1
|
Guy MM, Bian T, Sun L, Hao Y, Jiao X, Zhang W, Zhang T, Cui B. SLC7A11 is a potential therapeutic target and prognostic biomarker correlated with immune cell infiltration in cervical cancer. Discov Oncol 2025; 16:125. [PMID: 39915437 PMCID: PMC11802985 DOI: 10.1007/s12672-025-01888-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND SLC7A11 is importantly in both ferroptosis and disulfidptosis which participated in human development and homeostasis. By utilizing bioinformatics and in vitro validation, we explored SLC7A11's role in cervical cancer. METHODS From the TCGA database, we analyzed SLC7A11 expression profiles and validated its promoting role in cervical cancer by in vitro. Patients were divided into two subgroups according to SLC7A11 expression levels, and differentially expressed genes (DEGs) were identified between these groups. Then SLC7A11's mechanism was then clarified by function enrichment analysis. The association between SLC7A11 and the immune microenvironment was investigated. Finally, we explore potential drugs by oncoPredict. RESULTS Our findings suggest that SLC7A11 could serve as a valuable prognostic biomarker in cervical cancer. Besides, SLC7A11 was positively regulated cervical cancer cells' proliferation, migration and invasion. We identified 113 DEGs between two subgroups. Functional enrichment analysis revealed that these DEGs are linked to immune-related pathways. The SLC7A11 high expression group showed greater enrichment of resting NK cells, neutrophils, M0 macrophages, and activated mast cells, whereas the SLC7A11 low expression group had higher levels of resting dendritic cells, resting mast cells, and follicular helper T cells. Besides, there were higher TMB scores in patients with high SLC7A11 expression. Finally, we explore 37 kinds of drugs may improve prognosis. CONCLUSION SLC7A11, correlated with immune pathway, is a risk factor affecting the prognosis of cervical cancer. We also explore 37 kinds of drugs that may benefit cervical cancer patients.
Collapse
Affiliation(s)
- Mutangala Muloye Guy
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Obstetrics and Gynecology, School of Medicine, University of Lubumbashi, Lubumbashi, Democratic Republic of the Congo
| | - Tingting Bian
- Department of Medical Imaging, Affiliated Hospital of Jining Medical University, Jining City, China
| | - Longyun Sun
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yiping Hao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xinlin Jiao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Wenjing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Teng Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Baoxia Cui
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
2
|
See WR, Yousefi M, Ooi YS. A review of virus host factor discovery using CRISPR screening. mBio 2024; 15:e0320523. [PMID: 39422472 PMCID: PMC11559068 DOI: 10.1128/mbio.03205-23] [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] [Indexed: 10/19/2024] Open
Abstract
The emergence of genome-scale forward genetic screening techniques, such as Haploid Genetic screen and clustered regularly interspaced short palindromic repeats (CRISPR) knockout screen has opened new horizons in our understanding of virus infection biology. CRISPR screening has become a popular tool for the discovery of novel host factors for several viruses due to its specificity and efficiency in genome editing. Here, we review how CRISPR screening has revolutionized our understanding of virus-host interactions from scientific and technological viewpoints. A summary of the published screens conducted thus far to uncover virus host factors is presented, highlighting their experimental design and significant findings. We will outline relevant methods for customizing the CRISPR screening process to answer more specific hypotheses and compile a glossary of conducted CRISPR screens to show their design aspects. Furthermore, using flaviviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as examples, we hope to offer a broad-based perspective on the capabilities of CRISPR screening to serve as a reference point to guide future unbiased discovery of virus host factors.
Collapse
Affiliation(s)
- Wayne Ren See
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Meisam Yousefi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Yaw Shin Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
3
|
Koppin A, Chase L. Lysine 473 Regulates the Progression of SLC7A11, the Cystine/Glutamate Exchanger, through the Secretory Pathway. Int J Mol Sci 2024; 25:10271. [PMID: 39408599 PMCID: PMC11476549 DOI: 10.3390/ijms251910271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
System xc-, the cystine/glutamate exchanger, is a membrane transporter that plays a critical role in the antioxidant response of cells. Recent work has shown that System xc- localizes to the plasma membrane during oxidative stress, allowing for increased activity to support the production of glutathione. In this study, we used site-directed mutagenesis to examine the role of C-terminal lysine residues (K422, K472, and K473) of xCT (SLC7A11) in regulating System xc-. We observed that K473R exhibits loss of transporter activity and membrane localization and is 7.5 kD lower in molecular weight, suggesting that K473 regulates System xc- trafficking and is modified under basal conditions. After ruling out ubiquitination and neddylation, we demonstrated that unlike WT xCT, K473R lacks N- and O-glycosylation and is sequestered in the endoplasmic reticulum. Next, we demonstrated that K473Q, a constitutively acetylated lysine mimic, also exhibits loss of transporter activity, decreased membrane expression, and a 4 kD decrease in molecular weight; however, it is N- and O-glycosylated and localized to the endoplasmic reticulum and Golgi. These results suggest that acetylation and deacetylation of K473 in the endoplasmic reticulum and Golgi, respectively, serve to regulate the progression of the transporter through the biosynthetic pathway.
Collapse
Affiliation(s)
- Anna Koppin
- Departments of Biology and Chemistry, Hope College, Holland, MI 49423, USA;
| | - Leah Chase
- Neuroscience Program, Departments of Biology and Chemistry, Hope College, Holland, MI 49423, USA
| |
Collapse
|
4
|
Zhou Q, Yu H, Chen Y, Ren J, Lu Y, Sun Y. The CRL3 KCTD10 ubiquitin ligase-USP18 axis coordinately regulates cystine uptake and ferroptosis by modulating SLC7A11. Proc Natl Acad Sci U S A 2024; 121:e2320655121. [PMID: 38959043 PMCID: PMC11252818 DOI: 10.1073/pnas.2320655121] [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: 11/24/2023] [Accepted: 05/22/2024] [Indexed: 07/04/2024] Open
Abstract
SLC7A11 is a cystine transporter and ferroptosis inhibitor. How the stability of SLC7A11 is coordinately regulated in response to environmental cystine by which E3 ligase and deubiquitylase (DUB) remains elusive. Here, we report that neddylation inhibitor MLN4924 increases cystine uptake by causing SLC7A11 accumulation, via inactivating Cullin-RING ligase-3 (CRL-3). We identified KCTD10 as the substrate-recognizing subunit of CRL-3 for SLC7A11 ubiquitylation, and USP18 as SLC7A11 deubiquitylase. Upon cystine deprivation, the protein levels of KCTD10 or USP18 are decreased or increased, respectively, contributing to SLC7A11 accumulation. By destabilizing or stabilizing SLC7A11, KCTD10, or USP18 inversely regulates the cystine uptake and ferroptosis. Biologically, MLN4924 combination with SLC7A11 inhibitor Imidazole Ketone Erastin (IKE) enhanced suppression of tumor growth. In human breast tumor tissues, SLC7A11 levels were negatively or positively correlated with KCTD10 or USP18, respectively. Collectively, our study defines how SLC7A11 and ferroptosis is coordinately regulated by the CRL3KCTD10/E3-USP18/DUB axis, and provides a sound rationale of drug combination to enhance anticancer efficacy.
Collapse
Affiliation(s)
- Qiyin Zhou
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310029, China
- Cancer Center, Zhejiang University, Hangzhou310058, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou310009, China
| | - Hongfei Yu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310029, China
- Cancer Center, Zhejiang University, Hangzhou310058, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou310053, China
| | - Yongxia Chen
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310029, China
- Cancer Center, Zhejiang University, Hangzhou310058, China
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310016, China
| | - Jiayi Ren
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310029, China
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310016, China
| | - Yan Lu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310029, China
- Cancer Center, Zhejiang University, Hangzhou310058, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Department of Gynecologic Oncology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou310006, China
| | - Yi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310029, China
- Cancer Center, Zhejiang University, Hangzhou310058, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou310053, China
| |
Collapse
|
5
|
Ashraf AA, Aljuhani M, Hubens CJ, Jeandriens J, Parkes HG, Geraki K, Mahmood A, Herlihy AH, So PW. Inflammation subsequent to mild iron excess differentially alters regional brain iron metabolism, oxidation and neuroinflammation status in mice. Front Aging Neurosci 2024; 16:1393351. [PMID: 38836051 PMCID: PMC11148467 DOI: 10.3389/fnagi.2024.1393351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/26/2024] [Indexed: 06/06/2024] Open
Abstract
Iron dyshomeostasis and neuroinflammation, characteristic features of the aged brain, and exacerbated in neurodegenerative disease, may induce oxidative stress-mediated neurodegeneration. In this study, the effects of potential priming with mild systemic iron injections on subsequent lipopolysaccharide (LPS)-induced inflammation in adult C57Bl/6J mice were examined. After cognitive testing, regional brain tissues were dissected for iron (metal) measurements by total reflection X-ray fluorescence and synchrotron radiation X-Ray fluorescence-based elemental mapping; and iron regulatory, ferroptosis-related, and glia-specific protein analysis, and lipid peroxidation by western blotting. Microglial morphology and astrogliosis were assessed by immunohistochemistry. Iron only treatment enhanced cognitive performance on the novel object location task compared with iron priming and subsequent LPS-induced inflammation. LPS-induced inflammation, with or without iron treatment, attenuated hippocampal heme oxygenase-1 and augmented 4-hydroxynonenal levels. Conversely, in the cortex, elevated ferritin light chain and xCT (light chain of System Xc-) were observed in response to LPS-induced inflammation, without and with iron-priming. Increased microglial branch/process lengths and astrocyte immunoreactivity were also increased by combined iron and LPS in both the hippocampus and cortex. Here, we demonstrate iron priming and subsequent LPS-induced inflammation led to iron dyshomeostasis, compromised antioxidant function, increased lipid peroxidation and altered neuroinflammatory state in a brain region-dependent manner.
Collapse
Affiliation(s)
- Azhaar Ahmad Ashraf
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Manal Aljuhani
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Chantal J Hubens
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jérôme Jeandriens
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- Department of Human Biology and Toxicology, Faculty of Medicine, University of Mons, Mons, Belgium
| | - Harold G Parkes
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Kalotina Geraki
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, United Kingdom
| | - Ayesha Mahmood
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | | | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
6
|
Zheng S, Li Y, Yin L, Lu M. Identification of sulfur metabolism-related gene signature in osteoarthritis and TM9SF2's sustenance effect on M2 macrophages' phagocytic activity. J Orthop Surg Res 2024; 19:62. [PMID: 38218914 PMCID: PMC10787471 DOI: 10.1186/s13018-023-04384-2] [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: 10/12/2023] [Accepted: 11/18/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic and low-grade inflammatory disease associated with metabolism disorder and multiple cell death types in the synovial tissues. Sulfur metabolism has not been studied in OA. METHODS First, we calculated the single sample gene set enrichment analysis score of sulfur metabolism-associated annotations (i.e., cysteine metabolism process, regulation of sulfur metabolism process, and disulfidptosis) between healthy and synovial samples from patients with OA. Sulfur metabolism-related differentially expressed genes (DEGs) were analyzed in OA. Least absolute shrinkage and selection operator COX regression were used to identify the sulfur metabolism-associated gene signature for diagnosing OA. Correlation and immune cell deconvolution analyses were used to explore the correlated functions and cell specificity of the signature gene, TM9SF2. TM9SF2's effect on the phagocytosis of macrophages M2 was analyzed by coculturing macrophages with IgG-coated beads or apoptotic Jurkat cells. RESULTS A diagnostic six gene signature (i.e., MTHFD1, PDK4, TM9SF2, POU4F1, HOXA2, NCKAP1) was identified based on the ten DEGs, validated using GSE12021 and GSE1919 datasets. TM9SF2 was upregulated in the synovial tissues of OA at both mRNA and protein levels. The relationship between TM9SF2 and several functional annotations, such as antigen processing and presentation, lysosome, phagosome, Fcγ-mediated phagocytosis, and tyrosine metabolism, was identified. TM9SF2 and macrophages M2 were significantly correlated. After silencing TM9SF2 in THP-1-derived macrophages M2, a significantly reduced phagocytosis and attenuated activation of PLC-γ1 were observed. CONCLUSION A sulfur metabolism-associated six-gene signature for OA diagnosis was constructed and upregulation of the phagocytosis-associated gene, TM9SF2, was identified. The findings are expected to deepen our understanding of the molecular mechanism underlying OA development and be used as potential therapeutic targets.
