1
|
Ma J, Li Z, Xu J, Lai J, Zhao J, Ma L, Sun X. PRDM1 promotes the ferroptosis and immune escape of thyroid cancer by regulating USP15-mediated SELENBP1 deubiquitination. J Endocrinol Invest 2024; 47:2981-2997. [PMID: 39014173 DOI: 10.1007/s40618-024-02385-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/25/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND The deubiquitinating enzyme Ubiquitin-specific peptidase 15 (USP15) is upregulated in various cancers and promotes tumor progression by increasing the expression of several oncogenes. This project is designed to explore the role and mechanism of USP15 in thyroid cancer (TC) progression. METHODS Selenium-binding protein 1 (SELENBP1), USP15, CCL2/5, CXCL10/11, IL-4, and TGF-β1 mRNA levels were detected using real-time quantitative polymerase chain reaction (RT-qPCR). SELENBP1, USP15, GPX4, IL-10, Arg-1, Granzyme B, TNF-α, and PR domain zinc finger protein 1 (PRDM1) protein levels were examined by western blot assay. Fe+ level, malondialdehyde (MDA), and lipid-ROS levels were determined using special kits. The proportion of CD11b+CD206+ positive cells was detected using a flow cytometry assay. The role of SELENBP1 on TC cell growth was examined using a xenograft tumor model in vivo. After GeneMANIA prediction, the interaction between USP15 and SELENBP1 was verified using Co-immunoprecipitation (CoIP) assay. The binding between PRDM1 and USP15 promoter was predicted by JASPAR and validated using Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays. RESULTS SELENBP1 was increased in TC subjects and cell lines, and its knockdown repressed TC cell proliferation, migration, invasion, immune escape, and induced ferroptosis in vitro, as well as blocked tumor growth in vivo. In mechanism, USP15 interacted with SELENBP1 and maintained its stabilization by removing ubiquitin. Meanwhile, the upregulation of USP15 was induced by the transcription factor PRDM1. CONCLUSION USP15 transcriptionally mediated by PRDM1 might boost TC cell malignant behaviors through deubiquitinating SELENBP1, providing a promising therapeutic target for TC treatment.
Collapse
Affiliation(s)
- J Ma
- Department of Vascular Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710004, Shaanxi, China
| | - Z Li
- Department of Vascular Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710004, Shaanxi, China
| | - J Xu
- Department of General Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710004, Shaanxi, China
| | - J Lai
- Department of Vascular Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710004, Shaanxi, China
| | - J Zhao
- Department of General Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710004, Shaanxi, China
| | - L Ma
- Department of Laboratory Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, 710061, Shaanxi, China
| | - X Sun
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an City, 710061, Shaanxi, China.
| |
Collapse
|
2
|
Liang B, Lin W, Tang Y, Li T, Chen Q, Zhang W, Zhou X, Ma J, Liu B, Yu Z, Zha L, Zhang M. Selenium supplementation elevated SELENBP1 to inhibit fibroblast activation in pulmonary arterial hypertension. iScience 2024; 27:111036. [PMID: 39435142 PMCID: PMC11492086 DOI: 10.1016/j.isci.2024.111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/28/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease induced by abnormal activation of pulmonary adventitial fibroblasts (PAFs) in the early stage. The association between selenium deficiency and PAH is not yet fully understood. In this study, we found that the serum selenium content of PAH patients was significantly lower than that of healthy volunteers in two independent cohorts. Moreover, PAH patients with lower selenium levels may present poorer prognosis. Prophylactic selenium supplementation could effectively improve hemodynamics and pulmonary vascular remodeling in monocrotaline-induced pulmonary hypertension rat models. Mechanistically, selenium supplementation restored the level of selenium binding protein 1 (SELENBP1) which could exert an antagonistic effect on PAF activation. The rescue assay further proved that selenium supplementation worked in a SELENBP1-dependent manner. These findings demonstrated that selenium deficiency is an important risk factor in PAH, and the selenium-SELENBP1 axis represents a promising target for PAH prevention.
Collapse
Affiliation(s)
- Benhui Liang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenchao Lin
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiyang Tang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tangzhiming Li
- Department of Cardiology, Shenzhen People’s Hospital, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Guangzhou, Guangdong, China
| | - Qin Chen
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Zhang
- Department of Cardiology, Xiangya Third Hospital, Central South University, Changsha, Hunan, China
| | - Xinyi Zhou
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiayao Ma
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Boqing Liu
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zaixin Yu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lihuang Zha
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengqiu Zhang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Dervisi I, Koletti A, Agalou A, Haralampidis K, Flemetakis E, Roussis A. Selenium-Binding Protein 1 (SBP1): A New Putative Player of Stress Sensing in Plants. Int J Mol Sci 2024; 25:9372. [PMID: 39273319 PMCID: PMC11394908 DOI: 10.3390/ijms25179372] [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: 08/04/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Selenium-binding proteins (SBPs) represent a ubiquitous and conserved protein family with yet unclear biochemical and molecular functions. The importance of the human homolog has been extensively studied as it is implicated in many cancer types and other diseases. On the other hand, little is known regarding plant homologs. In plants, there is evidence that SBP participates in developmental procedures, oxidative stress responses, selenium and cadmium binding, and pathogenic tolerance. Moreover, recent studies have revealed that SBP is a methanethiol oxidase (MTO) catalyzing the conversion of methanethiol into formaldehyde, H2S, and H2O2. The two later products emerge as key signal molecules, playing pivotal roles in physiological processes and environmental stress responses. In this review, we highlight the available information regarding plants in order to introduce and emphasize the importance of SBP1 and its role in plant growth, development, and abiotic/biotic stress.
