PMC9742377 DOI: 10.3389/fmolb.2022.1055356] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/15/2022] [Indexed: 09/17/2023] Open
Abstract
More than a decade ago, studies on cellular cisplatin accumulation via active membrane transport established the role of the high affinity copper uptake protein 1 (CTR1) as a main uptake route besides passive diffusion. In this work, CTR1 expression, cisplatin accumulation and intracellular copper concentration was assessed for single cells revisiting the case of CTR1 in the context of acquired cisplatin resistance. The single-cell workflow designed for in vitro experiments enabled quantitative imaging at resolutions down to 1 µm by laser ablation-inductively coupled plasma-time-of-flight mass spectrometry (LA-ICP-TOFMS). Cisplatin-sensitive ovarian carcinoma cells A2780 as compared to the cisplatin-resistant subline A2780cis were investigated. Intracellular cisplatin and copper levels were absolutely quantified for thousands of individual cells, while for CTR1, relative differences of total CTR1 versus plasma membrane-bound CTR1 were determined. A markedly decreased intracellular cisplatin concentration accompanied by reduced copper concentrations was observed for single A2780cis cells, along with a distinctly reduced (total) CTR1 level as compared to the parental cell model. Interestingly, a significantly different proportion of plasma membrane-bound versus total CTR1 in untreated A2780 as compared to A2780cis cells was observed. This proportion changed in both models upon cisplatin exposure. Statistical analysis revealed a significant correlation between total and plasma membrane-bound CTR1 expression and cisplatin accumulation at the single-cell level in both A2780 and A2780cis cells. Thus, our study recapitulates the crosstalk of copper homeostasis and cisplatin uptake, and also indicates a complex interplay between subcellular CTR1 localization and cellular cisplatin accumulation as a driver for acquired resistance development.
Collapse
Affiliation(s)
- Anna Schoeberl
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Michael Gutmann
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sarah Theiner
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Mario Corte-Rodríguez
- Department of Physical and Analytical Chemistry, Faculty of Chemistry and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain
| | - Gabriel Braun
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Vician
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gunda Koellensperger
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
McCann C, Quinteros M, Adelugba I, Morgada MN, Castelblanco AR, Davis EJ, Lanzirotti A, Hainer SJ, Vila AJ, Navea JG, Padilla-Benavides T. The mitochondrial Cu+ transporter PiC2 (SLC25A3) is a target of MTF1 and contributes to the development of skeletal muscle in vitro. Front Mol Biosci 2022; 9:1037941. [PMID: 36438658 PMCID: PMC9682256 DOI: 10.3389/fmolb.2022.1037941] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
The loading of copper (Cu) into cytochrome c oxidase (COX) in mitochondria is essential for energy production in cells. Extensive studies have been performed to characterize mitochondrial cuproenzymes that contribute to the metallation of COX, such as Sco1, Sco2, and Cox17. However, limited information is available on the upstream mechanism of Cu transport and delivery to mitochondria, especially through Cu-impermeable membranes, in mammalian cells. The mitochondrial phosphate transporter SLC25A3, also known as PiC2, binds Cu+ and transports the ion through these membranes in eukaryotic cells, ultimately aiding in the metallation of COX. We used the well-established differentiation model of primary myoblasts derived from mouse satellite cells, wherein Cu availability is necessary for growth and maturation, and showed that PiC2 is a target of MTF1, and its expression is both induced during myogenesis and favored by Cu supplementation. PiC2 deletion using CRISPR/Cas9 showed that the transporter is required for proliferation and differentiation of primary myoblasts, as both processes are delayed upon PiC2 knock-out. The effects of PiC2 deletion were rescued by the addition of Cu to the growth medium, implying the deleterious effects of PiC2 knockout in myoblasts may be in part due to a failure to deliver sufficient Cu to the mitochondria, which can be compensated by other mitochondrial cuproproteins. Co-localization and co-immunoprecipitation of PiC2 and COX also suggest that PiC2 may participate upstream in the copper delivery chain into COX, as verified by in vitro Cu+-transfer experiments. These data indicate an important role for PiC2 in both the delivery of Cu to the mitochondria and COX, favoring the differentiation of primary myoblasts.
Collapse
|
24
|
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: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
BACKGROUND A 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. METHODS By 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. RESULTS ATP7B 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. CONCLUSION Copper 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
| |
Collapse
|
25
|
Decreased Expression of the Slc31a1 Gene and Cytoplasmic Relocalization of Membrane CTR1 Protein in Renal Epithelial Cells: A Potent Protective Mechanism against Copper Nephrotoxicity in a Mouse Model of Menkes Disease. Int J Mol Sci 2022; 23:ijms231911441. [PMID: 36232742 PMCID: PMC9570402 DOI: 10.3390/ijms231911441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
Kidneys play an especial role in copper redistribution in the organism. The epithelial cells of proximal tubules perform the functions of both copper uptake from the primary urine and release to the blood. These cells are equipped on their apical and basal membrane with copper transporters CTR1 and ATP7A. Mosaic mutant mice displaying a functional dysfunction of ATP7A are an established model of Menkes disease. These mice exhibit systemic copper deficiency despite renal copper overload, enhanced by copper therapy, which is indispensable for their life span extension. The aim of this study was to analyze the expression of Slc31a1 and Slc31a2 genes (encoding CTR1/CTR2 proteins) and the cellular localization of the CTR1 protein in suckling, young and adult mosaic mutants. Our results indicate that in the kidney of both intact and copper-injected 14-day-old mutants showing high renal copper content, CTR1 mRNA level is not up-regulated compared to wild-type mice given a copper injection. The expression of the Slc31a1 gene in 45-day-old mice is even reduced compared with intact wild-type animals. In suckling and young copper-injected mutants, the CTR1 protein is relocalized from the apical membrane to the cytoplasm of epithelial cells of proximal tubules, the process which prevents copper transport from the primary urine and, thus, protects cells against copper toxicity.