Collapse
Affiliation(s)
- Shuang Zheng
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China.
| | - Yetian Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China
| | - Li Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China
| | - Ming Lu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
7
|
Li Z, Hu B, Du L, Hou C, Li Q. Involvement of B-aat1 and Cbs in regulating mantle pigmentation in the Pacific oyster (Crassostrea gigas). Mol Biol Rep 2023; 50:377-387. [PMID: 36335521 DOI: 10.1007/s11033-022-08037-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Shell color formation is an important physiological process in bivalves, the molecular genetic basis has potential application in bivalve aquaculture, but there is still remaining unclear about this issue. The cystine/glutamate transporter (Slc7a11) and cystathionine beta-synthase (Cbs) are integral genes in pheomelanin synthesis pathway, which is vital to skin pigmentation. METHODS AND RESULTS Here, the sequences of b (0, +) -type amino acid transporter 1 (B-aat1) and Cbs in Pacific oyster (Crassostrea gigas) (CgB-aat1, CgCbs) were characterized. Phylogenetically, the deduced amino acid sequences of CgB-aat1 and CgCbs both possessed conserved features. Genes were both ubiquitously expressed in six tested tissues with more abundant expression level in central mantle. Besides, the polyclonal antibodies of CgB-aat1, CgCbs, CgTyr, and CgTyrp2 were successfully prepared. Immunofluorescence analysis revealed that CgB-aat1 and CgCbs proteins were both expressed in gill rudiments of eyed-larvae and concentrated mainly in cytoplasm of epithelial cell and nerve axons in mantle. Additionally, after CgB-aat1 or CgCbs silencing, expressions at mRNA and protein levels of CgB-aat1 and CgCbs involved in pheomelanin synthesis were significantly suppressed, and CgTyr, CgTyrp1 and CgTyrp2 related to eumelanin synthesis were also down-regulated but no apparent differences, respectively. Moreover, micrographic examination found less brown-granules at mantle edge in CgB-aat1 interference group. CONCLUSION These results implied that pheomelanin synthesis was possible induced by CgB-aat1-CgTyr-CgCbs axis, and it played an essential role on mantle pigmentation in the oysters. These findings provide the useful genetic knowledge and enrich the physiological information for the shell color formation in bivalve aquaculture.
Collapse
Affiliation(s)
- Zhuanzhuan Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Biyang Hu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Lijie Du
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Chunhao Hou
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China. .,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
8
|
Hang TD, Hung HM, Beckers P, Desmet N, Lamrani M, Massie A, Hermans E, Vanommeslaeghe K. Structural investigation of human cystine/glutamate antiporter system xc− (Sxc−) using homology modeling and molecular dynamics. Front Mol Biosci 2022; 9:1064199. [DOI: 10.3389/fmolb.2022.1064199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
The cystine/glutamate antiporter system xc− (Sxc−) belongs to the SLC7 family of plasma membrane transporters. It exports intracellular glutamate along the latter’s concentration gradient as a driving force for cellular uptake of cystine. Once imported, cystine is mainly used for the production of glutathione, a tripeptide thiol crucial in maintenance of redox homeostasis and protection of cells against oxidative stress. Overexpression of Sxc− has been found in several cancer cells, where it is thought to counteract the increased oxidative stress. In addition, Sxc− is important in the central nervous system, playing a complex role in regulating glutamatergic neurotransmission and glutamate toxicity. Accordingly, this transporter is considered a potential target for the treatment of cancer as well as neurodegenerative diseases. Till now, no specific inhibitors are available. We herein present four conformations of Sxc− along its transport pathway, obtained using multi-template homology modeling and refined by means of Molecular Dynamics. Comparison with a very recently released cryo-EM structure revealed an excellent agreement with our inward-open conformation. Intriguingly, our models contain a structured N-terminal domain that is unresolved in the experimental structures and is thought to play a gating role in the transport mechanism of other SLC7 family members. In contrast to the inward-open model, there is no direct experimental counterpart for the other three conformations we obtained, although they are in fair agreement with the other stages of the transport mechanism seen in other SLC7 transporters. Therefore, our models open the prospect for targeting alternative Sxc− conformations in structure-based drug design efforts.
Collapse
|
9
|
The Role of SLC7A11 in Cancer: Friend or Foe? Cancers (Basel) 2022; 14:cancers14133059. [PMID: 35804831 PMCID: PMC9264807 DOI: 10.3390/cancers14133059] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/12/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
SLC7A11 controls the uptake of extracellular cystine in exchange for glutamate at a ratio of 1:1, and it is overexpressed in a variety of tumours. Accumulating evidence has shown that the expression of SLC7A11 is fine-tuned at multiple levels, and plays diverse functional and pharmacological roles in tumours, such as cellular redox homeostasis, cell growth and death, and cell metabolism. Many reports have suggested that the inhibition of SLC7A11 expression and activity is favourable for tumour therapy; thus, SLC7A11 is regarded as a potential therapeutic target. However, emerging evidence also suggests that on some occasions, the inhibition of SLC7A11 is beneficial to the survival of cancer cells, and confers the development of drug resistance. In this review, we first briefly introduce the biological properties of SLC7A11, including its structure and physiological functions, and further summarise its regulatory network and potential regulators. Then, focusing on its role in cancer, we describe the relationships of SLC7A11 with tumourigenesis, survival, proliferation, metastasis, and therapeutic resistance in more detail. Finally, since SLC7A11 has been linked to cancer through multiple approaches, we propose that its contribution and regulatory mechanism require further elucidation. Thus, more personalised therapeutic strategies should be adapted when targeting SLC7A11.
Collapse
|
10
|
Pardieu B, Pasanisi J, Ling F, Dal Bello R, Penneroux J, Su A, Joudinaud R, Chat L, Wu HC, Duchmann M, Sodaro G, Chauvel C, Castelli FA, Vasseur L, Pacchiardi K, Belloucif Y, Laiguillon MC, Meduri E, Vaganay C, Alexe G, Berrou J, Benaksas C, Forget A, Braun T, Gardin C, Raffoux E, Clappier E, Adès L, de Thé H, Fenaille F, Huntly BJ, Stegmaier K, Dombret H, Fenouille N, Lobry C, Puissant A, Itzykson R. Cystine uptake inhibition potentiates front-line therapies in acute myeloid leukemia. Leukemia 2022; 36:1585-1595. [PMID: 35474100 DOI: 10.1038/s41375-022-01573-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 12/17/2022]
Abstract
By querying metabolic pathways associated with leukemic stemness and survival in multiple AML datasets, we nominated SLC7A11 encoding the xCT cystine importer as a putative AML dependency. Genetic and chemical inhibition of SLC7A11 impaired the viability and clonogenic capacity of AML cell lines in a cysteine-dependent manner. Sulfasalazine, a broadly available drug with xCT inhibitory activity, had anti-leukemic activity against primary AML samples in ex vivo cultures. Multiple metabolic pathways were impacted upon xCT inhibition, resulting in depletion of glutathione pools in leukemic cells and oxidative stress-dependent cell death, only in part through ferroptosis. Higher expression of cysteine metabolism genes and greater cystine dependency was noted in NPM1-mutated AMLs. Among eight anti-leukemic drugs, the anthracycline daunorubicin was identified as the top synergistic agent in combination with sulfasalazine in vitro. Addition of sulfasalazine at a clinically relevant concentration significantly augmented the anti-leukemic activity of a daunorubicin-cytarabine combination in a panel of 45 primary samples enriched in NPM1-mutated AML. These results were confirmed in vivo in a patient-derived xenograft model. Collectively, our results nominate cystine import as a druggable target in AML and raise the possibility to repurpose sulfasalazine for the treatment of AML, notably in combination with chemotherapy.
Collapse
Affiliation(s)
- Bryann Pardieu
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Justine Pasanisi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Frank Ling
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Reinaldo Dal Bello
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Justine Penneroux
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Angela Su
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Romane Joudinaud
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Laureen Chat
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Hsin Chieh Wu
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR1050, CNRS UMR, 7241, Paris, France
| | - Matthieu Duchmann
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Gaetano Sodaro
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Clémentine Chauvel
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Florence A Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif-sur-Yvette, France
| | - Loic Vasseur
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Kim Pacchiardi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Yannis Belloucif
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Marie-Charlotte Laiguillon
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Eshwar Meduri
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - Camille Vaganay
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeannig Berrou
- Université Paris Cité, Leukemia Transfer Lab, EA 3518, Institut de Recherche Saint-Louis, F-75010, Paris, France
| | - Chaima Benaksas
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Antoine Forget
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Thorsten Braun
- Université Paris Cité, Leukemia Transfer Lab, EA 3518, Institut de Recherche Saint-Louis, F-75010, Paris, France
| | - Claude Gardin
- Université Paris Cité, Leukemia Transfer Lab, EA 3518, Institut de Recherche Saint-Louis, F-75010, Paris, France
| | - Emmanuel Raffoux
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Emmanuelle Clappier
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Lionel Adès
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Hugues de Thé
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR1050, CNRS UMR, 7241, Paris, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif-sur-Yvette, France
| | - Brian J Huntly
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hervé Dombret
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
- The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nina Fenouille
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Camille Lobry
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Alexandre Puissant
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Raphael Itzykson
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France.
| |
Collapse
|
11
|
Xu R, Huang Y, Zhu D, Guo J. Iron promotes Slc7a11-deficient valvular interstitial cell osteogenic differentiation: A possible mechanism by which ferroptosis participates in intraleaflet hemorrhage-induced calcification. Free Radic Biol Med 2022; 184:158-169. [PMID: 35331838 DOI: 10.1016/j.freeradbiomed.2022.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 01/18/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most frequent pathogeny of aortic valve replacement in developed countries. Iron deposits are found in the intraleaflet hemorrhage (IH) areas of calcific aortic valves. Ferroptosis is a form of regulated cell death that involves metabolic dysfunction resulting from iron overload-dependent excessive lipid peroxidation. In this study, histological analysis showed that ferroptosis occurs in the IH areas of calcific aortic valves. We also demonstrated that Slc7a11 is expressed at low levels in OM-treated valvular interstitial cells (VICs) and IH areas and that low Slc7a11 expression is associated with calcification in CAVD. However, iron overload treatment did not promote VIC calcification under osteogenic conditions in vitro. Using lentiviral transfection to knockdown Slc7a11 in VICs, we found that the degree of iron overload-induced ferroptosis was positively increased in vitro. Finally, we also found that Slc7a11 knockdown promoted the osteogenic differentiation of VICs in vitro. In summary, this study reports a novel mechanism linking ferroptosis and CAVD development in which iron may promote Slc7a11-deficient VIC osteogenic differentiation by aggravating ferroptosis in vitro, thereby accelerating the progression of aortic valve calcification.