Collapse
Affiliation(s)
- Irene Dervisi
- Department of Botany, Faculty of Biology, National & Kapodistrian University of Athens, 15784 Athens, Greece; (I.D.)
| | - Aikaterini Koletti
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (A.K.); (E.F.)
| | - Adamantia Agalou
- Laboratory of Toxicological Control of Pesticides, Scientific Directorate of Pesticides’ Control & Phytopharmacy, Benaki Phytopathological Institute (BPI), 14561 Athens, Greece;
| | - Kosmas Haralampidis
- Department of Botany, Faculty of Biology, National & Kapodistrian University of Athens, 15784 Athens, Greece; (I.D.)
| | - Emmanouil Flemetakis
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (A.K.); (E.F.)
| | - Andreas Roussis
- Department of Botany, Faculty of Biology, National & Kapodistrian University of Athens, 15784 Athens, Greece; (I.D.)
| |
Collapse
|
4
|
Turan TL, Klein HJ, Hackler J, Hoerner L, Rijntjes E, Graf TR, Plock JA, Schomburg L. Serum Selenium-Binding Protein 1 (SELENBP1) in Burn Injury: A Potential Biomarker of Disease Severity and Clinical Course. Antioxidants (Basel) 2023; 12:1927. [PMID: 38001780 PMCID: PMC10669776 DOI: 10.3390/antiox12111927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Oxidative stress, systemic inflammation, and metabolic derangements are hallmarks of burn pathophysiology. Severely burned patients are highly susceptible to infectious complications. Selenium-binding protein 1 (SELENBP1) modulates intracellular redox homeostasis, and elevated serum concentrations have been associated with adverse clinical outcomes in trauma patients. We hypothesized that serum SELENBP1 at hospital admission and during hospitalization may constitute a meaningful biomarker of disease severity and the clinical course in burn injury, with pulmonary infection as primary endpoint. To this end, we conducted a prospective cohort study that included 90 adult patients admitted to the Burn Center of the University Hospital Zurich, Switzerland. Patients were treated according to the local standard of care, with high-dose selenium supplementation during the first week. Serum SELENBP1 was determined at nine time-points up to six months postburn and the data were correlated to clinical parameters. SELENBP1 was initially elevated and rapidly declined within the first day. Baseline SELENBP1 levels correlated positively with the Abbreviated Burn Severity Index (ABSI) (R = 0.408; p < 0.0001). In multiple logistic regression, a higher ABSI was significantly associated with increased pulmonary infection risk (OR, 14.4; 95% CI, 3.2-88.8; p = 0.001). Similarly, baseline SELENBP1 levels constituted a novel but less accurate predictor of pulmonary infection risk (OR, 2.5; 95% CI, 0.7-8.9; p = 0.164). Further studies are needed to explore the additional value of serum SELENBP1 when stratifying patients with respect to the clinical course following major burns and, potentially, for monitoring therapeutic measures aimed at reducing tissue damage and oxidative stress.
Collapse
Affiliation(s)
- Tabael L. Turan
- Institute for Experimental Endocrinology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (T.L.T.); (L.H.); (E.R.)
| | - Holger J. Klein
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (H.J.K.); (J.A.P.)
- Department of Plastic Surgery and Hand Surgery, Cantonal Hospital Aarau, 5001 Aarau, Switzerland
| | - Julian Hackler
- Institute for Experimental Endocrinology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (T.L.T.); (L.H.); (E.R.)
| | - Livia Hoerner
- Institute for Experimental Endocrinology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (T.L.T.); (L.H.); (E.R.)
| | - Eddy Rijntjes
- Institute for Experimental Endocrinology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (T.L.T.); (L.H.); (E.R.)
| | - Theresia Reding Graf
- Department of Visceral Surgery and Transplantation, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Jan A. Plock
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (H.J.K.); (J.A.P.)
- Department of Plastic Surgery and Hand Surgery, Cantonal Hospital Aarau, 5001 Aarau, Switzerland
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (T.L.T.); (L.H.); (E.R.)
| |
Collapse
|
5
|
Maritz-Olivier C, Ferreira M, Olivier NA, Crafford J, Stutzer C. Mining gene expression data for rational identification of novel drug targets and vaccine candidates against the cattle tick, Rhipicephalus microplus. EXPERIMENTAL & APPLIED ACAROLOGY 2023; 91:291-317. [PMID: 37755526 PMCID: PMC10562289 DOI: 10.1007/s10493-023-00838-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/09/2023] [Indexed: 09/28/2023]
Abstract
Control of complex parasites via vaccination remains challenging, with the current combination of vaccines and small drugs remaining the choice for an integrated control strategy. Studies conducted to date, are providing evidence that multicomponent vaccines will be needed for the development of protective vaccines against endo- and ectoparasites, though multicomponent vaccines require an in-depth understanding of parasite biology which remains insufficient for ticks. With the rapid development and spread of acaricide resistance in ticks, new targets for acaricide development also remains to be identified, along with novel targets that can be exploited for the design of lead compounds. In this study, we analysed the differential gene expression of Rhipicephalus microplus ticks that were fed on cattle vaccinated with a multi-component vaccine (Bm86 and 3 putative Bm86-binding proteins). The data was scrutinised for the identification of vaccine targets, small drug targets and novel pathways that can be evaluated in future studies. Limitations associated with targeting novel proteins for vaccine and/or drug design is also discussed and placed into the context of challenges arising when targeting large protein families and intracellular localised proteins. Lastly, this study provide insight into how Bm86-based vaccines may reduce successful uptake and digestion of the bloodmeal and overall tick fecundity.
Collapse
Affiliation(s)
- Christine Maritz-Olivier
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, Gauteng, South Africa.
| | - Mariëtte Ferreira
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Nicholas A Olivier
- DNA Microarray Laboratory, Department of Plant Sciences, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Jan Crafford
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Christian Stutzer
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, Gauteng, South Africa.
| |
Collapse
|
6
|
Withdrawal Notice. Cancer Med 2023; 12:19353. [PMID: 36372952 PMCID: PMC10557851 DOI: 10.1002/cam4.5306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Withdrawal Notice: Zhu, Y, Pu, Q, Zhang, Q, et al. Selenium-binding protein 1 inhibits malignant progression and induces apoptosis via distinct mechanisms in non-small-cell lung cancer. Cancer Med. 2022; 00: 1-22. doi: 10.1002/cam4.5306. The above article, published online on 13th November 2022 in Wiley Online Library (https://onlinelibrary.wiley.com/doi/10.1002/cam4.5306), has been withdrawn by agreement between the journal Editor in Chief, Dr Stephen Tait, the Authors, and John Wiley & Sons, Ltd. The withdrawal has been agreed due to an editorial office error that led to the publication of the article without peer review. The revised article, which has undergone peer review may be read here: https://onlinelibrary.wiley.com/doi/10.1002/cam4.6309.