Collapse
|
26
|
Choi H, Oh D, Kim M, Cai L, Lee J, Kim E, Lee G, Hyun SH. Copper deficiency affects the developmental competence of porcine oocytes matured in vitro. Front Cell Dev Biol 2022; 10:993030. [PMID: 36158185 PMCID: PMC9490373 DOI: 10.3389/fcell.2022.993030] [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: 07/13/2022] [Accepted: 08/17/2022] [Indexed: 12/03/2022] Open
Abstract
The trace element Cu is required for the activity of various enzymes essential for physiological processes. In this study, we elucidated the copper transport system in porcine follicular cells and investigated the effect of Cu chelation during in vitro maturation (IVM) of porcine oocytes and subsequent embryonic development after parthenogenetic activation (PA). Cu chelation was induced by adding tetraethylenepentamine (TEPA) to the maturation media (TCM199-PVA). First, we identified the localization and relative levels of the copper transporter CTR1 in follicular cells. The level of CTR1 protein was the highest in mature cumulus cells; moreover, CTR1 was mainly localized in the cytoplasmic vesicular compartment in oocytes, whereas it was evenly distributed in the cytoplasm in cumulus cells. A total of 42 h after IVM, the TEPA-treated group showed reduced maturation rates compared to those of the control (p < 0.05). This negative effect of TEPA disappeared when it was added to the media with Cu (Cu + TEPA group). The TEPA treatment during IVM significantly increased the mRNA levels of the Has2 gene, which is related to cumulus expansion (p < 0.05). Both Cu supplementation and chelation significantly increased the reactive oxygen species (ROS) levels in porcine oocytes (p < 0.05). When we analyzed the transcript levels of folliculogenesis-related genes in Cu chelation conditions, only the expression of MAPK3 in cumulus cells significantly increased compared to that of the control. We also evaluated the subsequent embryonic development of PA embryos. TEPA-treated oocytes showed significantly decreased blastocyst formation rates compared to those of the control. The TEPA-induced toxic effect was alleviated when Cu was added with TEPA. Our findings suggest that the Cu transport system plays an important role in the porcine follicular development process and that the Cu deficiency negatively affects porcine oocyte maturation, as well as their subsequent developmental competence.
Collapse
Affiliation(s)
- Hyerin Choi
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, South Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Dongjin Oh
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, South Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Mirae Kim
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, South Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Lian Cai
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, South Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, South Korea
| | - Joohyeong Lee
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, South Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Eunhye Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Koreaa
| | - Gabsang Lee
- Department of Neurology, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sang-Hwan Hyun
- Veterinary Medical Center and College of Veterinary Medicine, Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Chungbuk National University, Cheongju, South Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, South Korea
- *Correspondence: Sang-Hwan Hyun,
| |
Collapse
|
27
|
Focarelli F, Giachino A, Waldron KJ. Copper microenvironments in the human body define patterns of copper adaptation in pathogenic bacteria. PLoS Pathog 2022; 18:e1010617. [PMID: 35862345 PMCID: PMC9302775 DOI: 10.1371/journal.ppat.1010617] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Copper is an essential micronutrient for most organisms that is required as a cofactor for crucial copper-dependent enzymes encoded by both prokaryotes and eukaryotes. Evidence accumulated over several decades has shown that copper plays important roles in the function of the mammalian immune system. Copper accumulates at sites of infection, including the gastrointestinal and respiratory tracts and in blood and urine, and its antibacterial toxicity is directly leveraged by phagocytic cells to kill pathogens. Copper-deficient animals are more susceptible to infection, whereas those fed copper-rich diets are more resistant. As a result, copper resistance genes are important virulence factors for bacterial pathogens, enabling them to detoxify the copper insult while maintaining copper supply to their essential cuproenzymes. Here, we describe the accumulated evidence for the varied roles of copper in the mammalian response to infections, demonstrating that this metal has numerous direct and indirect effects on immune function. We further illustrate the multifaceted response of pathogenic bacteria to the elevated copper concentrations that they experience when invading the host, describing both conserved and species-specific adaptations to copper toxicity. Together, these observations demonstrate the roles of copper at the host–pathogen interface and illustrate why bacterial copper detoxification systems can be viable targets for the future development of novel antibiotic drug development programs. Copper is required by both animals and bacteria in small quantities as a micronutrient. During infection, the mammalian immune system increases the local concentration of copper, which gives rise to copper toxicity in the pathogen. In turn, bacterial pathogens possess specialized systems to resist this copper toxicity. Copper also plays important, indirect roles in the function of the immune system. In this review, we explain the diverse roles of copper in the human body with a focus on its functions within the immune system. We also describe how bacterial pathogens respond to the copper toxicity that they experience within the host during infection, illustrating both conserved copper homeostasis and detoxification systems in bacteria and species-specific adaptations that have been shown to be important to pathogenicity. The key role of copper at the host–pathogen interface and the essential requirement for pathogenic bacteria to resist copper toxicity makes the protein components that confer resistance on pathogens potential targets for future development of novel antibiotic drugs.
Collapse
Affiliation(s)
- Francesca Focarelli
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrea Giachino
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kevin John Waldron
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
Structural elucidation and cytotoxicity profile of neocuproine-Cu(II) and Cu(I)-based chemotherapeutic agents: Effect of picric acid-derived cocrystals. Polyhedron 2022. [DOI: 10.1016/j.poly.2022.115848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
29
|
Kong L, Price NM. Light Stimulates Copper-Limited Growth of an Oceanic Diatom by Increasing Cellular Copper(II) Reduction─A Rate-Determining Step in Copper Uptake. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:9103-9111. [PMID: 35549243 DOI: 10.1021/acs.est.2c01479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Uptake of Cu by Thalassiosira oceanica requires that Cu(II) is reduced to Cu(I) prior to transport across the cell membrane. The reduction step is mediated biochemically by cellular reductases active with a broad range of Cu chemical species. Here, we report on the cellular Cu(II) reduction and Cu(I) uptake of a diatom under saturating and subsaturating irradiance. An increase in growth irradiance, from 50 to 400 μmol photons m-2 s-1, increased the rate of extracellular Cu(II) reduction and steady-state Cu uptake. Under these conditions, Cu-limited cells acquired Cu more efficiently and maintained faster rates of growth than Cu-limited cells in low light. Pseudo-first-order reaction rate constants were about 70-fold faster for Cu(I) uptake than for Cu(II) reduction so that reduction was the rate-determining step in Cu acquisition. Accordingly, steady-state Cu uptake rates predicted from the reduction rate constants agreed well with measured rates of Cu uptake obtained from cultures growing at low nanomolar Cu concentrations. Transcript abundance of putative Cu(II) reductases followed a similar pattern to cupric reductase activity, increasing in Cu-limited cells and with increasing growth irradiance. The results are significant in showing Cu(II) reduction as the rate-determining step in Cu uptake: they suggest that biologically mediated Cu(II) reduction may be an important part of the Cu cycle in surface waters of the open sea.