Collapse
Affiliation(s)
- Ran Xu
- Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Huaihai Road, Shanghai, 200000, China.
| | - Ying Huang
- Department of Cardiology, Shanghai East Hospital, Tongji University, Jimo Road, Shanghai, 200000, China.
| | - Dan Zhu
- Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Huaihai Road, Shanghai, 200000, China.
| | - Jianghong Guo
- The Rugao People's Hospital, Teaching Hospital of Nantong University, Rugao, Ninghai Road, Nantong, 226500, China.
| |
Collapse
|
12
|
Parker JL, Deme JC, Kolokouris D, Kuteyi G, Biggin PC, Lea SM, Newstead S. Molecular basis for redox control by the human cystine/glutamate antiporter system xc . Nat Commun 2021; 12:7147. [PMID: 34880232 PMCID: PMC8654953 DOI: 10.1038/s41467-021-27414-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Cysteine plays an essential role in cellular redox homoeostasis as a key constituent of the tripeptide glutathione (GSH). A rate limiting step in cellular GSH synthesis is the availability of cysteine. However, circulating cysteine exists in the blood as the oxidised di-peptide cystine, requiring specialised transport systems for its import into the cell. System xc- is a dedicated cystine transporter, importing cystine in exchange for intracellular glutamate. To counteract elevated levels of reactive oxygen species in cancerous cells system xc- is frequently upregulated, making it an attractive target for anticancer therapies. However, the molecular basis for ligand recognition remains elusive, hampering efforts to specifically target this transport system. Here we present the cryo-EM structure of system xc- in both the apo and glutamate bound states. Structural comparisons reveal an allosteric mechanism for ligand discrimination, supported by molecular dynamics and cell-based assays, establishing a mechanism for cystine transport in human cells.
Collapse
Affiliation(s)
- Joanne L Parker
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
| | - Justin C Deme
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- Central Oxford Structural Molecular Imaging Centre, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | | | - Gabriel Kuteyi
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Susan M Lea
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
- Central Oxford Structural Molecular Imaging Centre, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA.
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK.
| |
Collapse
|
13
|
Rolih V, Caldeira J, Bolli E, Salameh A, Conti L, Barutello G, Riccardo F, Magri J, Lamolinara A, Parra K, Valenzuela P, Francia G, Iezzi M, Pericle F, Cavallo F. Development of a VLP-Based Vaccine Displaying an xCT Extracellular Domain for the Treatment of Metastatic Breast Cancer. Cancers (Basel) 2020; 12:cancers12061492. [PMID: 32521631 PMCID: PMC7352461 DOI: 10.3390/cancers12061492] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 01/17/2023] Open
Abstract
Metastatic breast cancer (MBC) is the leading cause of cancer death in women due to recurrence and resistance to conventional therapies. Thus, MBC represents an important unmet clinical need for new treatments. In this paper we generated a virus-like particle (VLP)-based vaccine (AX09) to inhibit de novo metastasis formation and ultimately prolong the survival of patients with MBC. To this aim, we engineered the bacteriophage MS2 VLP to display an extracellular loop of xCT, a promising therapeutic target involved in tumor progression and metastasis formation. Elevated levels of this protein are observed in a high percentage of invasive mammary ductal tumors including triple negative breast cancer (TNBC) and correlate with poor overall survival. Moreover, xCT expression is restricted to only a few normal cell types. Here, we tested AX09 in several MBC mouse models and showed that it was well-tolerated and elicited a strong antibody response against xCT. This antibody-based response resulted in the inhibition of xCT's function in vitro and reduced metastasis formation in vivo. Thus, AX09 represents a promising novel approach for MBC, and it is currently advancing to clinical development.
Collapse
Affiliation(s)
- Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; (V.R.); (E.B.); (L.C.); (G.B.); (F.R.); (J.M.)
| | - Jerri Caldeira
- AgilVax Inc., Albuquerque, NM 87110, USA; (J.C.); (A.S.); (F.P.)
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; (V.R.); (E.B.); (L.C.); (G.B.); (F.R.); (J.M.)
| | - Ahmad Salameh
- AgilVax Inc., Albuquerque, NM 87110, USA; (J.C.); (A.S.); (F.P.)
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; (V.R.); (E.B.); (L.C.); (G.B.); (F.R.); (J.M.)
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; (V.R.); (E.B.); (L.C.); (G.B.); (F.R.); (J.M.)
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; (V.R.); (E.B.); (L.C.); (G.B.); (F.R.); (J.M.)
| | - Jolanda Magri
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; (V.R.); (E.B.); (L.C.); (G.B.); (F.R.); (J.M.)
| | - Alessia Lamolinara
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technologies (CAST), G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy; (A.L.); (M.I.)
| | - Karla Parra
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (K.P.); (P.V.); (G.F.)
| | - Paloma Valenzuela
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (K.P.); (P.V.); (G.F.)
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA; (K.P.); (P.V.); (G.F.)
| | - Manuela Iezzi
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technologies (CAST), G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy; (A.L.); (M.I.)
| | - Federica Pericle
- AgilVax Inc., Albuquerque, NM 87110, USA; (J.C.); (A.S.); (F.P.)
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; (V.R.); (E.B.); (L.C.); (G.B.); (F.R.); (J.M.)
- Correspondence: ; Tel.: +39-011-670-6457; Fax: +39-011-236-6457
| |
Collapse
|
14
|
Choi J, Li W, Schindell B, Ni L, Liu S, Zhao X, Gong J, Nyachoti M, Yang C. Molecular cloning, tissue distribution and the expression of cystine/glutamate exchanger (xCT, SLC7A11) in different tissues during development in broiler chickens. ACTA ACUST UNITED AC 2020; 6:107-114. [PMID: 32211536 PMCID: PMC7082690 DOI: 10.1016/j.aninu.2019.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 01/29/2023]
Abstract
The cystine/glutamate exchanger (xCT, SLC7A11) is a component of the system Xc amino-acid antiporter that is able to export glutamate and import cysteine into cells. The xCT amino acid exchanger has received a lot of attention, due to the fact that cysteine is an essential substrate for the synthesis of glutathione (GSH), an endogenous antioxidant in cells. The objective of this research was to clone the full-length cDNA of chicken xCT, and to investigate the gene expression of xCT in different tissues, including intestinal segments of broiler chickens during development. The full-length cDNA of chicken xCT (2,703 bp) was obtained from the jejunum by reverse transcription-PCR and sequenced. Homology tests showed that chicken xCT had 80.4%, 80.2%, and 71.2% homology at the nucleotide level with humans, cattle, and rats, respectively. Likewise, amino acid sequence analysis showed that chicken xCT protein is 86.4%, 79.3%, and 75.6% homologous with humans, cattle, and rats, respectively. Additionally, phylogenetic analysis indicated that chicken xCT genes share a closer genetic relationship with humans and cattle, than with rats. The chicken xCT protein has 12 transmembrane helixes, 6 extracellular loops, and 5 intracellular loops. The mRNA of xCT was detected in all tissues, including intestinal segments, in which the mRNA expression of xCT was significantly higher (P < 0.05) within the colon, compared to the jejunum and ileum. During development, a linear pattern of changes regarding the levels of the xCT mRNA was found, indicating that there was an abundance of xCT within the duodenum (P < 0.05). Furthermore, there were changes of the xCT mRNA abundance in the colon during development, which displayed linear and cubic patterns (P < 0.05). These results indicated that xCT is widely expressed both in intestinal segments, as well as other organs that are not associated with nutrient absorption. Further investigation is needed to characterize the functional relevance of xCT activity in oxidative stress and inflammation in the small intestine of broiler chickens.
Collapse
Affiliation(s)
- Janghan Choi
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Weiqi Li
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Brayden Schindell
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Liju Ni
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.,Shanghai Lab-Animal Research Center, Shanghai, 201203, China
| | - Shangxi Liu
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Xiaoya Zhao
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Joshua Gong
- Guelph Research and Development Centre, Agriculture Agri-Food Canada, Guelph, ON, N1G 5C9, Canada
| | - Martin Nyachoti
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Chengbo Yang
- Department of Animal Science, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| |
Collapse
|
15
|
Laurent Q, Berthet M, Cheng Y, Sakai N, Barluenga S, Winssinger N, Matile S. Probing for Thiol-Mediated Uptake into Bacteria. Chembiochem 2020; 21:69-73. [PMID: 31603284 DOI: 10.1002/cbic.201900378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Indexed: 01/02/2023]
Abstract
Cellular uptake mediated by cyclic oligochalcogenides (COCs) is emerging as a conceptually innovative method to penetrate mammalian cells. Their mode of action is based on dynamic covalent oligochalcogenide exchange with cellular thiols. To test thiol-mediated uptake in bacteria, five antibiotics have been equipped with up to three different COCs: One diselenolane and two dithiolanes. We found that the COCs do not activate antibiotics in Gram-negative bacteria. In Gram-positive bacteria, the COCs inactivate antibiotics that act in the cytoplasm and reduce the activity of antibiotics that act on the cell surface. These results indicate that thiol-mediated uptake operates in neither of the membranes of bacteria. COCs are likely to exchange with thiols on the inner, maybe also on the outer membrane, but do not move on. Concerning mammalian cells, the absence of a COC-mediated uptake into bacteria observed in this study disfavors trivial mechanisms, such as passive diffusion, and supports the existence of more sophisticated, so far poorly understood uptake pathways.
Collapse
Affiliation(s)
- Quentin Laurent
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Mathéo Berthet
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Yangyang Cheng
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Naomi Sakai
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Sofia Barluenga
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Nicolas Winssinger
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Stefan Matile
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| |
Collapse
|
16
|
Solute carrier transporters: the metabolic gatekeepers of immune cells. Acta Pharm Sin B 2020; 10:61-78. [PMID: 31993307 PMCID: PMC6977534 DOI: 10.1016/j.apsb.2019.12.006] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/29/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
Solute carrier (SLC) transporters meditate many essential physiological functions, including nutrient uptake, ion influx/efflux, and waste disposal. In its protective role against tumors and infections, the mammalian immune system coordinates complex signals to support the proliferation, differentiation, and effector function of individual cell subsets. Recent research in this area has yielded surprising findings on the roles of solute carrier transporters, which were discovered to regulate lymphocyte signaling and control their differentiation, function, and fate by modulating diverse metabolic pathways and balanced levels of different metabolites. In this review, we present current information mainly on glucose transporters, amino-acid transporters, and metal ion transporters, which are critically important for mediating immune cell homeostasis in many different pathological conditions.
