1
|
Wu H, Shi JY, Zhao YS, Shi YW, Zhong LQY, Feng HN, Liu Y, Chen JH, Chen X. Low Expression of Selenoprotein S Promotes Osteogenic Differentiation in Bone Marrow Mesenchymal Stromal Cells. Biol Trace Elem Res 2025:10.1007/s12011-025-04657-3. [PMID: 40342061 DOI: 10.1007/s12011-025-04657-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/03/2025] [Indexed: 05/11/2025]
Abstract
Selenium performs biological functions in the human body primarily through selenoproteins, which are known to play critical roles in bone metabolism. Normal osteogenic function is maintained by steady bone metabolism in the mandible. The role of selenoprotein S (SelS), one of the 25 identified selenoproteins, in osteogenic differentiation remains unclear. This study aimed to investigate whether SelS is involved in osteogenic differentiation and to analyze the effect of low expression of SelS on osteogenic differentiation. Osteogenic differentiation was induced in vitro, and the expression dynamics of SelS, Sp7 transcription factor (Sp7), and alkaline phosphatase (Alp) were analyzed using quantitative PCR (qPCR) and Western blotting. Lentivirus-mediated RNA interference (RNAi) was employed to achieve stable SelS knockdown. Alp activity was evaluated after osteogenic differentiation. Mandibular bone formation was evaluated by micro-CT scanning and histological staining (Masson's trichrome staining). Transcriptome sequencing and bioinformatics analysis were conducted to identify genes and pathways regulated by SelS low expression. During osteogenic differentiation, the expression of SelS protein decreased. After low expression of SelS, osteogenic differentiation was performed and the expression of Alp and Sp7 increased. Alp staining showed that the number of mineralized particles in the low expression SelS group increased, indicating that low expression of SelS enhanced the osteogenic differentiation ability of cells. Micro-CT and histological staining showed that the mandibular collagen synthesis and bone formation in the low expression SelS group were higher than those in the control group. Transcriptomic profiling identified 2268 differentially expressed genes (DEGs) under SelS low expression, suggesting that SelS may play a regulatory role by affecting the expression of these genes. Our data suggest that SelS expression is related to osteogenic differentiation. At low levels, SelS can promote osteogenic differentiation, collagen synthesis, and enhance bone formation.
Collapse
Affiliation(s)
- Hao Wu
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an JiaoTong University, 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Jing-Yi Shi
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an JiaoTong University, 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yun-Shan Zhao
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an JiaoTong University, 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Ya-Wen Shi
- School of Public Health, Xi'an JiaoTong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Liang-Qiu-Yue Zhong
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an JiaoTong University, 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Hai-Nan Feng
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an JiaoTong University, 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Yan Liu
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an JiaoTong University, 277 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Jing-Hong Chen
- School of Public Health, Xi'an JiaoTong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, No. 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| | - Xi Chen
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an JiaoTong University, 277 Yanta West Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
2
|
Ozhava D, Lee K, Bektas C, Jackson A, Patel K, Mao Y. Optimized Adipogenic Differentiation and Delivery of Bovine Umbilical Cord Stem Cells for Cultivated Meat. Gels 2024; 10:488. [PMID: 39195017 DOI: 10.3390/gels10080488] [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: 06/26/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/29/2024] Open
Abstract