Collapse
Affiliation(s)
- Liangliang Kong
- Department of Biology, McGill University, Montréal, Québec H3A 1B1, Canada
- College of Marine Life Science, Ocean University of China, Qingdao 266001, Shandong, China
| | - Neil M Price
- Department of Biology, McGill University, Montréal, Québec H3A 1B1, Canada
| |
Collapse
|
30
|
Identification of Copper Transporter 1 as a Receptor for Feline Endogenous Retrovirus ERV-DC14. J Virol 2022; 96:e0022922. [PMID: 35652657 DOI: 10.1128/jvi.00229-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vertebrates harbor hundreds of endogenous retroviral (ERV) sequences in their genomes, which are considered signs of past infections that occurred during evolution. On rare occasions, ERV genes like env are maintained and coopted by hosts for physiological functions, but they also participate in recombination events with exogenous retroviruses to generate rearranged viruses with novel tropisms. In domestic cats, feline leukemia virus type D (FeLV-D) has been described as a recombinant virus between the infectious FeLV-A and likely the ERV-DC14 env gene that resulted in an extended tropism due to the usage of a new uncharacterized retroviral receptor. Here, we report the identification of SLC31A1 encoding the copper transporter 1 (CTR1) as a susceptibility gene for ERV-DC14 infection. Expression of human CTR1 into nonpermissive cells was sufficient to confer sensitivity to ERV-DC14 pseudotype infection and to increase the binding of an ERV-DC14 Env ligand. Moreover, inactivation of CTR1 by genome editing or cell surface downmodulation of CTR1 by a high dose of copper dramatically decreased ERV-DC14 infection and binding, while magnesium treatment had no effect. We also investigated the role of CTR1 in the nonpermissivity of feline and hamster cells. While feline CTR1 was fully functional for ERV-DC14, we found that binding was strongly reduced upon treatment with conditioned medium of feline cells, suggesting that the observed resistance to infection was a consequence of CTR1 saturation. In contrast, hamster CTR1 was inactive due to the presence of a N-linked glycosylation site at position 27, which is absent in the human ortholog. These results provide evidence that CTR1 is a receptor for ERV-DC14. Along with chimpanzee endogenous retrovirus type 2, ERV-DC14 is the second family of endogenous retrovirus known to have used CTR1 during past infections of vertebrates. IMPORTANCE Receptor usage is an important determinant of diseases induced by pathogenic retroviruses. In the case of feline leukemia viruses, three subgroups (A, B, and C) based on their ability to recognize different cell host receptors, respectively, the thiamine transporter THTR1, the phosphate transporter PiT1, and the heme exporter FLVCR1, are associated with distinct feline diseases. FeLV-A is horizontally transmitted and found in all naturally infected cats, while FeLV-B and FeLV-C have emerged from FeLV-A, respectively, by recombination with endogenous retroviral env sequences or by mutations in the FeLV-A env gene, both leading to a switch in receptor usage and in subsequent in vivo tropism. Here, we set up a genetic screen to identify the retroviral receptor of ERV-DC14, a feline endogenous provirus whose env gene has been captured by infectious FeLV-A to give rise to FeLV-D in a process similar to FeLV-B. Our results reveal that the copper transporter CTR1 was such a receptor and provide new insights into the acquisition of an expanded tropism by FeLV-D.
Collapse
|
31
|
Copper(II) import and reduction are dependent on His-Met clusters in the extracellular amino terminus of human copper transporter-1. J Biol Chem 2022; 298:101631. [PMID: 35090891 PMCID: PMC8867124 DOI: 10.1016/j.jbc.2022.101631] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/22/2022] Open
Abstract
Copper(I) is an essential metal for all life forms. Though Cu(II) is the most abundant and stable state, its reduction to Cu(I) via an unclear mechanism is prerequisite for its bioutilization. In eukaryotes, the copper transporter-1 (CTR1) is the primary high-affinity copper importer, although its mechanism and role in Cu(II) reduction remain uncharacterized. Here we show that extracellular amino-terminus of human CTR1 contains two methionine-histidine clusters and neighboring aspartates that distinctly bind Cu(I) and Cu(II) preceding its import. We determined that hCTR1 localizes at the basolateral membrane of polarized MDCK-II cells and that its endocytosis to Common-Recycling-Endosomes is regulated by reduction of Cu(II) to Cu(I) and subsequent Cu(I) coordination by the methionine cluster. We demonstrate the transient binding of both Cu(II) and Cu(I) during the reduction process is facilitated by aspartates that also act as another crucial determinant of hCTR1 endocytosis. Mutating the first Methionine cluster (7Met-Gly-Met9) and Asp13 abrogated copper uptake and endocytosis upon copper treatment. This phenotype could be reverted by treating the cells with reduced and nonreoxidizable Cu(I). We show that histidine clusters, on other hand, bind Cu(II) and are crucial for hCTR1 functioning at limiting copper. Finally, we show that two N-terminal His-Met-Asp clusters exhibit functional complementarity, as the second cluster is sufficient to preserve copper-induced CTR1 endocytosis upon complete deletion of the first cluster. We propose a novel and detailed mechanism by which the two His-Met-Asp residues of hCTR1 amino-terminus not only bind copper, but also maintain its reduced state, crucial for intracellular uptake.
Collapse
|
32
|
Wen MH, Xie X, Huang PS, Yang K, Chen TY. Crossroads between membrane trafficking machinery and copper homeostasis in the nerve system. Open Biol 2021; 11:210128. [PMID: 34847776 PMCID: PMC8633785 DOI: 10.1098/rsob.210128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Imbalanced copper homeostasis and perturbation of membrane trafficking are two common symptoms that have been associated with the pathogenesis of neurodegenerative and neurodevelopmental diseases. Accumulating evidence from biophysical, cellular and in vivo studies suggest that membrane trafficking orchestrates both copper homeostasis and neural functions-however, a systematic review of how copper homeostasis and membrane trafficking interplays in neurons remains lacking. Here, we summarize current knowledge of the general trafficking itineraries for copper transporters and highlight several critical membrane trafficking regulators in maintaining copper homeostasis. We discuss how membrane trafficking regulators may alter copper transporter distribution in different membrane compartments to regulate intracellular copper homeostasis. Using Parkinson's disease and MEDNIK as examples, we further elaborate how misregulated trafficking regulators may interplay parallelly or synergistically with copper dyshomeostasis in devastating pathogenesis in neurodegenerative diseases. Finally, we explore multiple unsolved questions and highlight the existing challenges to understand how copper homeostasis is modulated through membrane trafficking.