Collapse
Key Words
- 3-PG, 3-phosphoglyceric acid
- ABC, ATP-binding cassette
- AIF, apoptosis-inducing factor
- AP-1, activator protein 1
- ASCT2, alanine serine and cysteine transporter system 2
- ATP, adenosine triphosphate
- BCR, B cell receptor
- BMDMs, bone marrow-derived macrophages
- CD45R, a receptor-type protein tyrosine phosphatase
- CTL, cytotoxic T lymphocytes
- DC, dendritic cells
- EAATs, excitatory amino acid transporters
- ER, endoplasmic reticulum
- ERRα, estrogen related receptor alpha
- FFA, free fatty acids
- G-6-P, glucose 6-phosphate
- GLUT, glucose transporters
- GSH, glutathione
- Glucose
- Glutamine
- HIF-1α, hypoxia-inducible factor 1-alpha
- HIV-1, human immunodeficiency virus type 1
- Hk1, hexokinase-1
- IFNβ, interferon beta
- IFNγ, interferon gamma
- IKK, IκB kinase
- IKKβ, IκB kinase beta subunit
- IL, interleukin
- LDHA, lactate dehydrogenase A
- LPS, lipopolysaccharide
- Lymphocytes
- Lyn, tyrosine-protein kinase
- MAPK, mitogen-activated protein kinase
- MCT, monocarboxylate transporters
- MS, multiple sclerosis
- Metal ion
- NADPH, nicotinamide adenine dinucleotide phosphate
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NO, nitric oxide
- NOD2, nucleotide-binding oligomerization domain containing 2
- PEG2, prostaglandin E2
- PI-3K/AKT, phosphatidylinositol-3-OH kinase/serine–threonine kinase
- PPP, pentose phosphate pathway
- Pfk, phosphofructokinase
- RA, rheumatoid arthritis
- RLR, RIG-I-like receptor
- ROS, reactive oxygen species
- SLC, solute carrier
- SLE, systemic lupus erythematosus
- SNAT, sodium-coupled neutral amino acid transporters
- STAT, signal transducers and activators of transcription
- Solute carrier
- TAMs, tumor-associated macrophages
- TCA, tricarboxylic acid
- TCR, T cell receptor
- TLR, toll-like receptor
- TNF, tumor necrosis factor
- TRPM7, transient receptor potential cation channel subfamily M member 7
- Teffs, effector T cells
- Th1/2/17, type 1/2/17 helper T cells
- Tregs, regulatory T cells
- VEGF, vascular endothelial growth factor
- ZIP, zrt/irt-like proteins
- iNOS, inducible nitric oxide synthase
- iTregs, induced regulatory T cells
- mTORC1, mammalian target of rapamycin complex 1
- α-KG, α-ketoglutaric acid
Collapse
|
17
|
Sharma M, Anirudh CR. In silico characterization of residues essential for substrate binding of human cystine transporter, xCT. J Mol Model 2019; 25:336. [PMID: 31705320 DOI: 10.1007/s00894-019-4233-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
xCT is a sodium-independent amino acid antiporter that imports L-cystine and exports L-glutamate in a 1:1 ratio. It is a component of heterodimeric amino acid transporter system Xc- working at the cross-roads of maintaining neurological processes and regulating antioxidant defense. The transporter has 12 transmembrane domains with intracellular N- and C-termini, and like other transporter proteins can undergo various conformational changes while switching the ligand accessibilities from intracellular to extracellular site. In the present study, we generated two homology models of human xCT in two distinct conformations: inward-facing occluded state and outward-facing open state. Our results indicated the substrate translocation channel composed of transmembrane helices TMs 1, 3, 6, 8, and 10. We docked anionic L-cystine and L-glutamate within the cavities to assess the two distinct binding scenarios for xCT as antiporter. We also assessed the interactions between the ligands and transporter and observed that ligands bind to similar residues within the channel. Using MM-PBSA/MM-GBSA approach, we computed the binding energies of these ligands to different conformational states. Cystine and glutamate bind xCT with favorable binding energies, with more favorable binding observed in inward occluded state than in outward open state. We further computed the residue-wise decomposition of these binding energies and identified the residues as essential for substrate binding/permeation. Filtering the residues that form favorable energetic contributions to the ligand binding in both the states, our studies suggest T56, A60, R135, A138, V141, Y244, A247, F250, S330, L392, and R396 as critical residues for ligand binding as well as ligand transport for any conformational state adopted by xCT during its transport cycle. .Graphical Abstract.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Sector 81, Knowledge City, SAS, Nagar, Punjab, India.
| | - C R Anirudh
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Sector 81, Knowledge City, SAS, Nagar, Punjab, India
| |
Collapse
|
18
|
Wang LM, Bu HY, Song FB, Zhu WB, Fu JJ, Dong ZJ. Characterization and functional analysis of slc7a11 gene, involved in skin color differentiation in the red tilapia. Comp Biochem Physiol A Mol Integr Physiol 2019; 236:110529. [DOI: 10.1016/j.cbpa.2019.110529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 10/26/2022]
|
19
|
Effect of oxidative stress on cystine transportation by xC‾ antiporter. Arch Biochem Biophys 2019; 674:108114. [PMID: 31557465 DOI: 10.1016/j.abb.2019.108114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/19/2019] [Accepted: 09/22/2019] [Indexed: 12/15/2022]
Abstract
We performed computer simulations to investigate the effect of oxidation on the extracellular cystine (CYC) uptake by the xC- antiporter. The latter is important for killing of cancer cells. Specifically, applying molecular dynamics (MD) simulations we studied the transport of CYC across xCT, i.e., the light subunit of the xC- antiporter, in charge of bidirectional transport of CYC and glutamate. We considered the outward facing (OF) configuration of xCT, and to study the effect of oxidation, we modified the Cys327 residue, located in the vicinity of the extracellular milieu, to cysteic acid (CYO327). Our computational results showed that oxidation of Cys327 results in a free energy barrier for CYC translocation, thereby blocking the access of CYC to the substrate binding site of the OF system. The formation of the energy barrier was found to be due to the conformational changes in the channel. Analysis of the MD trajectories revealed that the reorganization of the side chains of the Tyr244 and CYO327 residues play a critical role in the OF channel blocking. Indeed, the calculated distance between Tyr244 and either Cys327 or CYO327 showed a narrowing of the channel after oxidation. The obtained free energy barrier for CYC translocation was found to be 33.9kJmol-1, indicating that oxidation of Cys327, by e.g., cold atmospheric plasma, is more effective in inhibiting the xC- antiporter than in the mutation of this amino acid to Ala (yielding a barrier of 32.4kJmol-1). The inhibition of the xC- antiporter may lead to Cys starvation in some cancer cells, eventually resulting in cancer cell death.
Collapse
|
20
|
Slc7a11 Modulated by POU2F1 is Involved in Pigmentation in Rabbit. Int J Mol Sci 2019; 20:ijms20102493. [PMID: 31137576 PMCID: PMC6566412 DOI: 10.3390/ijms20102493] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/17/2019] [Accepted: 05/18/2019] [Indexed: 01/22/2023] Open
Abstract
Solute carrier family 7 member 11 (Slc7a11) is a cystine/glutamate xCT transporter that controls the production of pheomelanin pigment to change fur and skin color in animals. Previous studies have found that skin expression levels of Slc7a11 varied significantly with fur color in Rex rabbits. However, the molecular regulation mechanism of Slc7a11 in pigmentation is unknown. Here, rabbit melanocytes were first isolated and identified. The distribution and expression pattern of Slc7a11 was confirmed in skin from rabbits with different fur colors. Slc7a11 affected the expression of pigmentation related genes and thus affected melanogenesis. Meanwhile, Slc7a11 decreased melanocyte apoptosis, but inhibition of Slc7a11 enhanced apoptosis. Furthermore, the POU2F1 protein was found to bind to the −713 to −703 bp region of Slc7a11 promoter to inhibit its activity in a dual-luciferase reporter and site-directed mutagenesis assay. This study reveals the function of the Slc7a11 in melanogenesis and provides in-depth analysis of the mechanism of fur pigmentation.
Collapse
|
21
|
L amino acid transporter structure and molecular bases for the asymmetry of substrate interaction. Nat Commun 2019; 10:1807. [PMID: 31000719 PMCID: PMC6472337 DOI: 10.1038/s41467-019-09837-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 04/02/2019] [Indexed: 11/26/2022] Open
Abstract
L-amino acid transporters (LATs) play key roles in human physiology and are implicated in several human pathologies. LATs are asymmetric amino acid exchangers where the low apparent affinity cytoplasmic side controls the exchange of substrates with high apparent affinity on the extracellular side. Here, we report the crystal structures of an LAT, the bacterial alanine-serine-cysteine exchanger (BasC), in a non-occluded inward-facing conformation in both apo and substrate-bound states. We crystallized BasC in complex with a nanobody, which blocks the transporter from the intracellular side, thus unveiling the sidedness of the substrate interaction of BasC. Two conserved residues in human LATs, Tyr 236 and Lys 154, are located in equivalent positions to the Na1 and Na2 sites of sodium-dependent APC superfamily transporters. Functional studies and molecular dynamics (MD) calculations reveal that these residues are key for the asymmetric substrate interaction of BasC and in the homologous human transporter Asc-1. L-Amino acid Transporters (LATs) are asymmetric amino acid exchangers. Here the authors determine the crystal structure of a prokaryotic LAT, the alanine-serine-cysteine exchanger (BasC) and identify key residues for asymmetric substrate interaction in both BasC and the homologous human transporter Asc-1 through functional studies.
Collapse
|
22
|
Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, Song S, Tavana O, Gu W. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol 2019; 21:579-591. [PMID: 30962574 PMCID: PMC6624840 DOI: 10.1038/s41556-019-0305-6] [Citation(s) in RCA: 618] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/27/2019] [Indexed: 12/23/2022]
Abstract
It is well established that ferroptosis is primarily controlled by glutathione peroxidase 4 (GPX4). Surprisingly, we observed that p53 activation modulates ferroptotic responses without apparent effects on GPX4 function. Instead, ALOX12 inactivation diminishes p53-mediated ferroptosis induced by ROS stress and abrogates p53-dependent inhibition of tumor growth in xenograft models, suggesting that ALOX12 is critical for p53-mediated ferroptosis. The ALOX12 gene resides on human chromosome 17p13.1, a hot spot of monoallelic deletion in human cancers. Loss of one ALOX12 allele is sufficient to accelerate tumorigenesis in Eμ-Myc lymphoma models. Moreover, ALOX12 missense mutations from human cancers abrogate its ability to oxygenate polyunsaturated fatty acids and to induce p53-mediated ferroptosis. Notably, ALOX12 is dispensable for ferroptosis induced by erastin or GPX4 inhibitors; conversely, ACSL4 is required for ferroptosis upon GPX4 inhibition but dispensable for p53-mediated ferroptosis. Thus, our study identifies an ALOX12-mediated, ACSL4-independent ferroptosis pathway that is critical for p53-dependent tumor suppression.
Collapse
Affiliation(s)
- Bo Chu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Ning Kon
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Delin Chen
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Tongyuan Li
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Tong Liu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Le Jiang
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Shujuan Song
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Omid Tavana
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
23
|
Transport of cystine across xC− antiporter. Arch Biochem Biophys 2019; 664:117-126. [DOI: 10.1016/j.abb.2019.01.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 01/17/2023]
|
24
|
Bolli E, O'Rourke JP, Conti L, Lanzardo S, Rolih V, Christen JM, Barutello G, Forni M, Pericle F, Cavallo F. A Virus-Like-Particle immunotherapy targeting Epitope-Specific anti-xCT expressed on cancer stem cell inhibits the progression of metastatic cancer in vivo. Oncoimmunology 2017; 7:e1408746. [PMID: 29399412 PMCID: PMC5790338 DOI: 10.1080/2162402x.2017.1408746] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/17/2017] [Accepted: 11/18/2017] [Indexed: 01/17/2023] Open
Abstract
Aggressive forms of breast cancer, such as Her2+ and triple negative breast cancer (TNBC), are enriched in breast cancer stem cells (BCSC) and have limited therapeutic options. BCSC represent a key cellular reservoir for relapse, metastatic progression and therapeutic resistance. Their ability to resist common cytotoxic therapies relies on different mechanisms, including improved detoxification. The cystine-glutamate antiporter protein xCT (SLC7A11) regulates cystine intake, conversion to cysteine and subsequent glutathione synthesis, protecting cells against oxidative and chemical insults. Our previous work showed that xCT is highly expressed in tumorspheres derived from breast cancer cell lines and downregulation of xCT altered BCSC function in vitro and inhibited pulmonary metastases in vivo. We further strengthened these observations by developing a virus-like-particle (VLP; AX09-0M6) immunotherapy targeting the xCT protein. AX09-0M6 elicited a strong antibody response against xCT including high levels of IgG2a antibody. IgG isolated from AX09-0M6 treated mice bound to tumorspheres, inhibited xCT function as assessed by reactive oxygen species generation and decreased BCSC growth and self-renewal. To assess if AX09-0M6 impacts BCSC in vivo seeding, Her2+ TUBO-derived tumorspheres were injected into the tail vein of AX09-0M6 or control treated female BALB/c mice. AX09-0M6 significantly inhibited formation of pulmonary nodules. To evaluate its ability to impact metastases, AX09-0M6 was administered to mice with established subcutaneous 4T1 tumors. AX09-0M6 administration significantly hampered tumor growth and development of pulmonary metastases. These data show that a VLP-based immunization approach inhibits xCT activity, impacts BCSC biology and significantly reduces metastatic progression in preclinical models.