Cultivated meat, also known as cell-based or clean meat, utilizes mesenchymal stem cells to cultivate mature cell types like adipocytes, which are pivotal for imparting the desired taste and texture. The delivery of differentiated cells, crucial in cultivated meat production, is facilitated through extensive exploration of 3D culturing techniques mimicking physiological environments. In this study, we investigated the adipogenic differentiation potential of bovine umbilical cord stem cells (BUSCs), sourced from discarded birth tissue, and assessed the feasibility of delivering differentiated cells for cultivated meat using gelatin methacrylate (GelMA) as a carrier for adipose tissue. Various adipogenic inducers, previously reported to be effective for human mesenchymal stem cells (hMSCs), were evaluated individually or in combination for their efficacy in promoting the adipogenesis of BUSCs. Surprisingly, while the traditional adipogenic inducers, including insulin, dexamethasone, isobutylmethylxantine (IBMX), indomethacin, and rosiglitazone, showed no significant effect on the adipogenic differentiation of BUSCs, efficient differentiation was achieved in the presence of a fatty acid cocktail. Furthermore, we explored methods for the delivery of BUSCs. Differentiated cells were delivered either encapsulated in GelMA hydrogel or populated on the surface of GelMA microparticles (MPs) as the adipose component of cultivated meat. Our findings reveal that after adipogenic induction, the lipid production per cell was comparable when cultured either within hydrogel or on MPs. However, GelMA-MPs supported better cell growth compared to hydrogel encapsulation. Consequently, the overall lipid production is higher when BUSCs are delivered via GelMA-MPs rather than encapsulation. This study not only systematically evaluated the impact of common adipogenic inducers on BUSCs, but also identified GelMA-MPs as a promising carrier for delivering bovine adipocytes for cultivated meat production.
Collapse
Affiliation(s)
- Derya Ozhava
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Kathleen Lee
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Cemile Bektas
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Anisha Jackson
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Krishi Patel
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
3
|
Xu D, Yuan L, Che M, Lu D, Liu W, Meng F, Yang Y, Du Y, Hou S, Nan Y. Molecular mechanism of Gan-song Yin inhibiting the proliferation of renal tubular epithelial cells by regulating miR-21-5p in adipocyte exosomes. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117530. [PMID: 38043753 DOI: 10.1016/j.jep.2023.117530] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gan-song Yin is derived from the classic ancient prescription " Gan-song pill " for the treatment of wasting-thirst in Ningxia combined with the characteristic "fragrant medicine". It is clinically used for the treatment of early renal fibrosis caused by diabetic nephropathy. Previous studies have shown that it has a good effect and great potential in the prevention and treatment of diabetic nephropathy, but its mechanism research is still limited. AIM OF THE STUDY To investigate the mechanism of GSY to improve DN by interfering with miR-21-5p and glycolipid metabolism in adipocyte exosomes using 3T3-L1 and TCMK-1 co-culture system. MATERIALS AND METHODS The co-culture system of 3T3-L3 and TCMK-1 was established, the IR model was established, and the stability, lipid drop change, glucose consumption, triglyceride content, cell viability, cell cycle and apoptosis level, protein content and mRNA expression of the IR model were detected. RESULTS GSY inhibited 3T3-L1 activity, increased glucose consumption and decreased TG content. Decreased TCMK-1 cell viability, inhibited apoptosis, cell cycle arrest occurred in G0/G1 phase and S phase. Adipocyte IR model and co-culture system were stable within 48 h. After GSY intervention, lipid droplet decomposition and glucose consumption increased. The TG content of adipocytes increased, while the TG content of co-culture system decreased. GSY can regulate the expression of TGF-β1/SMAD signaling pathway protein in IR state. After GSY intervention, the expression of miR-21-5p was increased in 3T3-L1 and Exo cells, and decreased in TCMK-1 cells. CONCLUSIONS GSY can regulate TGF-β1/SMAD signaling pathway through the secretion of miR-21-5p from adipocytes, protect IR TCMK-1, regulate the protein and mRNA expression levels of PPARγ, GLUT4, FABP4, and improve glucose and lipid metabolism.