Collapse
Affiliation(s)
- Meng-Hsuan Wen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Xihong Xie
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Pei-San Huang
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Karen Yang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
33
|
Lutsenko S. Dynamic and cell-specific transport networks for intracellular copper ions. J Cell Sci 2021; 134:272704. [PMID: 34734631 DOI: 10.1242/jcs.240523] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Copper (Cu) homeostasis is essential for the development and function of many organisms. In humans, Cu misbalance causes serious pathologies and has been observed in a growing number of diseases. This Review focuses on mammalian Cu(I) transporters and highlights recent studies on regulation of intracellular Cu fluxes. Cu is used by essential metabolic enzymes for their activity. These enzymes are located in various intracellular compartments and outside cells. When cells differentiate, or their metabolic state is otherwise altered, the need for Cu in different cell compartments change, and Cu has to be redistributed to accommodate these changes. The Cu transporters SLC31A1 (CTR1), SLC31A2 (CTR2), ATP7A and ATP7B regulate Cu content in cellular compartments and maintain Cu homeostasis. Increasing numbers of regulatory proteins have been shown to contribute to multifaceted regulation of these Cu transporters. It is becoming abundantly clear that the Cu transport networks are dynamic and cell specific. The comparison of the Cu transport machinery in the liver and intestine illustrates the distinct composition and dissimilar regulatory response of their Cu transporters to changing Cu levels.
Collapse
Affiliation(s)
- Svetlana Lutsenko
- Johns Hopkins Medical Institutes, Department of Physiology, Baltimore, MD 21205, USA
| |
Collapse
|
34
|
Tabbì G, Cucci LM, Pinzino C, Munzone A, Marzo T, Pizzanelli S, Satriano C, Magrì A, La Mendola D. Peptides Derived from Angiogenin Regulate Cellular Copper Uptake. Int J Mol Sci 2021; 22:9530. [PMID: 34502439 PMCID: PMC8430698 DOI: 10.3390/ijms22179530] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/31/2022] Open
Abstract
The angiogenin protein (ANG) is one of the most potent endogenous angiogenic factors. In this work we characterized by means of potentiometric, spectroscopic and voltammetric techniques, the copper complex species formed with peptide fragments derived from the N-terminal domain of the protein, encompassing the sequence 1-17 and having free amino, Ang1-17, or acetylated N-terminus group, AcAng1-17, so to explore the role of amino group in metal binding and cellular copper uptake. The obtained data show that amino group is the main copper anchoring site for Ang1-17. The affinity constant values, metal coordination geometry and complexes redox-potentials strongly depend, for both peptides, on the number of copper equivalents added. Confocal laser scanning microscope analysis on neuroblastoma cells showed that in the presence of one equivalent of copper ion, the free amino Ang1-17 increases cellular copper uptake while the acetylated AcAng1-17 strongly decreases the intracellular metal level. The activity of peptides was also compared to that of the protein normally present in the plasma (wtANG) as well as to the recombinant form (rANG) most commonly used in literature experiments. The two protein isoforms bind copper ions but with a different coordination environment. Confocal laser scanning microscope data showed that the wtANG induces a strong increase in intracellular copper compared to control while the rANG decreases the copper signal inside cells. These data demonstrate the relevance of copper complexes' geometry to modulate peptides' activity and show that wtANG, normally present in the plasma, can affect cellular copper uptake.
Collapse
Affiliation(s)
- Giovanni Tabbì
- Institute of Crystallography—National Council of Research—CNR, via Paolo Gaifami 18, 95126 Catania, Italy;
| | - Lorena Maria Cucci
- Nano Hybrid BioInterfaces Lab (NHBIL), Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy;
| | - Calogero Pinzino
- Institute for the Chemistry of OrganoMetallic Compounds (ICCOM), National Council of Research—CNR, via G. Moruzzi 1, 56124 Pisa, Italy;
| | - Alessia Munzone
- Aix-Marseille Univesité, 52 Avenue Escadrille Normandie Niemen, 13013 Marseille, France;
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy;
| | - Silvia Pizzanelli
- Institute for the Chemistry of OrganoMetallic Compounds (ICCOM), National Council of Research—CNR, via G. Moruzzi 1, 56124 Pisa, Italy;
| | - Cristina Satriano
- Nano Hybrid BioInterfaces Lab (NHBIL), Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy;
| | - Antonio Magrì
- Institute of Crystallography—National Council of Research—CNR, via Paolo Gaifami 18, 95126 Catania, Italy;
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy;
| |
Collapse
|
35
|
Maung MT, Carlson A, Olea-Flores M, Elkhadragy L, Schachtschneider KM, Navarro-Tito N, Padilla-Benavides T. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J 2021; 35:e21810. [PMID: 34390520 DOI: 10.1096/fj.202100273rr] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022]
Abstract
Copper (Cu) is an essential micronutrient required for the activity of redox-active enzymes involved in critical metabolic reactions, signaling pathways, and biological functions. Transporters and chaperones control Cu ion levels and bioavailability to ensure proper subcellular and systemic Cu distribution. Intensive research has focused on understanding how mammalian cells maintain Cu homeostasis, and how molecular signals coordinate Cu acquisition and storage within organs. In humans, mutations of genes that regulate Cu homeostasis or facilitate interactions with Cu ions lead to numerous pathologic conditions. Malfunctions of the Cu+ -transporting ATPases ATP7A and ATP7B cause Menkes disease and Wilson disease, respectively. Additionally, defects in the mitochondrial and cellular distributions and homeostasis of Cu lead to severe neurodegenerative conditions, mitochondrial myopathies, and metabolic diseases. Cu has a dual nature in carcinogenesis as a promotor of tumor growth and an inducer of redox stress in cancer cells. Cu also plays role in cancer treatment as a component of drugs and a regulator of drug sensitivity and uptake. In this review, we provide an overview of the current knowledge of Cu metabolism and transport and its relation to various human pathologies.