Collapse
|
7
|
Zhu Y, Pu Q, Zhang Q, Liu Y, Ma Y, Yuan Y, Liu L, Zhu W. Selenium-binding protein 1 inhibits malignant progression and induces apoptosis via distinct mechanisms in non-small cell lung cancer. Cancer Med 2023; 12:17149-17170. [PMID: 37606338 PMCID: PMC10501285 DOI: 10.1002/cam4.6309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 05/30/2023] [Accepted: 06/23/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Selenium is an essential trace element in the human body. In epidemiological and clinical studies, Se supplementation significantly reduced the incidence of lung cancer in individuals with low baseline Se levels. The significant action of selenium is based on the selenium-containing protein as a mediator. Of note, the previous studies reported that the expression of selenium-binding protein 1 (SELENBP1) was obviously decreased in many human cancer tissues including non-small cell lung cancer (NSCLC). However, its roles in the origin and development of NSCLC are still unclear. METHODS The expression of SELENBP1 was measured by qRT-PCR, Western blotting and IHC in our collected clinical NSCLC tissues and cell lines. Next, the CCK-8, colony formation, wound-haeling, Millicell, Transwell, FCM assay, and in vivo xenograft model were performed to explore the function of SELENBP1 in NSCLC. The molecular mechanisms of SELENBP1 were investigated by Western blotting or IF assay. RESULTS We further identified that the expression of SELENBP1 was significantly decreased in NSCLC tissues in TCGA database and 45 out of 59 collected clinical NSCLC tissues compared with adjacent nontumor tissues, as well as in four NSCLC cell lines compared with normal lung cells. Particularly, we unexpectedly discovered that SELENBP1 was obviously expressed in alveolar type 2 (AT-II) cells for the first time. Then, a series of in vitro experiments uncovered that overexpression of SELENBP1 inhibited the proliferation, migration, and invasion of NSCLC cells, and induced cell apoptosis. Moreover, overexpression of SELENBP1 also inhibited growth and induced apoptosis of NSCLC cells in vivo. Mechanistically, we demonstrated that overexpression of SELENBP1 inhibited the malignant characteristics of NSCLC cells in part via inactivating the PI3K/AKT/mTOR signal pathway. Meanwhile, we found that overexpression of SELENBP1 inducing the apoptosis of NSCLC cells was associated with the activation of caspase-3 signaling pathway under nonhigh level of oxidative stress, but overexpression of SELENBP1 facilitating the cell apoptosis might be related to its combining with GPX1 and colocalizing in the nucleus under high level of oxidative stress. CONCLUSIONS Our findings highlighted that SELENBP1 was an important tumor suppressor during the origin and development of NSCLC. It may help to discover novel biomarkers or drug therapy targets for NSCLC.
Collapse
Affiliation(s)
- Ying Zhu
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Qiang Pu
- Department of Thoracic SurgeryInstitute of Thoracic Oncology, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Qiongyin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yang Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yongfang Ma
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yue Yuan
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Lunxu Liu
- Department of Thoracic SurgeryInstitute of Thoracic Oncology, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Wen Zhu
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
8
|
Zhang Y, He Q. The role of SELENBP1 and its epigenetic regulation in carcinogenic progression. Front Genet 2022; 13:1027726. [PMID: 36386843 PMCID: PMC9663989 DOI: 10.3389/fgene.2022.1027726] [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: 08/25/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
The initiation and progression of cancer is modulated through diverse genetic and epigenetic modifications. The epigenetic machinery regulates gene expression through intertwined DNA methylation, histone modifications, and miRNAs without affecting their genome sequences. SELENBP1 belongs to selenium-binding proteins and functions as a tumor suppressor. Its expression is significantly downregulated and correlates with carcinogenic progression and poor survival in various cancers. The role of SELENBP1 in carcinogenesis has not been fully elucidated, and its epigenetic regulation remains poorly understood. In this review, we summarize recent findings on the function and regulatory mechanisms of SELENBP1 during carcinogenic progression, with an emphasis on epigenetic mechanisms. We also discuss the potential cancer treatment targeting epigenetic modification of SELENBP1, either alone or in combination with selenium-containing compounds or dietary selenium.
Collapse
|
9
|
Qin B, Zhou L, Wang F, Wang Y. Ubiquitin-specific protease 20 in human disease: emerging role and therapeutic implications. Biochem Pharmacol 2022; 206:115352. [DOI: 10.1016/j.bcp.2022.115352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
|
10
|
Shang P, Zhang B, Li P, Ahmed Z, Hu X, Chamba Y, Zhang H. Plateau Adaptation Gene Analyses Reveal Transcriptomic, Proteomic, and Dual Omics Expression in the Lung Tissues of Tibetan and Yorkshire Pigs. Animals (Basel) 2022; 12:ani12151919. [PMID: 35953907 PMCID: PMC9367445 DOI: 10.3390/ani12151919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/29/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Elevated environments such as plateaus are often classified as low oxygen environments. The hypoxic adaptation mechanisms utilized by organisms in these conditions are not well understood. To address this, the differentially expressed genes (DEGs) involved in hypoxia adaptation were assessed using two pig breeds (Tibetan pig [TP] and Yorkshire sow [YY]). Genes related to lung tissue responses to hypoxia were assessed using transcriptomic (using RNA-seq) and proteomic (using iTRAQ) analysis. A total of 1021 DEGs were screened out. In the iTRAQ omics data, a total of 22,100 peptides were obtained and 4518 proteins were found after filtering. A total of 271 differentially expressed proteins [DEPs] were screened using the conditions of p < 0.05; FC ≤ 0.833; and FC ≥ 1.2. A total of 14 DEGs at the mRNA and protein levels were identified and found to be associated with regulation of the inflammatory response; blood particles; and MAPK cascade response regulation. Among the DEGs, six were associated with hypoxia adaptation function (mitochondria and glycolysis) in pigs. The results of this study identify novel candidate genes involved in porcine hypoxia adaptation mechanisms.
Collapse
Affiliation(s)
- Peng Shang
- Laboratory National Engineering for Animal Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing 100193, China; (P.S.); (B.Z.); (P.L.); (X.H.)