Collapse
Affiliation(s)
- Duojie Xu
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Mengying Che
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Wenjing Liu
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Fandi Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yating Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Shaozhang Hou
- Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yi Nan
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
4
|
Yu SS, Du JL. Current views on selenoprotein S in the pathophysiological processes of diabetes-induced atherosclerosis: potential therapeutics and underlying biomarkers. Diabetol Metab Syndr 2024; 16:5. [PMID: 38172976 PMCID: PMC10763436 DOI: 10.1186/s13098-023-01247-y] [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: 08/02/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) consistently ranks as the primary mortality factor among diabetic people. A thorough comprehension of the pathophysiological routes and processes activated by atherosclerosis (AS) caused by diabetes mellitus (DM), together with the recognition of new contributing factors, could lead to the discovery of crucial biomarkers and the development of innovative drugs against atherosclerosis. Selenoprotein S (SELENOS) has been implicated in the pathology and progression of numerous conditions, including diabetes, dyslipidemia, obesity, and insulin resistance (IR)-all recognized contributors to endothelial dysfunction (ED), a precursor event to diabetes-induced AS. Hepatic-specific deletion of SELENOS accelerated the onset and progression of obesity, impaired glucose tolerance and insulin sensitivity, and increased hepatic triglycerides (TG) and diacylglycerol (DAG) accumulation; SELENOS expression in subcutaneous and omental adipose tissue was elevated in obese human subjects, and act as a positive regulator for adipogenesis in 3T3-L1 preadipocytes; knockdown of SELENOS in Min6 β-cells induced β-cell apoptosis and reduced cell proliferation. SELENOS also participates in the early stages of AS, notably by enhancing endothelial function, curbing the expression of adhesion molecules, and lessening leukocyte recruitment-actions that collectively reduce the formation of foam cells. Furthermore, SELENOS forestalls the apoptosis of vascular smooth muscle cells (VSMCs) and macrophages, mitigates vascular calcification, and alleviates inflammation in macrophages and CD4+ T cells. These actions help stifle the creation of unstable plaque characterized by thinner fibrous caps, larger necrotic cores, heightened inflammation, and more extensive vascular calcification-features seen in advanced atherosclerotic lesion development. Additionally, serum SELENOS could function as a potential biomarker, and SELENOS single nucleotide polymorphisms (SNPs) rs4965814, rs28628459, and rs9806366, might be effective gene markers for atherosclerosis-related diseases in diabetes. This review accentuates the pathophysiological processes of atherosclerosis in diabetes and amasses current evidence on SELENOS's potential therapeutic benefits or as predictive biomarkers in the various stages of diabetes-induced atherosclerosis.
Collapse
Affiliation(s)
- Shan-Shan Yu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
- Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, 116011, Liaoning, China
| | - Jian-Ling Du
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
- Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, 116011, Liaoning, China.
| |
Collapse
|
5
|
Ghelichkhani F, Gonzalez FA, Kapitonova MA, Rozovsky S. Selenoprotein S Interacts with the Replication and Transcription Complex of SARS-CoV-2 by Binding nsp7. J Mol Biol 2023; 435:168008. [PMID: 36773692 PMCID: PMC9911985 DOI: 10.1016/j.jmb.2023.168008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/05/2022] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) replicates and evades detection using ER membranes and their associated protein machinery. Among these hijacked human proteins is selenoprotein S (selenos). This selenoprotein takes part in the protein quality control, signaling, and the regulation of cytokine secretion. While the role of selenos in the viral life cycle is not yet known, it has been reported to interact with SARS-CoV-2 nonstructural protein 7 (nsp7), a viral protein essential for the replication of the virus. We set to study whether selenos and nsp7 interact directly and if they can still bind when nsp7 is bound to the replication and transcription complex of the virus. Using biochemical assays, we show that selenos binds directly to nsp7. In addition, we found that selenos can bind to nsp7 when it is in a complex with the coronavirus's minimal replication and transcription complex, comprised of nsp7, nsp8, and the RNA-dependent RNA polymerase nsp12. In addition, through crosslinking experiments, we mapped the interaction sites of selenos and nsp7 in the replication complex and showed that the hydrophobic segment of selenos is essential for binding to nsp7. This arrangement leaves an extended helix and the intrinsically disordered segment of selenos-including the reactive selenocysteine-exposed and free to potentially recruit additional proteins to the replication and transcription complex.