Collapse
Affiliation(s)
- May T Maung
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Alyssa Carlson
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Monserrat Olea-Flores
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | - Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Napoleon Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | | |
Collapse
|
36
|
Meng T, Gao L, Xie C, Xiang Y, Huang Y, Zhang Y, Wu X. Manganese methionine hydroxy analog chelated affects growth performance, trace element deposition and expression of related transporters of broilers. ACTA ACUST UNITED AC 2021; 7:481-487. [PMID: 34258436 PMCID: PMC8245798 DOI: 10.1016/j.aninu.2020.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 11/27/2022]
Abstract
The present study aimed to evaluate the effects of manganese methionine hydroxyl analog chelated (Mn-MHAC) as a manganese (Mn) source on growth performance and trace element deposition in broilers. A total of 432 Arbor Acres commercial female broilers were fed a basal corn-soybean diet containing Mn at 25.64 mg/kg diet for 10 d. They were then randomly assigned to 6 groups, including a control group (the basal diet), a Mn sulfate group (the basal diet supplemented with Mn at 100 mg/kg diet), and 4 Mn-MHAC groups (the basal diet supplemented with Mn-MHAC at 25, 50, 75 and 100 mg Mn/kg diet, respectively). The results showed that compared with the control group, groups supplemented with Mn-MHAC had a positive effect on BW (quadratic, P = 0.017) and ADG (quadratic, P = 0.017). Moreover, the Mn-MHAC (50 mg Mn/kg diet) group had significantly greater BW and ADG (P < 0.05) compared with the other Mn-MHAC groups. Trace element deposition results also showed that tibial Mn increased (linear or quadratic, P = 0.002 and 0.009, respectively) in groups fed diets with increased levels of Mn-MHAC. In contrast, Fe deposition decreased both in the heart (linear, P = 0.020) and tibia (P < 0.05). In addition, the Mn-MHAC supplement noticeably lowered serum Mn-SOD activity (linear or quadratic, P = 0.048 and 0.019, respectively). The relative mRNA levels of divalent metal transporter 1 (DMT1) (P = 0.024), ferroportin 1 (FPN1) (P = 0.049), and Cu transporter-1(CTR1) (P < 0.001) in the duodenum, as well as CTR1 in the jejunum and ileum (P = 0.040 and 0.011, respectively) were higher in the Mn-supplemented group than in the control group. Furthermore, the relative mRNA level of DMT1 in the jejunum and ileum of broilers in the Mn-MHAC group (50 mg Mn/kg diet) did not differ from those in the control group, but was lower than those in the Mn sulfate group (P < 0.05). In conclusion, Mn-MHAC dietary supplementation improved the growth performance and trace element deposition in broilers. From this study, we recommend the optimum Mn-MHAC level to meet the Mn requirement of broilers is 50 to 75 mg Mn/kg diet.
Collapse
Affiliation(s)
- Tiantian Meng
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China.,Hunan Co-Innovation Center of Safety Animal Production, College of Animal Science and Technology; College of Resources and Environment, Hunan Agricultural University, Changsha, 410128, China
| | - Lumin Gao
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China
| | - Chunyan Xie
- Hunan Co-Innovation Center of Safety Animal Production, College of Animal Science and Technology; College of Resources and Environment, Hunan Agricultural University, Changsha, 410128, China
| | - Yangkui Xiang
- Hunan Provincial Research Center of Mineral Element Nutrition Engineering Technology, Xing-Jia Bio-engineering Co., Ltd., 410300, Changsha, China
| | - Yiqiang Huang
- Hunan Provincial Research Center of Mineral Element Nutrition Engineering Technology, Xing-Jia Bio-engineering Co., Ltd., 410300, Changsha, China
| | - Yawei Zhang
- Hunan Provincial Research Center of Mineral Element Nutrition Engineering Technology, Xing-Jia Bio-engineering Co., Ltd., 410300, Changsha, China
| | - Xin Wu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, 410125, China.,Hunan Co-Innovation Center of Safety Animal Production, College of Animal Science and Technology; College of Resources and Environment, Hunan Agricultural University, Changsha, 410128, China.,Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| |
Collapse
|
37
|
|
38
|
Ladeira LCM, Dos Santos EC, Valente GE, da Silva J, Santos TA, Dos Santos Costa Maldonado IR. Could biological tissue preservation methods change chemical elements proportion measured by energy dispersive X-ray spectroscopy? Biol Trace Elem Res 2020; 196:168-172. [PMID: 31654256 DOI: 10.1007/s12011-019-01909-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/16/2019] [Indexed: 12/27/2022]
Abstract
Energy dispersive X-ray spectroscopy (EDS) is a powerful technical tool used in the biomedical field to investigate the proportion of chemical elements of interest in research, such as heavy metal bioaccumulation and the enzymatic cofactors and nanoparticle therapy in various pathologies. However, the correct evaluation of the proportion of the elements is subject to some factors, including the method of sample preservation. In this study, we seek to investigate the effect of biological tissue preservation methods on the proportion of chemical elements obtained by the EDS methodology. For such, we used EDS to measure the proportion of chemical elements with biomedical interest in preserved livers, using three common methods for preserving biological tissues: (a) freezing, (b) paraformaldehyde fixative solution, and (c) Karnovsky solution. We found an increased level of sodium and reduced contents of potassium and copper in samples fixed in fixative solutions, when compared to frozen samples (p < 0.05). Our data indicate that preservation methods can change the proportion of chemical elements in biological samples, when measured by EDS. Frozen preservation should be preferred to retain the actual chemical content of samples and allow a correct assessment of the proportion of their elements.
Collapse
Affiliation(s)
- Luiz Carlos Maia Ladeira
- Departamento de Biologia Geral, Universidade Federal de Viçosa, Av. P.H. Rolfs, s/n, Campus Universitário, Viçosa, 36570-900, Brazil.
| | | | | | - Janaina da Silva
- Departamento de Biologia Geral, Universidade Federal de Viçosa, Av. P.H. Rolfs, s/n, Campus Universitário, Viçosa, 36570-900, Brazil
| | - Talita Amorim Santos
- Departamento de Biologia Geral, Universidade Federal de Viçosa, Av. P.H. Rolfs, s/n, Campus Universitário, Viçosa, 36570-900, Brazil
| | | |
Collapse
|
39
|
Guthrie LM, Soma S, Yuan S, Silva A, Zulkifli M, Snavely TC, Greene HF, Nunez E, Lynch B, De Ville C, Shanbhag V, Lopez FR, Acharya A, Petris MJ, Kim BE, Gohil VM, Sacchettini JC. Elesclomol alleviates Menkes pathology and mortality by escorting Cu to cuproenzymes in mice. Science 2020; 368:620-625. [PMID: 32381719 DOI: 10.1126/science.aaz8899] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/10/2020] [Accepted: 03/19/2020] [Indexed: 12/23/2022]
Abstract
Loss-of-function mutations in the copper (Cu) transporter ATP7A cause Menkes disease. Menkes is an infantile, fatal, hereditary copper-deficiency disorder that is characterized by progressive neurological injury culminating in death, typically by 3 years of age. Severe copper deficiency leads to multiple pathologies, including impaired energy generation caused by cytochrome c oxidase dysfunction in the mitochondria. Here we report that the small molecule elesclomol escorted copper to the mitochondria and increased cytochrome c oxidase levels in the brain. Through this mechanism, elesclomol prevented detrimental neurodegenerative changes and improved the survival of the mottled-brindled mouse-a murine model of severe Menkes disease. Thus, elesclomol holds promise for the treatment of Menkes and associated disorders of hereditary copper deficiency.