- Department of animal husbandry, College of Animal Science, Tibet Agriculture and Animal Husbandry College, Linzhi 860000, China
| | - Bo Zhang
- Laboratory National Engineering for Animal Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing 100193, China; (P.S.); (B.Z.); (P.L.); (X.H.)
| | - Pan Li
- Laboratory National Engineering for Animal Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing 100193, China; (P.S.); (B.Z.); (P.L.); (X.H.)
| | - Zulfiqar Ahmed
- Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Rawalakot 12350, Pakistan;
| | - Xiaoxiang Hu
- Laboratory National Engineering for Animal Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing 100193, China; (P.S.); (B.Z.); (P.L.); (X.H.)
| | - Yangzom Chamba
- Department of animal husbandry, College of Animal Science, Tibet Agriculture and Animal Husbandry College, Linzhi 860000, China
- Correspondence: (Y.C.); (H.Z.)
| | - Hao Zhang
- Laboratory National Engineering for Animal Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing 100193, China; (P.S.); (B.Z.); (P.L.); (X.H.)
- Correspondence: (Y.C.); (H.Z.)
| |
Collapse
|
11
|
Seelig J, Heller RA, Haubruck P, Sun Q, Georg Klingenberg J, Hackler J, Crowell HL, Daniel V, Moghaddam A, Schomburg L, Biglari B. Selenium-Binding Protein 1 (SELENBP1) as Biomarker for Adverse Clinical Outcome After Traumatic Spinal Cord Injury. Front Neurosci 2021; 15:680240. [PMID: 34140879 PMCID: PMC8204909 DOI: 10.3389/fnins.2021.680240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 04/29/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Traumatic spinal cord injury (TSCI) presents a diagnostic challenge as it may have dramatic consequences for the affected patient. Additional biomarkers are needed for improved care and personalized therapy. Objective: Serum selenium binding protein 1 (SELENBP1) has been detected in myocardial infarction, reflecting hypoxic tissue damage and recovery odds. As SELENBP1 is usually not detected in the serum of healthy subjects, we tested the hypothesis that it may become detectable in TSCI and indicate tissue damage and regeneration odds. Methods: In this prospective observational study, patients with comparable injuries were allocated to three groups; vertebral body fractures without neurological impairment (control “C”), TSCI without remission (“G0”), and TSCI with signs of remission (“G1”). Consecutive serum samples were available from different time points and analyzed for SELENBP1 by sandwich immunoassay, for trace elements by X-ray fluorescence and for cytokines by multiplex immunoassays. Results: Serum SELENBP1 was elevated at admission in relation to the degree of neurological impairment [graded as A, B, C, or D according to the American Spinal Injury Association (AISA) impairment scale (AIS)]. Patients with the most severe neurological impairment (classified as AIS A) exhibited the highest SELENBP1 concentrations (p = 0.011). During the first 3 days, SELENBP1 levels differed between G0 and G1 (p = 0.019), and dynamics of SELENBP1 correlated to monocyte chemoattractant protein 1, chemokine ligand 3 and zinc concentrations. Conclusion: Circulating SELENBP1 concentrations are related to the degree of neurological impairment in TSCI and provide remission odds information. The tight correlation of SELENBP1 with CCL2 levels provides a novel link between Se metabolism and immune cell activation, with potential relevance for neurological damage and regeneration processes, respectively.
Collapse
Affiliation(s)
- Julian Seelig
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Raban Arved Heller
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Heidelberg Trauma Research Group, Department of Trauma and Reconstructive Surgery, Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Heidelberg, Germany.,Department of General Practice and Health Services Research, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Haubruck
- Heidelberg Trauma Research Group, Department of Trauma and Reconstructive Surgery, Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Heidelberg, Germany.,Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Institute of Bone and Joint Research, University of Sydney, St Leonards, NSW, Australia
| | - Qian Sun
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Jochen Georg Klingenberg
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Julian Hackler
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Helena Lucia Crowell
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland.,Systems Biology Ph.D. Program, Life Science Zurich Graduate School, ETH Zürich and University of Zurich, Zurich, Switzerland
| | - Volker Daniel
- Transplantation Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Arash Moghaddam
- Aschaffenburg Trauma and Orthopaedic Research Group, Centre for Orthopaedics, Trauma Surgery and Sports Medicine, Hospital Aschaffenburg-Alzenau, Aschaffenburg, Germany
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Bahram Biglari
- Department of Paraplegiology, BG Trauma Centre Ludwigshafen, Ludwigshafen, Germany
| |
Collapse
|
12
|
Zhu W, Liu Y, Zhang W, Fan W, Wang S, Gu JH, Sun H, Liu F. Selenomethionine protects hematopoietic stem/progenitor cells against cobalt nanoparticles by stimulating antioxidant actions and DNA repair functions. Aging (Albany NY) 2021; 13:11705-11726. [PMID: 33875618 PMCID: PMC8109066 DOI: 10.18632/aging.202865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/22/2021] [Indexed: 01/13/2023]
Abstract
Hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) can differentiate into all blood lineages to maintain hematopoiesis, wound healing, and immune functions. Recently, cobalt-chromium alloy casting implants have been used extensively in total hip replacements; however, cobalt nanoparticles (CoNPs) released from the alloy were toxic to HSCs and HPCs. We aimed to investigate the mechanism underlying the toxic effect of CoNPs on HSCs/HPCs and to determine the protective effect of selenomethionine (SeMet) against CoNPs in vitro and in vivo. Human and rat CD34+ HSCs/HPCs were isolated from cord blood and bone marrow, respectively. CoNPs decreased the viability of CD34+ HSCs/HPCs and increased apoptosis. SeMet attenuated the toxicity of CoNPs by enhancing the antioxidant ability of cells. The protective effect of SeMet was not completely abolished after adding H2O2 to abrogate the improvement of the antioxidant capacity by SeMet. SeMet and CoNPs stimulated ATM/ATR DNA damage response signals and inhibited cell proliferation. Unlike CoNPs, SeMet did not damage the DNA, and cell proliferation recovered after removing SeMet. SeMet inhibited the CoNP-induced upregulation of hypoxia inducible factor (HIF)-1α, thereby disrupting the inhibitory effect of HIF-1α on breast cancer type 1 susceptibility protein (BRCA1). Moreover, SeMet promoted BRCA1-mediated ubiquitination of cyclin B by upregulating UBE2K. Thus, SeMet enhanced cell cycle arrest and DNA repair post-CoNP exposure. Overall, SeMet protected CD34+ HSCs/HPCs against CoNPs by stimulating antioxidant activity and DNA repair.