Collapse
Affiliation(s)
- Farid Ghelichkhani
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Fabio A Gonzalez
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Mariia A Kapitonova
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Sharon Rozovsky
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
6
|
Toh P, Seale LA, Berry MJ, Torres DJ. Prolonged maternal exposure to glucocorticoids alters selenoprotein expression in the developing brain. Front Mol Neurosci 2023; 16:1115993. [PMID: 37033382 PMCID: PMC10080067 DOI: 10.3389/fnmol.2023.1115993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Aberrant activation of the stress-response system in early life can alter neurodevelopment and cause long-term neurological changes. Activation of the hypothalamic-pituitary-adrenal axis releases glucocorticoids into the bloodstream, to help the organism adapt to the stressful stimulus. Elevated glucocorticoid levels can promote the accumulation of reactive oxygen species, and the brain is highly susceptible to oxidative stress. The essential trace element selenium is obtained through diet, is used to synthesize antioxidant selenoproteins, and can mitigate glucocorticoid-mediated oxidative damage. Glucocorticoids can impair antioxidant enzymes in the brain, and could potentially influence selenoprotein expression. We hypothesized that exposure to high levels of glucocorticoids would disrupt selenoprotein expression in the developing brain. C57 wild-type dams of recently birthed litters were fed either a moderate (0.25 ppm) or high (1 ppm) selenium diet and administered corticosterone (75 μg/ml) via drinking water during postnatal days 1 to 15, after which the brains of the offspring were collected for western blot analysis. Glutathione peroxidase 1 and 4 levels were increased by maternal corticosterone exposure within the prefrontal cortex, hippocampus, and hypothalamus of offspring. Additionally, levels of the glucocorticoid receptor were decreased in the hippocampus and selenoprotein W was elevated in the hypothalamus by corticosterone. Maternal consumption of a high selenium diet independently decreased glucocorticoid receptor levels in the hippocampus of offspring of both sexes, as well as in the prefrontal cortex of female offspring. This study demonstrates that early life exposure to excess glucocorticoid levels can alter selenoprotein levels in the developing brain.
Collapse
Affiliation(s)
| | | | | | - Daniel J. Torres
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
7
|
An KJ, Hanato AN, Hui KW, Pitts MW, Seale LA, Nicholson JL, Toh P, Kim JK, Berry MJ, Torres DJ. Selenium Protects Mouse Hypothalamic Cells from Glucocorticoid-Induced Endoplasmic Reticulum Stress Vulnerability and Insulin Signaling Impairment. Antioxidants (Basel) 2023; 12:526. [PMID: 36830084 PMCID: PMC9952756 DOI: 10.3390/antiox12020526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The use of glucocorticoid medications is known to cause metabolic side effects such as overeating, excess weight gain, and insulin resistance. The hypothalamus, a central regulator of feeding behavior and energy expenditure, is highly responsive to glucocorticoids, and it has been proposed that it plays a role in glucocorticoid-induced metabolic defects. Glucocorticoids can alter the expression and activity of antioxidant enzymes and promote the accumulation of reactive oxygen species. Recent evidence indicates that selenium can counter the effects of glucocorticoids, and selenium is critical for proper hypothalamic function. This study sought to determine whether selenium is capable of protecting hypothalamic cells from dysfunction caused by glucocorticoid exposure. We treated mHypoE-44 mouse hypothalamic cells with corticosterone to study the effects on cellular physiology and the involvement of selenium. We found that corticosterone administration rendered cells more vulnerable to endoplasmic reticulum stress and the subsequent impairment of insulin signaling. Supplementing the cell culture media with additional selenium alleviated endoplasmic reticulum stress and promoted insulin signaling. These findings implicate a protective role of selenium against chronic glucocorticoid-induced hypothalamic dysfunction.