Collapse
Affiliation(s)
- Liam M Guthrie
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Shivatheja Soma
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Sai Yuan
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Andres Silva
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Mohammad Zulkifli
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Thomas C Snavely
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Hannah Faith Greene
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Elyssa Nunez
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Brogan Lynch
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Courtney De Ville
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Vinit Shanbhag
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Franklin R Lopez
- Texas Veterinary Medicine Diagnostic Laboratory, College Station, TX 77843, USA
| | - Arjun Acharya
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Michael J Petris
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Byung-Eun Kim
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Vishal M Gohil
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA.
| | - James C Sacchettini
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
40
|
Boyd SD, Ullrich MS, Skopp A, Winkler DD. Copper Sources for Sod1 Activation. Antioxidants (Basel) 2020; 9:antiox9060500. [PMID: 32517371 PMCID: PMC7346115 DOI: 10.3390/antiox9060500] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
Copper ions (i.e., copper) are a critical part of several cellular processes, but tight regulation of copper levels and trafficking are required to keep the cell protected from this highly reactive transition metal. Cu, Zn superoxide dismutase (Sod1) protects the cell from the accumulation of radical oxygen species by way of the redox cycling activity of copper in its catalytic center. Multiple posttranslational modification events, including copper incorporation, are reliant on the copper chaperone for Sod1 (Ccs). The high-affinity copper uptake protein (Ctr1) is the main entry point of copper into eukaryotic cells and can directly supply copper to Ccs along with other known intracellular chaperones and trafficking molecules. This review explores the routes of copper delivery that are utilized to activate Sod1 and the usefulness and necessity of each.
Collapse
|
41
|
Reyes ME, de La Fuente M, Hermoso M, Ili CG, Brebi P. Role of CC Chemokines Subfamily in the Platinum Drugs Resistance Promotion in Cancer. Front Immunol 2020; 11:901. [PMID: 32499779 PMCID: PMC7243460 DOI: 10.3389/fimmu.2020.00901] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is a significant medical issue, being one of the main causes of mortality around the world. The therapies for this pathology depend on the stage in which the cancer is found, but it is usually diagnosed at an advanced stage in which the treatment is chemotherapy. Platinum drugs are among the most commonly used in therapy, unfortunately, one of the main obstacles to this treatment is the development of chemoresistance, which is the ability of cancer cells to evade the effects of drugs. Although some molecular mechanisms involved in resistance to platinum drugs are described, elucidation is still required of others. Secretion of inflammatory mediators such as cytokines and chemokines, by tumor microenvironment components or tumor cells, show direct influence on proliferation, metastasis and progression of cancer and are related to chemoresistance and poor prognosis. In this review, the general mechanisms associated with resistance to platinum drugs, inflammation on cancer development and chemoresistance in various types of cancer will be approached with special emphasis on the current history of CC chemokines subfamily-mediated chemoresistance.
Collapse
Affiliation(s)
- Maria E. Reyes
- Laboratorio de Biología Integrativa (LIBi), Centro de Excelencia en Medicina Traslacional-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de la Frontera, Temuco, Chile
| | - Marjorie de La Fuente
- Laboratorio de Inmunidad Innata, Programa de Inmunología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Dirección Académica, Clínica Las Condes, Santiago, Chile
| | - Marcela Hermoso
- Laboratorio de Inmunidad Innata, Programa de Inmunología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carmen G. Ili
- Laboratorio de Biología Integrativa (LIBi), Centro de Excelencia en Medicina Traslacional-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de la Frontera, Temuco, Chile
| | - Priscilla Brebi
- Laboratorio de Biología Integrativa (LIBi), Centro de Excelencia en Medicina Traslacional-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de la Frontera, Temuco, Chile
| |
Collapse
|
42
|
Lin W, Xu L, Li G. Molecular Insights Into Lysyl Oxidases in Cartilage Regeneration and Rejuvenation. Front Bioeng Biotechnol 2020; 8:359. [PMID: 32426343 PMCID: PMC7204390 DOI: 10.3389/fbioe.2020.00359] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Articular cartilage remains among the most difficult tissues to regenerate due to its poor self-repair capacity. The lysyl oxidase family (LOX; also termed as protein-lysine 6-oxidase), mainly consists of lysyl oxidase (LO) and lysyl oxidase-like 1-4 (LOXL1-LOXL4), has been traditionally defined as cuproenzymes that are essential for stabilization of extracellular matrix, particularly cross-linking of collagen and elastin. LOX is essential in the musculoskeletal system, particularly cartilage. LOXs-mediated collagen cross-links are essential for the functional integrity of articular cartilage. Appropriate modulation of the expression or activity of certain LOX members selectively may become potential promising strategy for cartilage repair. In the current review, we summarized the advances of LOX in cartilage homeostasis and functioning, as well as copper-mediated activation of LOX through hypoxia-responsive signaling axis during recent decades. Also, the molecular signaling network governing LOX expression has been summarized, indicating that appropriate modulation of hypoxia-responsive-signaling-directed LOX expression through manipulation of bioavailability of copper and oxygen is promising for further clinical implications of cartilage regeneration, which has emerged as a potential therapeutic approach for cartilage rejuvenation in tissue engineering and regenerative medicine. Therefore, targeted regulation of copper-mediated hypoxia-responsive signalling axis for selective modulation of LOX expression may become potential effective therapeutics for enhanced cartilage regeneration and rejuvenation in future clinical implications.
Collapse
Affiliation(s)
- Weiping Lin
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Liangliang Xu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.,MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
43
|
Abstract
Copper accumulation and deficiency are reciprocally connected to lipid metabolism. In Wilson disease (WD), which is caused by a genetic loss of function of the copper-transporting P-type ATPase beta, copper accumulates mainly in the liver and lipid metabolism is dysregulated. The underlying mechanisms linking copper and lipid metabolism in WD are not clear. Copper may impair metabolic machinery by direct binding to protein and lipid structures or by generating reactive oxygen species with consequent damage to cellular organelles vital to energy metabolism. In the liver, copper overload results in mitochondrial impairment, down-regulation of lipid metabolism, and the development of steatosis with an etiology not fully elucidated. Little is known regarding the effect of copper overload on extrahepatic energy homeostasis. This review aims to discuss alterations in hepatic energy metabolism associated with WD, highlights potential mechanisms involved in the development of hepatic and systemic dysregulation of lipid metabolism, and reviews current knowledge on the effects of copper overload on extrahepatic energy metabolism.