Collapse
Affiliation(s)
- Wenfeng Zhu
- Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.,Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Yake Liu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Weinan Zhang
- Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Wentao Fan
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Siqi Wang
- Orthopaedic Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province, China.,Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Huanjian Sun
- Department of Orthopaedics, The Sixth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, China
| | - Fan Liu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
13
|
Martins Alves AM, Pereira Menezes Reis S, Peres Gramacho K, Micheli F. The glutathione peroxidase family of Theobroma cacao: Involvement in the oxidative stress during witches' broom disease. Int J Biol Macromol 2020; 164:3698-3708. [PMID: 32882281 DOI: 10.1016/j.ijbiomac.2020.08.222] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/26/2020] [Accepted: 08/28/2020] [Indexed: 11/27/2022]
Abstract
The glutathione peroxidases (GPXs) are enzymes which are part of the cell antioxidant system inhibiting the ROS-induced damages of membranes and proteins. In cacao (Theobroma cacao L.) genome, five GPX genes were identified. Cysteine insertion codons (UGU) were found in TcPHGPX, TcGPX2, TcGPX4, TcGPX6 and tryptophan insertion codon (UGG) in TcGPX8. Multiple alignments revealed conserved domains between TcGPXs and other plants and human GPXs. Homology modeling was performed using the Populus trichocarpa GPX5 structure as template, and the molecular modeling showed that TcGPXs have affinity with selenometionine in their active site. In silico analysis of the TcGPXs promoter region revealed the presence of conserved cis-elements related to biotic stresses and hormone responsiveness. The expression analysis of TcGPXs in cacao plantlet meristems infected by M. perniciosa showed that TcGPXs are most expressed in susceptible variety than in resistant one, mainly in disease stages in which oxidative stress and programmed cell death occurred. This data, associated with phylogenetic and location analysis suggested that TcGPXs may play a role in protecting cells from oxidative stress as a try of disease progression reduction. To our knowledge, this is the first study of the overall GPX family from T. cacao.
Collapse
Affiliation(s)
- Akyla Maria Martins Alves
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Rodovia Ilhéus-Itabuna, km 16, 45662-900 Ilhéus, BA, Brazil
| | - Sara Pereira Menezes Reis
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Rodovia Ilhéus-Itabuna, km 16, 45662-900 Ilhéus, BA, Brazil
| | | | - Fabienne Micheli
- Universidade Estadual de Santa Cruz (UESC), Departamento de Ciências Biológicas (DCB), Centro de Biotecnologia e Genética (CBG), Rodovia Ilhéus-Itabuna, km 16, 45662-900 Ilhéus, BA, Brazil; CIRAD, UMR AGAP, F-34398 Montpellier, France.
| |
Collapse
|
14
|
Lee YM, Kim S, Park RY, Kim YS. Hepatitis B Virus-X Downregulates Expression of Selenium Binding Protein 1. Viruses 2020; 12:v12050565. [PMID: 32443734 PMCID: PMC7291177 DOI: 10.3390/v12050565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
Selenium binding protein 1 (SELENBP1) has been known to be reduced in various types cancer, and epigenetic change is shown to be likely to account for the reduction of SELNEBP1 expression. With cDNA microarray comparative analysis, we found that SELENBP1 is markedly decreased in hepatitis B virus-X (HBx)-expressing cells. To clarify the effect of HBx on SELENBP1 expression, we compared the expression levels of SELENBP1 mRNA and protein by semi-quantitative RT-PCR, Northern blot, and Western blot. As expected, SELENBP1 expression was shown to be reduced in cells expressing HBx, and reporter gene analysis showed that the SELENBP1 promoter is repressed by HBx. In addition, the stepwise deletion of 5′ flanking promoter sequences resulted in a gradual decrease in basal promoter activity and inhibition of SELENBP1 expression by HBx. Moreover, immunohistochemistry on tissue microarrays containing 60 pairs of human liver tissue showed decreased intensity of SELENBP1 in tumor tissues as compared with their matched non-tumor liver tissues. Taken together, our findings suggest that inhibition of SELENBP1 expression by HBx might act as one of the causes in the development of hepatocellular carcinoma caused by HBV infection.
Collapse
Affiliation(s)
- Young-Man Lee
- Dasan Undergraduate College, Ajou University, Suwon 16499, Korea;
| | - Soojin Kim
- Graduate School of New Drug Discovery & Development, Chungnam National University, Daejeon 34134, Korea;
| | - Ran-Young Park
- Department of Smart Food & Drugs, Inje University, Gimhae 50834, Korea;
| | - Yeon-Soo Kim
- Graduate School of New Drug Discovery & Development, Chungnam National University, Daejeon 34134, Korea;
- Correspondence: ; Tel.: +82-42-821-8631
| |
Collapse
|
15
|
Wang Y, Zhu W, Chen X, Wei G, Jiang G, Zhang G. Selenium-binding protein 1 transcriptionally activates p21 expression via p53-independent mechanism and its frequent reduction associates with poor prognosis in bladder cancer. J Transl Med 2020; 18:17. [PMID: 31918717 PMCID: PMC6953137 DOI: 10.1186/s12967-020-02211-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 01/03/2020] [Indexed: 01/31/2023] Open
Abstract
Background Recent studies have shown that selenium-binding protein 1 (SELENBP1) is significantly down-regulated in a variety of solid tumors. Nevertheless, the clinical relevance of SELENBP1 in human bladder cancer has not been described in any detail, and the molecular mechanism underlying its inhibitory role in cancer cell growth is largely unknown. Methods SELENBP1 expression levels in tumor tissues and adjacent normal tissues were evaluated using immunoblotting assay. The association of SELENBP1 expression, clinicopathological features, and clinical outcome was determined using publicly available dataset from The Cancer Genome Atlas bladder cancer (TCGA-BLCA) cohort. DNA methylation in SELENBP1 gene was assessed using online MEXPRESS tool. We generated stable SELENBP1-overexpression and their corresponding control cell lines to determine its potential effect on cell cycle and transcriptional activity of p21 by using flow cytometry and luciferase reporter assay, respectively. The dominant-negative mutant constructs, TAM67 and STAT1 Y701F, were employed to define the roles of c-Jun and STAT1 in the regulation of p21 protein. Results Here, we report that the reduction of SELENBP1 is a frequent event and significantly correlates with tumor progression as well as unfavorable prognosis in human bladder cancer. By utilizing TCGA-BLCA cohort, DNA hypermethylation, especially in gene body, is shown to be likely to account for the reduction of SELENBP1 expression. However, an apparent paradox is observed in its 3′-UTR region, in which DNA methylation is positively related to SELENBP1 expression. More importantly, we verify the growth inhibitory role for SELENBP1 in human bladder cancer, and further report a novel function for SELENBP1 in transcriptionally modulating p21 expression through a p53-independent mechanism. Instead, ectopic expression of SELENBP1 pronouncedly attenuates the phosphorylation of c-Jun and STAT1, both of which are indispensable for SELENBP1-mediated transcriptional induction of p21, thereby resulting in the G0/G1 phase cell cycle arrest in bladder cancer cell. Conclusions Taken together, our findings provide clinical and molecular insights into improved understanding of the tumor suppressive role for SELENBP1 in human bladder cancer, suggesting that SELENBP1 could potentially be utilized as a prognostic biomarker as well as a therapeutic target in future cancer therapy.