Collapse
Affiliation(s)
- Katlyn J. An
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai‘i, Honolulu, HI 96813, USA
| | - Ashley N. Hanato
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai‘i, Honolulu, HI 96813, USA
| | - Katherine W. Hui
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai‘i, Honolulu, HI 96813, USA
| | - Matthew W. Pitts
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai‘i, Honolulu, HI 96813, USA
| | - Lucia A. Seale
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawai‘i, Honolulu, HI 96822, USA
| | - Jessica L. Nicholson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai‘i, Honolulu, HI 96813, USA
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawai‘i, Honolulu, HI 96822, USA
| | - Pamela Toh
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawai‘i, Honolulu, HI 96822, USA
| | - Jun Kyoung Kim
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai‘i, Honolulu, HI 96813, USA
| | - Marla J. Berry
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawai‘i, Honolulu, HI 96822, USA
| | - Daniel J. Torres
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawai‘i, Honolulu, HI 96813, USA
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawai‘i, Honolulu, HI 96822, USA
| |
Collapse
|
8
|
Toh P, Nicholson JL, Vetter AM, Berry MJ, Torres DJ. Selenium in Bodily Homeostasis: Hypothalamus, Hormones, and Highways of Communication. Int J Mol Sci 2022; 23:15445. [PMID: 36499772 PMCID: PMC9739294 DOI: 10.3390/ijms232315445] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
The ability of the body to maintain homeostasis requires constant communication between the brain and peripheral tissues. Different organs produce signals, often in the form of hormones, which are detected by the hypothalamus. In response, the hypothalamus alters its regulation of bodily processes, which is achieved through its own pathways of hormonal communication. The generation and transmission of the molecules involved in these bi-directional axes can be affected by redox balance. The essential trace element selenium is known to influence numerous physiological processes, including energy homeostasis, through its various redox functions. Selenium must be obtained through the diet and is used to synthesize selenoproteins, a family of proteins with mainly antioxidant functions. Alterations in selenium status have been correlated with homeostatic disturbances in humans and studies with animal models of selenoprotein dysfunction indicate a strong influence on energy balance. The relationship between selenium and energy metabolism is complicated, however, as selenium has been shown to participate in multiple levels of homeostatic communication. This review discusses the role of selenium in the various pathways of communication between the body and the brain that are essential for maintaining homeostasis.
Collapse
Affiliation(s)
- Pamela Toh
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Jessica L. Nicholson
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Alyssa M. Vetter
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
- School of Human Nutrition, McGill University, Montreal, QC H3A 0G4, Canada
| | - Marla J. Berry
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Daniel J. Torres
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
9
|
Ghelichkhani F, Gonzalez FA, Kapitonova MA, Schaefer-Ramadan S, Liu J, Cheng R, Rozovsky S. Selenoprotein S: A versatile disordered protein. Arch Biochem Biophys 2022; 731:109427. [PMID: 36241082 PMCID: PMC10026367 DOI: 10.1016/j.abb.2022.109427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022]
Abstract
Selenoprotein S (selenos) is a small, intrinsically disordered membrane protein that is associated with various cellular functions, such as inflammatory processes, cellular stress response, protein quality control, and signaling pathways. It is primarily known for its contribution to the ER-associated degradation (ERAD) pathway, which governs the extraction of misfolded proteins or misassembled protein complexes from the ER to the cytosol for degradation by the proteasome. However, selenos's other cellular roles in signaling are equally vital, including the control of transcription factors and cytokine levels. Consequently, genetic polymorphisms of selenos are associated with increased risk for diabetes, dyslipidemia, and cardiovascular diseases, while high expression levels correlate with poor prognosis in several cancers. Its inhibitory role in cytokine secretion is also exploited by viruses. Since selenos binds multiple protein complexes, however, its specific contributions to various cellular pathways and diseases have been difficult to establish. Thus, the precise cellular functions of selenos and their interconnectivity have only recently begun to emerge. This review aims to summarize recent insights into the structure, interactome, and cellular roles of selenos.