Collapse
Affiliation(s)
- Tagreed A. Mazi
- Department of Nutrition, University of California Davis, Davis, CA, USA,Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Noreene M. Shibata
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, Sacramento, CA, USA
| | - Valentina Medici
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, Sacramento, CA, USA,Corresponding author. (V. Medici)
| |
Collapse
|
44
|
Tavera-Montañez C, Hainer SJ, Cangussu D, Gordon SJV, Xiao Y, Reyes-Gutierrez P, Imbalzano AN, Navea JG, Fazzio TG, Padilla-Benavides T. The classic metal-sensing transcription factor MTF1 promotes myogenesis in response to copper. FASEB J 2019; 33:14556-14574. [PMID: 31690123 PMCID: PMC6894080 DOI: 10.1096/fj.201901606r] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022]
Abstract
Metal-regulatory transcription factor 1 (MTF1) is a conserved metal-binding transcription factor in eukaryotes that binds to conserved DNA sequence motifs, termed metal response elements. MTF1 responds to both metal excess and deprivation, protects cells from oxidative and hypoxic stresses, and is required for embryonic development in vertebrates. To examine the role for MTF1 in cell differentiation, we use multiple experimental strategies [including gene knockdown (KD) mediated by small hairpin RNA and clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9), immunofluorescence, chromatin immunopreciptation sequencing, subcellular fractionation, and atomic absorbance spectroscopy] and report a previously unappreciated role for MTF1 and copper (Cu) in cell differentiation. Upon initiation of myogenesis from primary myoblasts, both MTF1 expression and nuclear localization increased. Mtf1 KD impaired differentiation, whereas addition of nontoxic concentrations of Cu+-enhanced MTF1 expression and promoted myogenesis. Furthermore, we observed that Cu+ binds stoichiometrically to a C terminus tetra-cysteine of MTF1. MTF1 bound to chromatin at the promoter regions of myogenic genes, and Cu addition stimulated this binding. Of note, MTF1 formed a complex with myogenic differentiation (MYOD)1, the master transcriptional regulator of the myogenic lineage, at myogenic promoters. These findings uncover unexpected mechanisms by which Cu and MTF1 regulate gene expression during myoblast differentiation.-Tavera-Montañez, C., Hainer, S. J., Cangussu, D., Gordon, S. J. V., Xiao, Y., Reyes-Gutierrez, P., Imbalzano, A. N., Navea, J. G., Fazzio, T. G., Padilla-Benavides, T. The classic metal-sensing transcription factor MTF1 promotes myogenesis in response to copper.
Collapse
Affiliation(s)
- Cristina Tavera-Montañez
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sarah J. Hainer
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and
| | - Daniella Cangussu
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shellaina J. V. Gordon
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Yao Xiao
- Department of Chemistry, Skidmore College, Saratoga Springs, New York, USA
| | - Pablo Reyes-Gutierrez
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anthony N. Imbalzano
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Juan G. Navea
- Department of Chemistry, Skidmore College, Saratoga Springs, New York, USA
| | - Thomas G. Fazzio
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
45
|
Horvath I, Blockhuys S, Šulskis D, Holgersson S, Kumar R, Burmann BM, Wittung-Stafshede P. Interaction between Copper Chaperone Atox1 and Parkinson's Disease Protein α-Synuclein Includes Metal-Binding Sites and Occurs in Living Cells. ACS Chem Neurosci 2019; 10:4659-4668. [PMID: 31600047 DOI: 10.1021/acschemneuro.9b00476] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Alterations in copper ion homeostasis appear coupled to neurodegenerative disorders, but mechanisms are unknown. The cytoplasmic copper chaperone Atox1 was recently found to inhibit amyloid formation in vitro of α-synuclein, the amyloidogenic protein in Parkinson's disease. As α-synuclein may have copper-dependent functions, and free copper ions promote α-synuclein amyloid formation, it is important to characterize the Atox1 interaction with α-synuclein on a molecular level. Here we applied solution-state nuclear magnetic resonance spectroscopy, with isotopically labeled α-synuclein and Atox1, to define interaction regions in both proteins. The α-synuclein interaction interface includes the whole N-terminal part up to Gln24; in Atox1, residues around the copper-binding cysteines (positions 11-16) are mostly perturbed, but additional effects are also found for residues elsewhere in both proteins. Because α-synuclein is N-terminally acetylated in vivo, we established that Atox1 also inhibits amyloid formation of this variant in vitro, and proximity ligation in human cell lines demonstrated α-synuclein-Atox1 interactions in situ. Thus, this interaction may provide the direct link between copper homeostasis and amyloid formation in vivo.
Collapse
Affiliation(s)
- Istvan Horvath
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Stéphanie Blockhuys
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Darius Šulskis
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg 405 30, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Stellan Holgersson
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Ranjeet Kumar
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Björn M. Burmann
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg 405 30, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Pernilla Wittung-Stafshede
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
46
|
Ghaffari R, Richburg JH. Mice with a Sertoli cell-specific knockout of the Ctr1 gene exhibit a reduced sensitivity to cisplatin-induced testicular germ cell apoptosis. Toxicol Res (Camb) 2019; 8:972-978. [PMID: 32665841 PMCID: PMC7344760 DOI: 10.1039/c9tx00142e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Abstract
Exposure to the chemotherapeutic agent cis-diamminedichloroplatinum(ii) (cDDP) is well known to instigate acute and prolonged testicular injury in male patients.
Exposure to the chemotherapeutic agent cis-diamminedichloroplatinum(ii) (cDDP) is well known to instigate acute and prolonged testicular injury in male patients. Many investigators have hypothesized that cDDP-induced dysfunction of Sertoli cells (SCs) may, in part, account for the cDDP-induced lasting testicular injury. Nevertheless, the relative contribution of cDDP-induced SC injury versus direct effects on germ cells (GCs) to the pathogenesis of GC loss remains to be elucidated. The expression of the copper transporter 1 (CTR1) protein in cells directly corresponds with cDDP uptake and its cellular toxicity. Therefore, to discern the role of SCs in the pathogenic mechanism, mice were developed with a SC-specific disruption of the Ctr1 gene (SCΔCtr1) as a strategy to prevent their exposure to cDDP. Adult mice at postnatal day (PND) 60 were treated with 5 mg kg–1 cDDP and then testis collected at 48 hours. A two-fold increase in GC-apoptosis occurred in the testis of cDDP-treated wildtype (WT) mice as compared to saline-treated WT mice. In contrast, cDDP-treated SCΔCtr1 mice exhibited only a half-fold increase in GC-apoptosis as compared to the saline-treated SCΔCtr1 mice. This reduced incidence of GC apoptosis in the SCΔCtr1 mice corresponded to a significantly lower level of platinum within the testis. Taken together, these findings reveal that the uptake of cDDP by CTR1 in SCs accounts for the accumulation of cDDP in the testis and plays a pivotal role in the pathogenic sequence of events leading to the loss of germ cells via apoptosis.