Collapse
Affiliation(s)
- Yulei Wang
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China. .,School of Medicine, South China University of Technology, Guangzhou, 510641, China.
| | - Wenzhen Zhu
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Xiaoqing Chen
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Guangnan Wei
- School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guochun Zhang
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China. .,School of Medicine, South China University of Technology, Guangzhou, 510641, China.
| |
Collapse
|
16
|
Selenium-Binding Protein 1 in Human Health and Disease. Int J Mol Sci 2018; 19:ijms19113437. [PMID: 30400135 PMCID: PMC6274749 DOI: 10.3390/ijms19113437] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/05/2018] [Accepted: 10/31/2018] [Indexed: 12/19/2022] Open
Abstract
Selenium-binding protein 1 (SBP1) is a highly conserved protein that covalently binds selenium. SBP1 may play important roles in several fundamental physiological functions, including protein degradation, intra-Golgi transport, cell differentiation, cellular motility, redox modulation, and the metabolism of sulfur-containing molecules. SBP1 expression is often reduced in many cancer types compared to the corresponding normal tissues and low levels of SBP1 are frequently associated with poor clinical outcome. In this review, the transcriptional regulation of SBP1, the different physiological roles reported for SBP1, as well as the implications of SBP1 function in cancer and other diseases are presented.
Collapse
|
17
|
Caswell DR, Chuang CH, Ma RK, Winters IP, Snyder EL, Winslow MM. Tumor Suppressor Activity of Selenbp1, a Direct Nkx2-1 Target, in Lung Adenocarcinoma. Mol Cancer Res 2018; 16:1737-1749. [PMID: 30002193 DOI: 10.1158/1541-7786.mcr-18-0392] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/07/2018] [Accepted: 06/29/2018] [Indexed: 12/18/2022]
Abstract
The Nkx2-1 transcription factor promotes differentiation of lung epithelial lineages and suppresses malignant progression of lung adenocarcinoma. However, targets of Nkx2-1 that limit tumor growth and progression remain incompletely understood. Here, direct Nkx2-1 targets are identified whose expression correlates with Nkx2-1 activity in human lung adenocarcinoma. Selenium-binding protein 1 (Selenbp1), an Nkx2-1 effector that limits phenotypes associated with lung cancer growth and metastasis, was investigated further. Loss- and gain-of-function approaches demonstrate that Nkx2-1 is required and sufficient for Selenbp1 expression in lung adenocarcinoma cells. Interestingly, Selenbp1 knockdown also reduced Nkx2-1 expression and Selenbp1 stabilized Nkx2-1 protein levels in a heterologous system, suggesting that these genes function in a positive feedback loop. Selenbp1 inhibits clonal growth and migration and suppresses growth of metastases in an in vivo transplant model. Genetic inactivation of Selenbp1, using CRISPR/Cas9, also enhanced primary tumor growth in autochthonous lung adenocarcinoma mouse models. Collectively, these data demonstrate that Selenbp1 is a direct target of Nkx2-1, which inhibits lung adenocarcinoma growth in vivo Implications: Selenbp1 is an important suppressor of lung tumor growth that functions in a positive feedback loop with Nkx2-1, and whose loss is associated with worse patient outcome. Mol Cancer Res; 16(11); 1737-49. ©2018 AACR.
Collapse
Affiliation(s)
- Deborah R Caswell
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California
| | - Chen-Hua Chuang
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Rosanna K Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Ian P Winters
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Eric L Snyder
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Monte M Winslow
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California. .,Department of Genetics, Stanford University School of Medicine, Stanford, California.,Department of Pathology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
18
|
Valassakis C, Livanos P, Minopetrou M, Haralampidis K, Roussis A. Promoter analysis and functional implications of the selenium binding protein (SBP) gene family in Arabidopsis thaliana. JOURNAL OF PLANT PHYSIOLOGY 2018; 224-225:19-29. [PMID: 29574326 DOI: 10.1016/j.jplph.2018.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 05/23/2023]
Abstract
Selenium Βinding Protein (SBP, originally termed SBP56) was identified in mouse liver as a cytosolic protein that could bind radioactive selenium. SBPs are highly conserved proteins present in a wide array of species across all kingdoms and are likely to be involved in selenium metabolism. In Arabidopsis, the selenium binding protein (SBP) gene family comprises three genes (AtSBP1, AtSBP2 and AtSBP3). AtSBP1 and AtSBP2 are clustered in a head-to-tail arrangement on chromosome IV, while AtSBP3 is located on chromosome III. In this work, we studied the promoter activity of the Arabidopsis SBP genes, determined their tissue specificity and showed that they are differentially regulated by sodium selenite and sodium selenate. All three SBP genes are upregulated in response to externally applied selenium compounds and the antioxidant NAC selectively downregulates SBP2. Although the effect on SBP2 levels was the most prominent, in all cases, the concurrent exposure of plants to selenite and the antioxidant supressed the expression of the SBP genes. We provide evidence that (at least) SBP1 expression is tightly linked to detoxification processes related to oxidative stress, since it is downregulated in the presence of NAC in selenium-treated plants. Furthermore, our results suggest that SBP genes may participate in the mechanisms that sense redox imbalance.