Collapse
Affiliation(s)
- Farid Ghelichkhani
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA
| | - Fabio A Gonzalez
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA
| | - Mariia A Kapitonova
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA
| | | | - Jun Liu
- Enlaza Therapeutics, 11099 N. Torrey Pines Rd, suite 290, La Jolla, CA, 92037, USA
| | - Rujin Cheng
- NGM Biopharmaceuticals, Inc., 333 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Sharon Rozovsky
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
10
|
Jehan C, Cartier D, Bucharles C, Anouar Y, Lihrmann I. Emerging roles of ER-resident selenoproteins in brain physiology and physiopathology. Redox Biol 2022; 55:102412. [PMID: 35917681 PMCID: PMC9344019 DOI: 10.1016/j.redox.2022.102412] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/05/2022] [Accepted: 07/14/2022] [Indexed: 12/23/2022] Open
Abstract
The brain has a very high oxygen consumption rate and is particularly sensitive to oxidative stress. It is also the last organ to suffer from a loss of selenium (Se) in case of deficiency. Se is a crucial trace element present in the form of selenocysteine, the 21st proteinogenic amino acid present in selenoproteins, an essential protein family in the brain that participates in redox signaling. Among the most abundant selenoproteins in the brain are glutathione peroxidase 4 (GPX4), which reduces lipid peroxides and prevents ferroptosis, and selenoproteins W, I, F, K, M, O and T. Remarkably, more than half of them are proteins present in the ER and recent studies have shown their involvement in the maintenance of ER homeostasis, glycoprotein folding and quality control, redox balance, ER stress response signaling pathways and Ca2+ homeostasis. However, their molecular functions remain mostly undetermined. The ER is a highly specialized organelle in neurons that maintains the physical continuity of axons over long distances through its continuous distribution from the cell body to the nerve terminals. Alteration of this continuity can lead to degeneration of distal axons and subsequent neuronal death. Elucidation of the function of ER-resident selenoproteins in neuronal pathophysiology may therefore become a new perspective for understanding the pathophysiology of neurological diseases. Here we summarize what is currently known about each of their molecular functions and their impact on the nervous system during development and stress.
Collapse
Affiliation(s)
- Cédric Jehan
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Dorthe Cartier
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Christine Bucharles
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Youssef Anouar
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Isabelle Lihrmann
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France.
| |
Collapse
|
11
|
Molecular characterization and tissue distribution of nine selenoprotein genes in grass carp Ctenopharyngodon idella and their mRNA expressions in response to high-fat diet and high-fat diet supplemented with selenium. Comp Biochem Physiol B Biochem Mol Biol 2021; 259:110706. [PMID: 34954089 DOI: 10.1016/j.cbpb.2021.110706] [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: 09/13/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022]
Abstract
Our previous study has found that selenium (Se) can alleviate lipid accumulation caused by high-fat diet (HFD) in fish. This study aims to explore the selenoproteins (SePs) in grass carp Ctenopharyngodon idella by characterizing cDNAs of nine SeP genes (SELENOF, SELENOM, SELENOS, SELENOP1, SELENOP2, SELENOE, SELENOL, SELENOU1a and SELENOU1b) and measuring their transcriptional activity in response to HFD and HFD supplemented with 0.3 mg/Kg and 0.6 mg/Kg of Se (HSe 0.3 and HSe 0.6). Firstly, the nine SeP genes in grass carp encoded proteins with conserved functional protein regions in fish and other vertebrates. Secondly, the nine SeP genes except SELENOS showed high expression levels in the hepatopancreas, but in the adipose tissue, only SELENOS, SELENOE and SELENOU1b showed high expression levels. Further, HFD significantly up-regulated the expressions of SELENOF and SELENOS in the hepatopancreas and SELENOM in the adipose tissue of grass carp (P < 0.05), but significantly down-regulated the expressions of SELENOU1b in the hepatopancreas, SELENOP2, SELENOE, SELENOL and SELENOU1a in the adipose tissue and SELENOM in the muscle of grass carp (P < 0.05). In addition, for the hepatopancreas, the expressions of SELENOS in the HSe 0.3 group and SELENOF, SELENOM and SELENOP2 in the HSe 0.6 group significantly decreased compared with the HFD group (P < 0.05). For the adipose tissue, the expressions of SELENOF, SELENOP2, SELENOL, SELENOU1a and SELENOU1b in the HSe 0.3 group and SELENOP2, SELENOE, SELENOU1a and SELENOU1b in the HSe 0.6 group significantly increased compared with the HFD group (P < 0.05). In summary, the transcriptional activities of the nine SeP genes were regulated by the HFD and HFD supplemented with Se, indicating the potential role of these genes in the Se regulated lipid metabolism processes in grass carp, which is worthy of in-depth study.