Collapse
Affiliation(s)
- Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | | | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Stefania Lanzardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | | | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Marco Forni
- EuroClone S.p.A Research Laboratory, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | | | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
25
|
Insights into the molecular basis for substrate binding and specificity of the fungal cystine transporter CgCYN1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2259-2268. [DOI: 10.1016/j.bbamem.2017.08.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 08/15/2017] [Accepted: 08/27/2017] [Indexed: 11/21/2022]
|
26
|
Wang H, An P, Xie E, Wu Q, Fang X, Gao H, Zhang Z, Li Y, Wang X, Zhang J, Li G, Yang L, Liu W, Min J, Wang F. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology 2017; 66:449-465. [PMID: 28195347 PMCID: PMC5573904 DOI: 10.1002/hep.29117] [Citation(s) in RCA: 483] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 02/09/2017] [Indexed: 12/11/2022]
Abstract
UNLABELLED Ferroptosis is a recently identified iron-dependent form of nonapoptotic cell death implicated in brain, kidney, and heart pathology. However, the biological roles of iron and iron metabolism in ferroptosis remain poorly understood. Here, we studied the functional role of iron and iron metabolism in the pathogenesis of ferroptosis. We found that ferric citrate potently induces ferroptosis in murine primary hepatocytes and bone marrow-derived macrophages. Next, we screened for ferroptosis in mice fed a high-iron diet and in mouse models of hereditary hemochromatosis with iron overload. We found that ferroptosis occurred in mice fed a high-iron diet and in two knockout mouse lines that develop severe iron overload (Hjv-/- and Smad4Alb/Alb mice) but not in a third line that develops only mild iron overload (Hfe-/- mice). Moreover, we found that iron overload-induced liver damage was rescued by the ferroptosis inhibitor ferrostatin-1. To identify the genes involved in iron-induced ferroptosis, we performed microarray analyses of iron-treated bone marrow-derived macrophages. Interestingly, solute carrier family 7, member 11 (Slc7a11), a known ferroptosis-related gene, was significantly up-regulated in iron-treated cells compared with untreated cells. However, genetically deleting Slc7a11 expression was not sufficient to induce ferroptosis in mice. Next, we studied iron-treated hepatocytes and bone marrow-derived macrophages isolated from Slc7a11-/- mice fed a high-iron diet. CONCLUSION We found that iron treatment induced ferroptosis in Slc7a11-/- cells, indicating that deleting Slc7a11 facilitates the onset of ferroptosis specifically under high-iron conditions; these results provide compelling evidence that iron plays a key role in triggering Slc7a11-mediated ferroptosis and suggest that ferroptosis may be a promising target for treating hemochromatosis-related tissue damage. (Hepatology 2017;66:449-465).
Collapse
Affiliation(s)
- Hao Wang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Peng An
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Enjun Xie
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Qian Wu
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Xuexian Fang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Hong Gao
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Zhuzhen Zhang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Yuzhu Li
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Xudong Wang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Jiaying Zhang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Guoli Li
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Lei Yang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Wei Liu
- Department of Biochemistry and Molecular Biology, Program in Molecular and Cell Biology, School of MedicineZhejiang UniversityHangzhouChina
| | - Junxia Min
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| | - Fudi Wang
- School of Public Health, Zhengzhou University; School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesSchool of Medicine, Zhejiang UniversityHangzhouChina
| |
Collapse
|
27
|
Gu Y, Albuquerque CP, Braas D, Zhang W, Villa GR, Bi J, Ikegami S, Masui K, Gini B, Yang H, Gahman TC, Shiau AK, Cloughesy TF, Christofk HR, Zhou H, Guan KL, Mischel PS. mTORC2 Regulates Amino Acid Metabolism in Cancer by Phosphorylation of the Cystine-Glutamate Antiporter xCT. Mol Cell 2017. [PMID: 28648777 DOI: 10.1016/j.molcel.2017.05.030] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mutations in cancer reprogram amino acid metabolism to drive tumor growth, but the molecular mechanisms are not well understood. Using an unbiased proteomic screen, we identified mTORC2 as a critical regulator of amino acid metabolism in cancer via phosphorylation of the cystine-glutamate antiporter xCT. mTORC2 phosphorylates serine 26 at the cytosolic N terminus of xCT, inhibiting its activity. Genetic inhibition of mTORC2, or pharmacologic inhibition of the mammalian target of rapamycin (mTOR) kinase, promotes glutamate secretion, cystine uptake, and incorporation into glutathione, linking growth factor receptor signaling with amino acid uptake and utilization. These results identify an unanticipated mechanism regulating amino acid metabolism in cancer, enabling tumor cells to adapt to changing environmental conditions.
Collapse
Affiliation(s)
- Yuchao Gu
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Claudio P Albuquerque
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel Braas
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; UCLA Metabolomics Center, Los Angeles, CA 90095, USA
| | - Wei Zhang
- Department of Medicine, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Genaro R Villa
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; Medical Scientist Training Program, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Junfeng Bi
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shiro Ikegami
- Division of Neurological Surgery, Chiba Cancer Center, Chiba 260-8717, Japan
| | - Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Beatrice Gini
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Huijun Yang
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Timothy C Gahman
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrew K Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Heather R Christofk
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; UCLA Metabolomics Center, Los Angeles, CA 90095, USA
| | - Huilin Zhou
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093 USA
| | - Kun-Liang Guan
- Department of Pharmacology, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093 USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pathology, UCSD School of Medicine, La Jolla, CA 92093 USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093 USA.
| |
Collapse
|
28
|
A time-resolved molecular map of the macrophage response to VSV infection. NPJ Syst Biol Appl 2016; 2:16027. [PMID: 28725479 PMCID: PMC5516859 DOI: 10.1038/npjsba.2016.27] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 01/30/2023] Open
Abstract
Studying the relationship between virus infection and cellular response is paradigmatic for our understanding of how perturbation changes biological systems. Immune response, in this context is a complex yet evolutionarily adapted and robust cellular change, and is experimentally amenable to molecular analysis. To visualize the full cellular response to virus infection, we performed temporal transcriptomics, proteomics, and phosphoproteomics analysis of vesicular stomatitis virus (VSV)-infected mouse macrophages. This enabled the understanding of how infection-induced changes in host gene and protein expression are coordinated with post-translational modifications by cells in time to best measure and control the infection process. The vast and complex molecular changes measured could be decomposed in a limited number of clusters within each category (transcripts, proteins, and protein phosphorylation) each with own kinetic parameter and characteristic pathways/processes, suggesting multiple regulatory options in the overall sensing and homeostatic program. Altogether, the data underscored a prevalent executive function to phosphorylation. Resolution of the molecular events affecting the RIG-I pathway, central to viral recognition, reveals that phosphorylation of the key innate immunity adaptor mitochondrial antiviral-signaling protein (MAVS) on S328/S330 is necessary for activation of type-I interferon and nuclear factor κ B (NFκB) pathways. To further understand the hierarchical relationships, we analyzed kinase–substrate relationships and found RAF1 and, to a lesser extent, ARAF to be inhibiting VSV replication and necessary for NFκB activation, and AKT2, but not AKT1, to be supporting VSV replication. Integrated analysis using the omics data revealed co-regulation of transmembrane transporters including SLC7A11, which was subsequently validated as a host factor in the VSV replication. The data sets are predicted to greatly empower future studies on the functional organization of the response of macrophages to viral challenges.
Collapse
|
29
|
Rodríguez-Banqueri A, Errasti-Murugarren E, Bartoccioni P, Kowalczyk L, Perálvarez-Marín A, Palacín M, Vázquez-Ibar JL. Stabilization of a prokaryotic LAT transporter by random mutagenesis. ACTA ACUST UNITED AC 2016; 147:353-68. [PMID: 26976827 PMCID: PMC4810068 DOI: 10.1085/jgp.201511510] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/22/2016] [Indexed: 12/31/2022]
Abstract
The knowledge of three-dimensional structures at atomic resolution of membrane transport proteins has improved considerably our understanding of their physiological roles and pathological implications. However, most structural biology techniques require an optimal candidate within a protein family for structural determination with (a) reasonable production in heterologous hosts and (b) good stability in detergent micelles. SteT, the Bacillus subtilis L-serine/L-threonine exchanger is the best-known prokaryotic paradigm of the mammalian L-amino acid transporter (LAT) family. Unfortunately, SteT's lousy stability after extracting from the membrane prevents its structural characterization. Here, we have used an approach based on random mutagenesis to engineer stability in SteT. Using a split GFP complementation assay as reporter of protein expression and membrane insertion, we created a library of 70 SteT mutants each containing random replacements of one or two residues situated in the transmembrane domains. Analysis of expression and monodispersity in detergent of this library permitted the identification of evolved versions of SteT with a significant increase in both expression yield and stability in detergent with respect to wild type. In addition, these experiments revealed a correlation between the yield of expression and the stability in detergent micelles. Finally, and based on protein delipidation and relipidation assays together with transport experiments, possible mechanisms of SteT stabilization are discussed. Besides optimizing a member of the LAT family for structural determination, our work proposes a new approach that can be used to optimize any membrane protein of interest.