Collapse
Affiliation(s)
- Chrysanthi Valassakis
- National and Kapodistrian University of Athens, Faculty of Biology, Department of Botany, 15784 Athens, Greece
| | - Pantelis Livanos
- National and Kapodistrian University of Athens, Faculty of Biology, Department of Botany, 15784 Athens, Greece
| | - Martha Minopetrou
- National and Kapodistrian University of Athens, Faculty of Biology, Department of Botany, 15784 Athens, Greece
| | - Kosmas Haralampidis
- National and Kapodistrian University of Athens, Faculty of Biology, Department of Botany, 15784 Athens, Greece
| | - Andreas Roussis
- National and Kapodistrian University of Athens, Faculty of Biology, Department of Botany, 15784 Athens, Greece.
| |
Collapse
|
19
|
Ji Z, Liu H, Fang L, Yu Y, Zhou Z. Use of immunoproteomics to identify immunogenic proteins in a rat model of acute respiratory distress syndrome. Mol Med Rep 2017; 16:7625-7632. [PMID: 28944852 DOI: 10.3892/mmr.2017.7557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/17/2017] [Indexed: 11/06/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and life‑threatening clinical syndrome, and seeking biomarkers of ARDS has been an area of continuing research. The present study hypothesized that alterations to certain immunogenic substances occur in injured lungs and are able to specifically bind with corresponding proteins in the blood, and that these proteins may be readily detected. To investigate this hypothesis, a rat model of ARDS was established by cecal ligation and puncture surgery, and an immunoproteomics approach, using serum as the primary antibody in a western blot analysis, was used with the aim of identifying immunogenic proteins in the injured lungs. Ingenuity Pathway Analysis (IPA) was used for bioinformatics analysis, and mass spectrometric analysis was used to identify a total of 38 differentially expressed immunogenic proteins. Bioinformatics analysis revealed that the top canonical pathways in which the identified proteins may be involved were gluconeogenesis I, glycolysis I, choline degradation I, NADH repair and heme degradation. IPA Biomarker Filter analysis with the terms 'acute respiratory distress syndrome/acute lung injury' was used to screen 13 proteins as candidate biomarkers. These proteins were described as antigens, and suggested that paired antibodies may be detected in the plasma of patients at high risk of ARDS. Analysis of these identified proteins may provide novel insights into the potential pathological mechanisms of ARDS.
Collapse
Affiliation(s)
- Zongshu Ji
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Haiyan Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Linsen Fang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Youxin Yu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Zheng Zhou
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
20
|
Maina PK, Shao P, Jia X, Liu Q, Umesalma S, Marin M, Long D, Concepción-Román S, Qi HH. Histone demethylase PHF8 regulates hypoxia signaling through HIF1α and H3K4me3. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:1002-1012. [PMID: 28734980 PMCID: PMC5776039 DOI: 10.1016/j.bbagrm.2017.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/10/2017] [Accepted: 07/18/2017] [Indexed: 11/12/2022]
Abstract
Hypoxia through transcription factor HIF1α plays a critical role in cancer development. In prostate cancer, HIF1α interplays with androgen receptor (AR) to contribute to the progression of this disease to its lethal form—castration-resistant prostate cancer (CRPC). Hypoxia upregulates several epigenetic factors including histone demethylase KDM3A which is a critical co-factor of HIF1α. However, how histone demethylases regulate hypoxia signaling is not fully understood. Here, we report that histone demethylase PHF8 plays an essential role in hypoxia signaling. Knockdown or knockout of PHF8 by RNAi or CRISPR-Cas9 system reduced the activation of HIF1α and the induction of HIF1α target genes including KDM3A. Mechanistically, PHF8 regulates hypoxia inducible genes mainly through sustaining the level of trimethylated histone 3 lysine 4 (H3K4me3), an active mark in transcriptional regulation. The positive role of PHF8 in hypoxia signaling extended to hypoxia-induced neuroendocrine differentiation (NED), wherein PHF8 cooperates with KDM3A to regulate the expression of NED genes. Moreover, we discovered that the role of PHF8 in hypoxia signaling is associated with the presence of full-length AR in CRPC cells. Collectively, our study identified PHF8 as a novel epigenetic factor in hypoxia signaling, and the underlying regulatory mechanisms likely apply to general cancer development involving HIF1α. Therefore, targeting PHF8 can potentially be a novel therapeutic strategy in cancer therapy.
Collapse
Affiliation(s)
- Peterson Kariuki Maina
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Peng Shao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Xiongfei Jia
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Qi Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Shaikamjad Umesalma
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Maximo Marin
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Donald Long
- Department of Biology, Southern Utah University, Cedar City, UT 84720, USA
| | | | - Hank Heng Qi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA.
| |
Collapse
|
21
|
SELENBP1 expression in the prefrontal cortex of subjects with schizophrenia. Transl Psychiatry 2015; 5:e615. [PMID: 26241353 PMCID: PMC4564563 DOI: 10.1038/tp.2015.108] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 06/10/2015] [Accepted: 06/14/2015] [Indexed: 12/30/2022] Open
Abstract
Selenium binding protein 1 (SELENBP1) messenger RNA (mRNA) has previously been shown to be upregulated in the brain and blood from subjects with schizophrenia. We aimed to validate these findings in a new cohort using real-time PCR in Brodmann's Area (BA) 9, and to determine the disease specificity of increased SELENBP1 expression by measuring SELENBP1 mRNA in subjects with major depressive disorder and bipolar disorder. We then extended the study to include other cortical regions such as BA8 and BA44. SELENBP1 mRNA was higher in BA9 (P = 0.001), BA8 (P = 0.003) and BA44 (P = 0.0007) from subjects with schizophrenia. Conversely, in affective disorders, there was no significant difference in SELENBP1 mRNA in BA9 (P = 0.67), suggesting that the upregulation may be diagnosis specific. Measurement of SELENBP1 protein levels showed that changes in mRNA did not translate to changes in protein. In addition, chronic treatment of rats with antipsychotics did not significantly affect the expression of Selenbp1 in the cortex (P = 0.24). Our data show that elevated SELENBP1 transcript expression is widespread throughout the prefrontal cortex in schizophrenia, and confirm that this change is a consistent feature of schizophrenia and not a simple drug effect.