Collapse
Affiliation(s)
- Rashin Ghaffari
- Institute of Cellular and Molecular Biology , College of Natural Sciences , The University of Texas at Austin , Austin , TX , USA .
| | - John H Richburg
- Institute of Cellular and Molecular Biology , College of Natural Sciences , The University of Texas at Austin , Austin , TX , USA . .,The Center for Molecular Carcinogenesis and Toxicology , Division of Pharmacology & Toxicology , College of Pharmacy , The University of Texas at Austin , Austin , TX , USA
| |
Collapse
|
47
|
De Luca A, Barile A, Arciello M, Rossi L. Copper homeostasis as target of both consolidated and innovative strategies of anti-tumor therapy. J Trace Elem Med Biol 2019; 55:204-213. [PMID: 31345360 DOI: 10.1016/j.jtemb.2019.06.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Copper was reported to be involved in the onset and progression of cancer. Proteins in charge of copper uptake and distribution, as well as cuproenzymes, are altered in cancer. More recently, proteins involved in signaling cascades, regulating cell proliferation, and anti-apoptotic protein factors were found to interact with copper. Therefore, therapeutic strategies using copper complexing molecules have been proposed for cancer therapy and used in clinical trials. OBJECTIVES This review will focus on novel findings about the involvement of copper and cupro-proteins in cancer dissemination process, epithelium to mesenchymal transition and vascularization. Particularly, implication of well-established (e.g. lysil oxidase) or newly identified copper-binding proteins (e.g. MEMO1), as well as their interplay, will be discussed. Moreover, we will describe recently synthesized copper complexes, including plant-derived ones, and their efficacy in contrasting cancer development. CONCLUSIONS The research on the involvement of copper in cancer is still an open field. Further investigation is required to unveil the mechanisms involved in copper delivery to the novel copper-binding proteins, which may identify other possible gene and protein targets for cancer therapy.
Collapse
Affiliation(s)
| | - Anna Barile
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Mario Arciello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
48
|
Abstract
Protein misfolding in cells is avoided by a network of protein chaperones that detect misfolded or partially folded species. When proteins escape these control systems, misfolding may result in protein aggregation and amyloid formation. We here show that aggregation of the amyloidogenic protein α-synuclein (αS), the key player in Parkinson's disease, is controlled by the copper transport protein Atox1 in vitro. Copper ions are not freely available in the cellular environment, but when provided by Atox1, the resulting copper-dependent ternary complex blocks αS aggregation. Because the same inhibition was found for a truncated version of αS, lacking the C-terminal part, it appears that Atox1 interacts with the N-terminal copper site in αS. Metal-dependent chaperoning may be yet another manner in which cells control its proteome.
Collapse
|
49
|
Pan W, Wang Y, Wang N. A new metal affinity NCTR 25 tag as a better alternative to the His-tag for the expression of recombinant fused proteins. Protein Expr Purif 2019; 164:105477. [PMID: 31419547 DOI: 10.1016/j.pep.2019.105477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/20/2019] [Accepted: 08/11/2019] [Indexed: 12/18/2022]
Abstract
His-tagging is commonly used in fusion protein production, but the His-tag is usually prohibited in medicinal proteins and must be removed. A fragment (NCTR25-tag) truncated from the N-terminus of human copper transporter 1 was tested for feasibility as a replacement for the His-tag in fusion proteins. The NCTR25-tag and His-tag were separately fused to the transthyretin (TTR) protein, and the expression, affinity purification, refolding and stability of the two kinds of fusions were compared. NCTR25 fusion produced a 63% higher yield of the recombinant protein, which was purified by metal affinity chromatography with an efficiency similar to that of His-tagged protein. NCTR25-tag fusion had much less impact on the foldability, kinetic and thermodynamic stability of tetrameric TTR than His-tag fusion. When the tags were individually fused to enhanced green fluorescent protein (EGFP), NCTR25 fusion yielded 29-128% more product than His-EGFP. NCTR25-EGFP could be purified by metal affinity chromatography and showed better foldability than His-EGFP. Furthermore, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) fusion with the third disulfide loop of TGF-α (TGF3L-TRAIL) fused with the NCTR25-tag retained the stability and superactivity of His-TGF3L-TRAIL. Therefore, the native tag NCTR25-tag is a feasible alternative to the His-tag in medicinal recombinant proteins.
Collapse
Affiliation(s)
- Weitong Pan
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yan Wang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| | - Nan Wang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
50
|
Skopp A, Boyd SD, Ullrich MS, Liu L, Winkler DD. Copper-zinc superoxide dismutase (Sod1) activation terminates interaction between its copper chaperone (Ccs) and the cytosolic metal-binding domain of the copper importer Ctr1. Biometals 2019; 32:695-705. [PMID: 31292775 PMCID: PMC6647829 DOI: 10.1007/s10534-019-00206-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/03/2019] [Indexed: 11/17/2022]
Abstract
Copper-zinc superoxide dismutase (Sod1) is a critical antioxidant enzyme that rids the cell of reactive oxygen through the redox cycling of a catalytic copper ion provided by its copper chaperone (Ccs). Ccs must first acquire this copper ion, directly or indirectly, from the influx copper transporter, Ctr1. The three proteins of this transport pathway ensure careful trafficking of copper ions from cell entry to target delivery, but the intricacies remain undefined. Biochemical examination of each step in the pathway determined that the activation of the target (Sod1) regulates the Ccs·Ctr1 interaction. Ccs stably interacts with the cytosolic C-terminal tail of Ctr1 (Ctr1c) in a copper-dependent manner. This interaction becomes tripartite upon the addition of an engineered immature form of Sod1 creating a stable Cu(I)-Ctr1c·Ccs·Sod1 heterotrimer in solution. This heterotrimer can also be made by the addition of a preformed Sod1·Ccs heterodimer to Cu(I)-Ctr1c, suggestive of multiple routes to the same destination. Only complete Sod1 activation (i.e. active site copper delivery and intra-subunit disulfide bond formation) breaks the Sod1·Ccs·Ctr1c complex. The results provide a new and extended view of the Sod1 activation pathway(s) originating at cellular copper import.
Collapse
Affiliation(s)
- Amélie Skopp
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Stefanie D Boyd
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Morgan S Ullrich
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Li Liu
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Duane D Winkler
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA.
| |
Collapse
|