Collapse
Affiliation(s)
- Arturo Rodríguez-Banqueri
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Ekaitz Errasti-Murugarren
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Paola Bartoccioni
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain Spanish Biomedical Research Center in Rare Diseases (CIBERER), 08028 Barcelona, Spain
| | - Lukasz Kowalczyk
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Alex Perálvarez-Marín
- Biophysics Unit, Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, 08193 Cerdanyola del Vallés, Spain
| | - Manuel Palacín
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain Spanish Biomedical Research Center in Rare Diseases (CIBERER), 08028 Barcelona, Spain Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - José Luis Vázquez-Ibar
- Institute for Integrative Biology of the Cell (I2BC), CEA, French National Centre for Scientific Research (CNRS) UMR 9198, University Paris-Sud, University Paris-Saclay, F-91198 Gif-sur-Yvette, France
| |
Collapse
|
30
|
Patel D, Kharkar PS, Nandave M. Emerging roles of system antiporter and its inhibition in CNS disorders. Mol Membr Biol 2015; 32:89-116. [PMID: 26508554 DOI: 10.3109/09687688.2015.1096972] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
| | - Prashant S. Kharkar
- Department of Pharmaceutical Chemistry, SPP School of Pharmacy and Technology Management, SVKM’s NMIMS University, Mumbai, India
| | | |
Collapse
|
31
|
Van Liefferinge J, Bentea E, Demuyser T, Albertini G, Follin-Arbelet V, Holmseth S, Merckx E, Sato H, Aerts JL, Smolders I, Arckens L, Danbolt NC, Massie A. Comparative analysis of antibodies to xCT (Slc7a11): Forewarned is forearmed. J Comp Neurol 2015; 524:1015-32. [DOI: 10.1002/cne.23889] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Thomas Demuyser
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Virginie Follin-Arbelet
- Department of Molecular Medicine, Institute of Basic Medical Sciences; University of Oslo; Oslo 0317 Norway
| | - Silvia Holmseth
- Department of Molecular Medicine, Institute of Basic Medical Sciences; University of Oslo; Oslo 0317 Norway
| | - Ellen Merckx
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Hideyo Sato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Technology; Niigata University; Niigata Niigata Prefecture 950-2181 Japan
| | - Joeri L. Aerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| | - Lutgarde Arckens
- Laboratory of Neuroplasticity and Neuroproteomics; KU Leuven; Leuven 3000 Belgium
| | - Niels C. Danbolt
- Department of Molecular Medicine, Institute of Basic Medical Sciences; University of Oslo; Oslo 0317 Norway
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences; Vrije Universiteit Brussel; Brussels 1090 Belgium
| |
Collapse
|
32
|
Hinz KM, Meyer K, Kinne A, Schülein R, Köhrle J, Krause G. Structural insights into thyroid hormone transport mechanisms of the L-type amino acid transporter 2. Mol Endocrinol 2015; 29:933-42. [PMID: 25945809 DOI: 10.1210/me.2015-1044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Thyroid hormones (THs) are transported across cell membranes by different transmembrane transporter proteins. In previous studies, we showed marked 3,3'-diiodothyronine (3,3'-T2) but moderate T3 uptake by the L-type amino acid transporter 2 (Lat2). We have now studied the structure-function relationships of this transporter and TH-like molecules. Our Lat2 homology model is based on 2 crystal structures of the homologous 12-transmembrane helix transporters arginine/agmatine antiporter and amino acid/polyamine/organocation transporter. Model-driven mutagenesis of residues lining an extracellular recognition site and a TH-traversing channel identified 9 sensitive residues. Using Xenopus laevis oocytes as expression system, we found that side chain shortening (N51S, N133S, N248S, and Y130A) expanded the channel and increased 3,3'-T2 transport. Side chain enlargements (T140F, Y130R, and I137M) decreased 3,3'-T2 uptake, indicating channel obstructions. The opposite results with mutations maintaining (F242W) or impairing (F242V) uptake suggest that F242 may have a gating function. Competitive inhibition studies of 14 TH-like compounds revealed that recognition by Lat2 requires amino and carboxylic acid groups. The size of the adjacent hydrophobic group is restricted. Bulky substituents in positions 3 and 5 of the tyrosine ring are allowed. The phenolic ring may be enlarged, provided that the whole molecule is flexible enough to fit into the distinctly shaped TH-traversing channel of Lat2. Taken together, the next Lat2 features were identified 1) TH recognition site; 2) TH-traversing channel in the center of Lat2; and 3) switch site that potentially facilitates intracellular substrate release. Together with identified substrate features, these data help to elucidate the molecular mechanisms and role of Lat2 in T2 transport.
Collapse
Affiliation(s)
- Katrin M Hinz
- Leibniz-Institut für Molekulare Pharmakologie (FMP) (K.M.H., K.M., A.K., R.S., G.K.), 13125 Berlin, Germany; and Institut für Experimentelle Endokrinologie (J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Katja Meyer
- Leibniz-Institut für Molekulare Pharmakologie (FMP) (K.M.H., K.M., A.K., R.S., G.K.), 13125 Berlin, Germany; and Institut für Experimentelle Endokrinologie (J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Anita Kinne
- Leibniz-Institut für Molekulare Pharmakologie (FMP) (K.M.H., K.M., A.K., R.S., G.K.), 13125 Berlin, Germany; and Institut für Experimentelle Endokrinologie (J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Ralf Schülein
- Leibniz-Institut für Molekulare Pharmakologie (FMP) (K.M.H., K.M., A.K., R.S., G.K.), 13125 Berlin, Germany; and Institut für Experimentelle Endokrinologie (J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Josef Köhrle
- Leibniz-Institut für Molekulare Pharmakologie (FMP) (K.M.H., K.M., A.K., R.S., G.K.), 13125 Berlin, Germany; and Institut für Experimentelle Endokrinologie (J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Gerd Krause
- Leibniz-Institut für Molekulare Pharmakologie (FMP) (K.M.H., K.M., A.K., R.S., G.K.), 13125 Berlin, Germany; and Institut für Experimentelle Endokrinologie (J.K.), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
33
|
Tian X, Meng X, Wang L, Song Y, Zhang D, Ji Y, Li X, Dong C. Molecular cloning, mRNA expression and tissue distribution analysis of Slc7a11 gene in alpaca (Lama paco) skins associated with different coat colors. Gene 2015; 555:88-94. [DOI: 10.1016/j.gene.2014.10.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/13/2014] [Accepted: 10/16/2014] [Indexed: 12/01/2022]
|
34
|
Mesci P, Zaïdi S, Lobsiger CS, Millecamps S, Escartin C, Seilhean D, Sato H, Mallat M, Boillée S. System xC- is a mediator of microglial function and its deletion slows symptoms in amyotrophic lateral sclerosis mice. ACTA ACUST UNITED AC 2014; 138:53-68. [PMID: 25384799 DOI: 10.1093/brain/awu312] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis is the most common adult-onset motor neuron disease and evidence from mice expressing amyotrophic lateral sclerosis-causing SOD1 mutations suggest that neurodegeneration is a non-cell autonomous process where microglial cells influence disease progression. However, microglial-derived neurotoxic factors still remain largely unidentified in amyotrophic lateral sclerosis. With excitotoxicity being a major mechanism proposed to cause motor neuron death in amyotrophic lateral sclerosis, our hypothesis was that excessive glutamate release by activated microglia through their system [Formula: see text] (a cystine/glutamate antiporter with the specific subunit xCT/Slc7a11) could contribute to neurodegeneration. Here we show that xCT expression is enriched in microglia compared to total mouse spinal cord and absent from motor neurons. Activated microglia induced xCT expression and during disease, xCT levels were increased in both spinal cord and isolated microglia from mutant SOD1 amyotrophic lateral sclerosis mice. Expression of xCT was also detectable in spinal cord post-mortem tissues of patients with amyotrophic lateral sclerosis and correlated with increased inflammation. Genetic deletion of xCT in mice demonstrated that activated microglia released glutamate mainly through system [Formula: see text]. Interestingly, xCT deletion also led to decreased production of specific microglial pro-inflammatory/neurotoxic factors including nitric oxide, TNFa and IL6, whereas expression of anti-inflammatory/neuroprotective markers such as Ym1/Chil3 were increased, indicating that xCT regulates microglial functions. In amyotrophic lateral sclerosis mice, xCT deletion surprisingly led to earlier symptom onset but, importantly, this was followed by a significantly slowed progressive disease phase, which resulted in more surviving motor neurons. These results are consistent with a deleterious contribution of microglial-derived glutamate during symptomatic disease. Therefore, we show that system [Formula: see text] participates in microglial reactivity and modulates amyotrophic lateral sclerosis motor neuron degeneration, revealing system [Formula: see text] inactivation, as a potential approach to slow amyotrophic lateral sclerosis disease progression after onset of clinical symptoms.
Collapse
Affiliation(s)
- Pinar Mesci
- 1 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Sakina Zaïdi
- 1 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Christian S Lobsiger
- 1 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Stéphanie Millecamps
- 1 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Carole Escartin
- 2 CEA, DSV, I2BM, MIRCen and CNRS URA2210, Fontenay-aux-Roses, France
| | - Danielle Seilhean
- 1 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Hideyo Sato
- 3 Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata, Japan
| | - Michel Mallat
- 1 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Séverine Boillée
- 1 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| |
Collapse
|
35
|
Detergent-induced stabilization and improved 3D map of the human heteromeric amino acid transporter 4F2hc-LAT2. PLoS One 2014; 9:e109882. [PMID: 25299125 PMCID: PMC4192586 DOI: 10.1371/journal.pone.0109882] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/12/2014] [Indexed: 11/19/2022] Open
Abstract
Human heteromeric amino acid transporters (HATs) are membrane protein complexes that facilitate the transport of specific amino acids across cell membranes. Loss of function or overexpression of these transporters is implicated in several human diseases such as renal aminoacidurias and cancer. HATs are composed of two subunits, a heavy and a light subunit, that are covalently connected by a disulphide bridge. Light subunits catalyse amino acid transport and consist of twelve transmembrane α-helix domains. Heavy subunits are type II membrane N-glycoproteins with a large extracellular domain and are involved in the trafficking of the complex to the plasma membrane. Structural information on HATs is scarce because of the difficulty in heterologous overexpression. Recently, we had a major breakthrough with the overexpression of a recombinant HAT, 4F2hc-LAT2, in the methylotrophic yeast Pichia pastoris. Microgram amounts of purified protein made possible the reconstruction of the first 3D map of a human HAT by negative-stain transmission electron microscopy. Here we report the important stabilization of purified human 4F2hc-LAT2 using a combination of two detergents, i.e., n-dodecyl-β-D-maltopyranoside and lauryl maltose neopentyl glycol, and cholesteryl hemisuccinate. The superior quality and stability of purified 4F2hc-LAT2 allowed the measurement of substrate binding by scintillation proximity assay. In addition, an improved 3D map of this HAT could be obtained. The detergent-induced stabilization of the purified human 4F2hc-LAT2 complex presented here paves the way towards its crystallization and structure determination at high-resolution, and thus the elucidation of the working mechanism of this important protein complex at the molecular level.
Collapse
|
36
|
Soria FN, Pérez-Samartín A, Martin A, Gona KB, Llop J, Szczupak B, Chara JC, Matute C, Domercq M. Extrasynaptic glutamate release through cystine/glutamate antiporter contributes to ischemic damage. J Clin Invest 2014; 124:3645-55. [PMID: 25036707 DOI: 10.1172/jci71886] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 05/21/2014] [Indexed: 01/21/2023] Open
Abstract
During brain ischemia, an excessive release of glutamate triggers neuronal death through the overactivation of NMDA receptors (NMDARs); however, the underlying pathways that alter glutamate homeostasis and whether synaptic or extrasynaptic sites are responsible for excess glutamate remain controversial. Here, we monitored ischemia-gated currents in pyramidal cortical neurons in brain slices from rodents in response to oxygen and glucose deprivation (OGD) as a real-time glutamate sensor to identify the source of glutamate release and determined the extent of neuronal damage. Blockade of excitatory amino acid transporters or vesicular glutamate release did not inhibit ischemia-gated currents or neuronal damage after OGD. In contrast, pharmacological inhibition of the cystine/glutamate antiporter dramatically attenuated ischemia-gated currents and cell death after OGD. Compared with control animals, mice lacking a functional cystine/glutamate antiporter exhibited reduced anoxic depolarization and neuronal death in response to OGD. Furthermore, glutamate released by the cystine/glutamate antiporter activated extrasynaptic, but not synaptic, NMDARs, and blockade of extrasynaptic NMDARs reduced ischemia-gated currents and cell damage after OGD. Finally, PET imaging showed increased cystine/glutamate antiporter function in ischemic rats. Altogether, these data suggest that cystine/glutamate antiporter function is increased in ischemia, contributing to elevated extracellular glutamate concentration, overactivation of extrasynaptic NMDARs, and ischemic neuronal death.