Collapse
|
22
|
The subcellular location of selenoproteins and the impact on their function. Nutrients 2015; 7:3938-48. [PMID: 26007340 PMCID: PMC4446787 DOI: 10.3390/nu7053938] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 01/31/2023] Open
Abstract
Most human selenium containing proteins contain selenium in the form of the amino acid selenocysteine, which is encoded in the corresponding mRNA as a UGA codon. Only a few non-selenocysteine containing selenoproteins are present and the nature of the association with selenium is not well understood. This review focuses on two selenocysteine-containing proteins that are members of the glutathione peroxidase family, GPx-1 and GPx-4, and the selenium-associated protein referred to as Selenium Binding Protein 1. Each of these proteins have been described to reside in two or more cellular compartments, and in the case of GPx-1 and SBP1, interact with each other. The enzymatic activity of GPx-1 and GPx-4 have been well described, but it is less clear how their cellular location impacts the health related phenotypes associated with activities, while no catalytic function is assigned to SBP1. The distribution of these proteins is presented as is the possible consequences of that compartmentalization.
Collapse
|
23
|
Ansong E, Ying Q, Ekoue DN, Deaton R, Hall AR, Kajdacsy-Balla A, Yang W, Gann PH, Diamond AM. Evidence that selenium binding protein 1 is a tumor suppressor in prostate cancer. PLoS One 2015; 10:e0127295. [PMID: 25993660 PMCID: PMC4436248 DOI: 10.1371/journal.pone.0127295] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 04/13/2015] [Indexed: 12/25/2022] Open
Abstract
Selenium-Binding Protein 1 (SBP1, SELENBP1, hSP56) is a selenium-associated protein shown to be at lower levels in tumors, and its lower levels are frequently predictive of a poor clinical outcome. Distinguishing indolent from aggressive prostate cancer is a major challenge in disease management. Associations between SBP1 levels, tumor grade, and disease recurrence following prostatectomy were investigated by duplex immunofluorescence imaging using a tissue microarray containing tissue from 202 prostate cancer patients who experienced biochemical (PSA) recurrence after prostatectomy and 202 matched control patients whose cancer did not recur. Samples were matched by age, ethnicity, pathological stage and Gleason grade, and images were quantified using the Vectra multispectral imaging system. Fluorescent labels were targeted for SBP1 and cytokeratins 8/18 to restrict scoring to tumor cells, and cell-by-cell quantification of SBP1 in the nucleus and cytoplasm was performed. Nuclear SBP1 levels and the nuclear to cytoplasm ratio were inversely associated with tumor grade using linear regression analysis. Following classification of samples into quartiles based on the SBP1 levels among controls, tumors in the lowest quartile were more than twice as likely to recur compared to those in any other quartile. Inducible ectopic SBP1 expression reduced the ability of HCT-116 human tumor cells to grow in soft agar, a measure of transformation, without affecting proliferation. Cells expressing SBP1 also demonstrated a robust induction in the phosphorylation of the p53 tumor suppressor at serine 15. These data indicate that loss of SBP1 may play an independent contributing role in prostate cancer progression and its levels might be useful in distinguishing indolent from aggressive disease.
Collapse
Affiliation(s)
- Emmanuel Ansong
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Qi Ying
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Dede N. Ekoue
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ryan Deaton
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Andrew R. Hall
- University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Andre Kajdacsy-Balla
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Wancai Yang
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Peter H. Gann
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Alan M. Diamond
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
24
|
Wang Y, Fang W, Huang Y, Hu F, Ying Q, Yang W, Xiong B. Reduction of selenium-binding protein 1 sensitizes cancer cells to selenite via elevating extracellular glutathione: a novel mechanism of cancer-specific cytotoxicity of selenite. Free Radic Biol Med 2015; 79:186-96. [PMID: 25445402 DOI: 10.1016/j.freeradbiomed.2014.11.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/14/2014] [Accepted: 11/20/2014] [Indexed: 01/14/2023]
Abstract
Selenium is an essential trace element and has been extensively studied for preventive effects on cancers. Recent emerging evidence has also shown that selenium at supranutritional dosage has a preferential cytotoxicity in cancer cells and chemotherapeutic drug-resistant cells, but the underlying mechanisms remain largely unknown. This study was to investigate the roles of two distinct representatives of selenium-containing proteins, selenium-binding protein 1 (SBP1) and glutathione peroxidase 1 (GPX1), in selenite-mediated cancer-specific cytotoxicity. We found that there was a significantly inverse correlation between SBP1 and GPX1 protein level in human breast cancers and adjacent matched nontumor tissues (Pearson r=-0.4347, P=0.0338). Ectopic expression of GPX1 enhanced selenite cytotoxicity through down-regulation of SBP1, and SBP1 was likely to be a crucial determinant for selenite-mediated cytotoxicity. Reduction of SBP1 in cancer cells and epirubicin-resistant cells on selenite exposure resulted in a dramatic increase in the generation of hydrogen peroxide and superoxide anion, which in turn caused oxidative stress and triggered apoptosis. Furthermore, knockdown SBP1 by small interfering RNA increased selenite sensitivity by elevating extracellular glutathione (GSH), which spontaneously reacted with selenite and led to the rapid depletion of selenium (IV) in growth medium and the high-affinity uptake of selenite. In conclusion, these findings would improve our understanding of the roles of selenium-containing proteins in selenite-mediated cytotoxicity, and revealed a potent mechanism of the selective cytotoxicity of selenite in cancer cells and drug-resistant cells, in which SBP1 was likely to play an important role in modulating the extracellular microenvironment by regulating the levels of extracellular GSH.
Collapse
Affiliation(s)
- Yulei Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, 430071, China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Ying Huang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Department of Oncology, the Fifth Hospital, Wuhan, Hubei, 430051, China
| | - Fen Hu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Qi Ying
- Department of Pathology, University of Illinois at Chicago, IL 60612, USA
| | - Wancai Yang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Department of Pathology, University of Illinois at Chicago, IL 60612, USA.
| | - Bin Xiong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, 430071, China.
| |
Collapse
|