Collapse
|
37
|
Nrf2- and ATF4-dependent upregulation of xCT modulates the sensitivity of T24 bladder carcinoma cells to proteasome inhibition. Mol Cell Biol 2014; 34:3421-34. [PMID: 25002527 DOI: 10.1128/mcb.00221-14] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The ubiquitin-proteasome pathway degrades ubiquitinated proteins to remove damaged or misfolded protein and thus plays an important role in the maintenance of many important cellular processes. Because the pathway is also crucial for tumor cell growth and survival, proteasome inhibition by specific inhibitors exhibits potent antitumor effects in many cancer cells. xCT, a subunit of the cystine antiporter system xc (-), plays an important role in cellular cysteine and glutathione homeostasis. Several recent reports have revealed that xCT is involved in cancer cell survival; however, it was unknown whether xCT affects the cytotoxic effects of proteasome inhibitors. In this study, we found that two stress-inducible transcription factors, Nrf2 and ATF4, were upregulated by proteasome inhibition and cooperatively enhance human xCT gene expression upon proteasome inhibition. In addition, we demonstrated that the knockdown of xCT by small interfering RNA (siRNA) or pharmacological inhibition of xCT by sulfasalazine (SASP) or (S)-4-carboxyphenylglycine (CPG) significantly increased the sensitivity of T24 cells to proteasome inhibition. These results suggest that the simultaneous inhibition of both the proteasome and xCT could have therapeutic benefits in the treatment of bladder tumors.
Collapse
|
38
|
Structural bases for the interaction and stabilization of the human amino acid transporter LAT2 with its ancillary protein 4F2hc. Proc Natl Acad Sci U S A 2014; 111:2966-71. [PMID: 24516142 DOI: 10.1073/pnas.1323779111] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Heteromeric amino acid transporters (HATs) are the unique example, known in all kingdoms of life, of solute transporters composed of two subunits linked by a conserved disulfide bridge. In metazoans, the heavy subunit is responsible for the trafficking of the heterodimer to the plasma membrane, and the light subunit is the transporter. HATs are involved in human pathologies such as amino acidurias, tumor growth and invasion, viral infection and cocaine addiction. However structural information about interactions between the heavy and light subunits of HATs is scarce. In this work, transmission electron microscopy and single-particle analysis of purified human 4F2hc/L-type amino acid transporter 2 (LAT2) heterodimers overexpressed in the yeast Pichia pastoris, together with docking analysis and crosslinking experiments, reveal that the extracellular domain of 4F2hc interacts with LAT2, almost completely covering the extracellular face of the transporter. 4F2hc increases the stability of the light subunit LAT2 in detergent-solubilized Pichia membranes, allowing functional reconstitution of the heterodimer into proteoliposomes. Moreover, the extracellular domain of 4F2hc suffices to stabilize solubilized LAT2. The interaction of 4F2hc with LAT2 gives insights into the structural bases for light subunit recognition and the stabilizing role of the ancillary protein in HATs.
Collapse
|
39
|
Newell JL, Keyari CM, McDaniel SW, Diaz PJ, Natale NR, Patel SA, Bridges RJ. Novel di-aryl-substituted isoxazoles act as noncompetitive inhibitors of the system Xc(-) cystine/glutamate exchanger. Neurochem Int 2013; 73:132-8. [PMID: 24333322 DOI: 10.1016/j.neuint.2013.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/26/2013] [Accepted: 11/29/2013] [Indexed: 01/18/2023]
Abstract
The system xc(-) antiporter is a plasma membrane transporter that mediates the exchange of extracellular l-cystine with intracellular l-glutamate. This exchange is significant within the context of the CNS because the import of l-cystine is required for the synthesis of the antioxidant glutathione, while the efflux of l-glutamate has the potential to contribute to either excitatory signaling or excitotoxic pathology. Changes in the activity of the transport system have been linked to the underlying pathological mechanisms of a variety of CNS disorders, one of the most prominent of which is its highly enriched expression in glial brain tumors. In an effort to produce more potent system xc(-) blockers, we have been using amino-3-carboxy-5-methylisoxazole propionic acid (ACPA) as a scaffold for inhibitor development. We previously demonstrated that the addition of lipophilic aryl groups to either the #4 or #5 position on the isoxazole ring markedly increased the inhibitory activity at system xc(-). In the present work a novel series of analogues has been prepared in which aryl groups have been introduced at both the #4 and #5 positions. In contrast to the competitive action of the mono-substituted analogues, kinetic analyses indicate that the di-substituted isoxazoles block system xc(-)-mediated uptake of (3)H-l-glutamate into SNB-19 cells by a noncompetitive mechanism. These new analogues appear to be the first noncompetitive inhibitors identified for this transport system, as well as being among the most potent blockers identified to date. These diaryl-isoxazoles should be of value in assessing the physiological roles and molecular pharmacology of system xc(-).
Collapse
Affiliation(s)
- J L Newell
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - C M Keyari
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - S W McDaniel
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - P J Diaz
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - N R Natale
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - S A Patel
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - R J Bridges
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States.
| |
Collapse
|
40
|
The SLC3 and SLC7 families of amino acid transporters. Mol Aspects Med 2013; 34:139-58. [PMID: 23506863 DOI: 10.1016/j.mam.2012.10.007] [Citation(s) in RCA: 499] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/15/2012] [Indexed: 01/18/2023]
Abstract
Amino acids are necessary for all living cells and organisms. Specialized transporters mediate the transfer of amino acids across plasma membranes. Malfunction of these proteins can affect whole-body homoeostasis giving raise to diverse human diseases. Here, we review the main features of the SLC3 and SLC7 families of amino acid transporters. The SLC7 family is divided into two subfamilies, the cationic amino acid transporters (CATs), and the L-type amino acid transporters (LATs). The latter are the light or catalytic subunits of the heteromeric amino acid transporters (HATs), which are associated by a disulfide bridge with the heavy subunits 4F2hc or rBAT. These two subunits are glycoproteins and form the SLC3 family. Most CAT subfamily members were functionally characterized and shown to function as facilitated diffusers mediating the entry and efflux of cationic amino acids. In certain cells, CATs play an important role in the delivery of L-arginine for the synthesis of nitric oxide. HATs are mostly exchangers with a broad spectrum of substrates and are crucial in renal and intestinal re-absorption and cell redox balance. Furthermore, the role of the HAT 4F2hc/LAT1 in tumor growth and the application of LAT1 inhibitors and PET tracers for reduction of tumor progression and imaging of tumors are discussed. Finally, we describe the link between specific mutations in HATs and the primary inherited aminoacidurias, cystinuria and lysinuric protein intolerance.
Collapse
|
41
|
Matti AA, Mirzaei J, Rudolph J, Smith SA, Newell JL, Patel SA, Braden MR, Bridges RJ, Natale NR. Microwave accelerated synthesis of isoxazole hydrazide inhibitors of the system xc- transporter: Initial homology model. Bioorg Med Chem Lett 2013; 23:5931-5. [PMID: 24042010 DOI: 10.1016/j.bmcl.2013.08.080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 08/16/2013] [Accepted: 08/19/2013] [Indexed: 01/18/2023]
Abstract
Microwave accelerated reaction system (MARS) technology provided a good method to obtain selective and open isoxazole ligands that bind to and inhibit the Sxc- antiporter. The MARS provided numerous advantages, including: shorter time, better yield and higher purity of the product. Of the newly synthesized series of isoxazoles the salicyl hydrazide 6 exhibited the highest level of inhibitory activity in the transport assay. A homology model has been developed to summarize the SAR results to date, and provide a working hypothesis for future studies.
Collapse
Affiliation(s)
- Afnan A Matti
- Department of Biomedical & Pharmaceutical Sciences, Center for Structural & Functional Neuroscience, The University of Montana, Missoula, MT 59812, United States; Medicinal Chemistry Graduate Program, The University of Montana, Missoula, MT 59812, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, Smith SB, Ganapathy V, Maher P. The cystine/glutamate antiporter system x(c)(-) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal 2013; 18:522-55. [PMID: 22667998 PMCID: PMC3545354 DOI: 10.1089/ars.2011.4391] [Citation(s) in RCA: 730] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The antiporter system x(c)(-) imports the amino acid cystine, the oxidized form of cysteine, into cells with a 1:1 counter-transport of glutamate. It is composed of a light chain, xCT, and a heavy chain, 4F2 heavy chain (4F2hc), and, thus, belongs to the family of heterodimeric amino acid transporters. Cysteine is the rate-limiting substrate for the important antioxidant glutathione (GSH) and, along with cystine, it also forms a key redox couple on its own. Glutamate is a major neurotransmitter in the central nervous system (CNS). By phylogenetic analysis, we show that system x(c)(-) is a rather evolutionarily new amino acid transport system. In addition, we summarize the current knowledge regarding the molecular mechanisms that regulate system x(c)(-), including the transcriptional regulation of the xCT light chain, posttranscriptional mechanisms, and pharmacological inhibitors of system x(c)(-). Moreover, the roles of system x(c)(-) in regulating GSH levels, the redox state of the extracellular cystine/cysteine redox couple, and extracellular glutamate levels are discussed. In vitro, glutamate-mediated system x(c)(-) inhibition leads to neuronal cell death, a paradigm called oxidative glutamate toxicity, which has successfully been used to identify neuroprotective compounds. In vivo, xCT has a rather restricted expression pattern with the highest levels in the CNS and parts of the immune system. System x(c)(-) is also present in the eye. Moreover, an elevated expression of xCT has been reported in cancer. We highlight the diverse roles of system x(c)(-) in the regulation of the immune response, in various aspects of cancer and in the eye and the CNS.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department of Neurology, University of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Expression of human heteromeric amino acid transporters in the yeast Pichia pastoris. Protein Expr Purif 2012; 87:35-40. [PMID: 23085088 DOI: 10.1016/j.pep.2012.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/08/2012] [Accepted: 10/08/2012] [Indexed: 11/20/2022]
Abstract
Human heteromeric amino acid transporters (HATs) play key roles in renal and intestinal re-absorption, cell redox balance and tumor growth. These transporters are composed of a heavy and a light subunit, which are connected by a disulphide bridge. Heavy subunits are the two type II membrane N-glycoproteins rBAT and 4F2hc, while L-type amino acid transporters (LATs) are the light and catalytic subunits of HATs. We tested the expression of human 4F2hc and rBAT as well as seven light subunits in the methylotrophic yeast Pichia pastoris. 4F2hc and the light subunit LAT2 showed the highest expression levels and yields after detergent solubilization. Co-transformation of both subunits in Pichia cells resulted in overexpression of the disulphide bridge-linked 4F2hc/LAT2 heterodimer. Two sequential affinity chromatography steps were applied to purify detergent-solubilized heterodimers yielding ~1mg of HAT from 2l of cell culture. Our results indicate that P. pastoris is a convenient system for the expression and purification of human 4F2hc/LAT2 for structural studies.
Collapse
|
44
|
Bridges RJ, Natale NR, Patel SA. System xc⁻ cystine/glutamate antiporter: an update on molecular pharmacology and roles within the CNS. Br J Pharmacol 2012; 165:20-34. [PMID: 21564084 DOI: 10.1111/j.1476-5381.2011.01480.x] [Citation(s) in RCA: 407] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
System x(c)(-) is an amino acid antiporter that typically mediates the exchange of extracellular l-cystine and intracellular L-glutamate across the cellular plasma membrane. Studied in a variety of cell types, the import of L-cystine through this transporter is critical to glutathione production and oxidative protection. The exchange-mediated export of L-glutamate takes on added significance within the CNS, as it represents a non-vesicular route of release through which this excitatory neurotransmitter can participate in either neuronal signalling or excitotoxic pathology. When both the import of L-cystine and the export of L-glutamate are taken into consideration, system x(c)(-) has now been linked to a wide range of CNS functions, including oxidative protection, the operation of the blood-brain barrier, neurotransmitter release, synaptic organization, viral pathology, drug addiction, chemosensitivity and chemoresistance, and brain tumour growth. The ability to selectively manipulate system x(c)(-), delineate its function, probe its structure and evaluate it as a therapeutic target is closely linked to understanding its pharmacology and the subsequent development of selective inhibitors and substrates. Towards that goal, this review will examine the current status of our understanding of system x(c)(-) pharmacology and the structure-activity relationships that have guided the development of an initial pharmacophore model, including the presence of lipophilic domains adjacent to the substrate binding site. A special emphasis is placed on the roles of system x(c)(-) within the CNS, as it is these actions that are among the most exciting as potential long-range therapeutic targets.
Collapse
Affiliation(s)
- Richard J Bridges
- Center for Structural and Functional Neuroscience, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana 59812, USA.
| | | | | |
Collapse
|
45
|
He X, Li H, Zhou Z, Zhao Z, Li W. Production of brown/yellow patches in the SLC7A11 transgenic sheep via testicular injection of transgene. J Genet Genomics 2012; 39:281-5. [PMID: 22749016 DOI: 10.1016/j.jgg.2012.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 04/27/2012] [Accepted: 04/30/2012] [Indexed: 10/28/2022]
Abstract
The gene, SLC7A11, which encodes the solute carrier family 7 member 11 (anionic amino acid transporter light chain, xCT), has been reported to be implicated in multiple processes such as in pheomelanin production, cell proliferation and migration, Kaposi's sarcoma herpesvirus (KSHV) entry into the host cells, learning and memory. Its involvement in cancer cell proliferation and metastasis has been widely studied. Its role in pheomelanogenesis is likely conserved in sheep. The full-length cDNA of sheep SLC7A11 was cloned from sheep skin fibroblasts for evaluating its role in regulating sheep coat color. The complete open reading frame of sheep xCT (sxCT) is 1512 bp in length, encoding a 503 amino acid polypeptide. We explored its function on pheomelanogenesis in vitro and in vivo. In the melan-a non-agouti mouse melanocytes that mainly produce eumelanin, overexpressed sxCT reduced the content of eumelanin. Using a testicular injection transgenic method, sxCT-transgenic sheep were generated and exhibited patches of brown/yellow coat, suggesting that sxCT can be selectively expressed to increase the pheomelanin production in wool. Our studies suggest that testicular injection of transgene can be used to genetically modify sheep coat color.
Collapse
Affiliation(s)
- Xin He
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | |
Collapse
|
46
|
Abstract
Thyroid hormones (TH) are essential for the development of the human brain, growth and cellular metabolism. Investigation of TH transporters became one of the emerging fields in thyroid research after the discovery of inactivating mutations in the Monocarboxylate transporter 8 (MCT8), which was found to be highly specific for TH transport. However, additional transmembrane transporters are also very important for TH uptake and efflux in different cell types. They transport TH as secondary substrates and include the aromatic amino acid transporting MCT10, the organic anion transporting polypeptides (e.g. OATP1C1, OATP1A2, OPTP1A4) and the large neutral amino acid transporters (LAT1 and LAT2). These TH transporters characteristically possess 12 transmembrane spanners but due to the strong differing sequences between the three transporter families we assume an identical conformation is not very likely. In contrast to the others, the LAT family members form a heterodimer with the escort protein 4F2hc/CD98. A comparison of sequence proportions, locations and types of functional sensitive features for TH transport discovered by mutations, revealed that transport sensitive charged residues occur as conserved amino acids only within each family of the transporter types but not in all putative TH transporters. Based on the lack of highly conserved sensitive charged residues throughout the three transporter families as a common counterpart for the amino acid moiety of the substrates, we conclude that the molecular transport mechanism is likely organized either a) by different molecular determinants in the divergent transporter types or b) the counterparts for the substrates` amino acid moiety at the transporter are not any charged side chains but other proton acceptors or donators. However, positions of transport sensitive residues coincide at transmembrane helix 8 in the TH transporter MCT8, OATP1C1 and another amino acid transporter, the L-cystine and L-glutamate exchanger xCT, which is highly homologous to LAT1 and LAT2. Here we review the data available and compare similarities and differences between these primary and secondary TH transporters regarding sequences, topology, potential structures, trafficking to the plasma membrane, molecular features and locations of transport sensitive functionalities. Thereby, we focus on TH transporters occurring in the blood-brain barrier.
Collapse
Affiliation(s)
- Anita Kinne
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Roessle-Str, 10, 13125 Berlin, Germany.
| | | | | |
Collapse
|
47
|
Abstract
Amino acids are essential building blocks of all mammalian cells. In addition to their role in protein synthesis, amino acids play an important role as energy fuels, precursors for a variety of metabolites and as signalling molecules. Disorders associated with the malfunction of amino acid transporters reflect the variety of roles that they fulfil in human physiology. Mutations of brain amino acid transporters affect neuronal excitability. Mutations of renal and intestinal amino acid transporters affect whole-body homoeostasis, resulting in malabsorption and renal problems. Amino acid transporters that are integral parts of metabolic pathways reduce the function of these pathways. Finally, amino acid uptake is essential for cell growth, thereby explaining their role in tumour progression. The present review summarizes the involvement of amino acid transporters in these roles as illustrated by diseases resulting from transporter malfunction.
Collapse
|
48
|
Costello LC, Fenselau CC, Franklin RB. Evidence for operation of the direct zinc ligand exchange mechanism for trafficking, transport, and reactivity of zinc in mammalian cells. J Inorg Biochem 2011; 105:589-99. [PMID: 21440525 PMCID: PMC3081963 DOI: 10.1016/j.jinorgbio.2011.02.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 11/28/2022]
Abstract
In addition to its critical role in normal cell function, growth, and metabolism, zinc is implicated as a major factor in the development and progression of many pathological conditions and diseases. Despite this importance of zinc, many important factors, processes, and mechanisms of the physiology, biochemistry, and molecular biology of zinc remain unknown. Especially important is the unresolved issue regarding the mechanism and process of the trafficking, transport, and reactivity of zinc in cells; especially in mammalian cells. This presentation focuses on the concept that, due to the existence of a negligible pool of free Zn(2+) ions in the mammalian cell environment, the trafficking, transport and reactivity of zinc occurs via a direct exchange of zinc from donor Zn-ligands to acceptor ligands. This Zn exchange process occurs without the requirement for production of free Zn(2+) ions. The direct evidence from mammalian cell studies is presented in support of the operation of the direct Zn-ligand exchange mechanism. The paper also provides important information and conditions that should be considered and employed in the conduct of studies regarding the role and effects of zinc in biological/biomedical research; and in its clinical interpretation and application.
Collapse
Affiliation(s)
- Leslie C. Costello
- Department of Oncology and Diagnostic Sciences, Dental School; and The Greenebaum Cancer Center; University of Maryland; Baltimore, Maryland, USA 21201
| | - Catherine C. Fenselau
- Department of Chemistry and Biochemistry; University of Maryland; College Park, Maryland, USA 20742; and The Greenebaum Cancer Center; University of Maryland; Baltimore, Maryland, USA 21201
| | - Renty B. Franklin
- Department of Oncology and Diagnostic Sciences, Dental School; and The Greenebaum Cancer Center; University of Maryland; Baltimore, Maryland, USA 21201
| |
Collapse
|
49
|
Conrad M, Sato H. The oxidative stress-inducible cystine/glutamate antiporter, system x (c) (-) : cystine supplier and beyond. Amino Acids 2011; 42:231-46. [PMID: 21409388 DOI: 10.1007/s00726-011-0867-5] [Citation(s) in RCA: 446] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 02/17/2011] [Indexed: 12/17/2022]
Abstract
The oxidative stress-inducible cystine/glutamate exchange system, system x (c) (-) , transports one molecule of cystine, the oxidized form of cysteine, into cells and thereby releases one molecule of glutamate into the extracellular space. It consists of two protein components, the 4F2 heavy chain, necessary for membrane location of the heterodimer, and the xCT protein, responsible for transport activity. Previously, system x (c) (-) has been regarded to be a mere supplier of cysteine to cells for the synthesis of proteins and the antioxidant glutathione (GSH). In that sense, oxygen, electrophilic agents, and bacterial lipopolysaccharide trigger xCT expression to accommodate with increased oxidative stress by stimulating GSH biosynthesis. However, emerging evidence established that system x (c) (-) may act on its own as a GSH-independent redox system by sustaining a redox cycle over the plasma membrane. Hallmarks of this cycle are cystine uptake, intracellular reduction to cysteine and secretion of the surplus of cysteine into the extracellular space. Consequently, increased levels of extracellular cysteine provide a reducing microenvironment required for proper cell signaling and communication, e.g. as already shown for the mechanism of T cell activation. By contrast, the enhanced release of glutamate in exchange with cystine may trigger neurodegeneration due to glutamate-induced cytotoxic processes. This review aims to provide a comprehensive picture from the early days of system x (c) (-) research up to now.
Collapse
Affiliation(s)
- Marcus Conrad
- DZNE, German Center for Neurodegenerative Diseases, Munich, 80336, Munich, Germany.
| | | |
Collapse
|
50
|
Lewerenz J, Maher P, Methner A. Regulation of xCT expression and system x (c) (-) function in neuronal cells. Amino Acids 2011; 42:171-9. [PMID: 21369940 DOI: 10.1007/s00726-011-0862-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 02/17/2011] [Indexed: 12/14/2022]
Abstract
The glutamate/cystine antiporter system x(c)(-) transports cystine into cells in exchange for glutamate at a ratio of 1:1. It is composed of a specific light chain, xCT, and a heavy chain, 4F2, linked by a disulfide bridge. Intracellularly, cystine is reduced into cysteine, the rate-limiting precursor of glutathione (GSH), an important small molecule antioxidant. Several lines of evidence suggest that the expression of xCT and thereby the presence system x(c)(-) activity plays an important role in the brain. First, it regulates extracellular glutamate concentrations. Second, as brain is prone to oxidative stress due to its high oxygen consumption and lipid content, system x(c)(-) by favoring GSH synthesis, may prevent oxidative damage. Thus, to understand how xCT expression and system x(c)(-) activity are regulated in the central nervous system is of utmost importance. In this review, we will summarize the current knowledge about the molecular basis by which xCT expression and system x(c)(-) activity are regulated in neuronal cell lines, especially the hippocampal cell line, HT22. In addition, we will relate these pathways to findings in other cell types, especially those found in the central nervous system. We will focus on the signaling pathways that modulate the transcription of the xCT gene. Furthermore, we describe possible pathways that modify system x(c)(-) activity beyond the level of xCT transcription, including regulation on the level of membrane trafficking and substrate availability, especially the regulation by glutamate transport through excitatory amino acid transporters.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20241, Hamburg, Germany.
| | | | | |
Collapse
|