Collapse
|
12
|
Al-Sayegh M, Ali H, Jamal MH, ElGindi M, Chanyong T, Al-Awadi K, Abu-Farha M. Mouse Embryonic Fibroblast Adipogenic Potential: A Comprehensive Transcriptome Analysis. Adipocyte 2021; 10:1-20. [PMID: 33345692 PMCID: PMC7757854 DOI: 10.1080/21623945.2020.1859789] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Our understanding of adipose tissue has progressed from an inert tissue for energy storage to be one of the largest endocrine organs regulating metabolic homoeostasis through its ability to synthesize and release various adipokines that regulate a myriad of pathways. The field of adipose tissue biology is growing due to this association with various chronic metabolic diseases. An important process in the regulation of adipose tissue biology is adipogenesis, which is the formation of new adipocytes. Investigating adipogenesis in vitro is currently a focus for identifying factors that might be utilized in clinically. A powerful tool for such work is high-throughput sequencing which can rapidly identify changes at gene expression level. Various cell models exist for studying adipogenesis and has been used in high-throughput studies, yet little is known about transcriptome profile that underlies adipogenesis in mouse embryonic fibroblasts. This study utilizes RNA-sequencing and computational analysis with DESeq2, gene ontology, protein–protein networks, and robust rank analysis to understand adipogenesis in mouse embryonic fibroblasts in-depth. Our analyses confirmed the requirement of mitotic clonal expansion prior to adipogenesis in this cell model and highlight the role of Cebpa and Cebpb in regulating adipogenesis through interactions of large numbers of genes.
Collapse
Affiliation(s)
- Mohamed Al-Sayegh
- New York University Abu Dhabi, Division of Biology, Abu Dhabi, United Arab Emirates
| | - Hamad Ali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Kuwait City, State of Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Kuwait City, State of Kuwait
| | - Mohammad H Jamal
- Department of Surgery, Faculty of Medicine, Health Sciences Center (HSC), Kuwait University, Kuwait City, State of Kuwait
| | - Mei ElGindi
- New York University Abu Dhabi, Division of Biology, Abu Dhabi, United Arab Emirates
| | - Tina Chanyong
- New York University Abu Dhabi, Division of Biology, Abu Dhabi, United Arab Emirates
| | - Khulood Al-Awadi
- New York University Abu Dhabi, Design Studio, Abu Dhabi, United Arab Emirates
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Kuwait City, State of Kuwait
| |
Collapse
|
13
|
Torres DJ, Alfulaij N, Berry MJ. Stress and the Brain: An Emerging Role for Selenium. Front Neurosci 2021; 15:666601. [PMID: 33935643 PMCID: PMC8081839 DOI: 10.3389/fnins.2021.666601] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/25/2021] [Indexed: 12/04/2022] Open
Abstract
The stress response is an important tool in an organism’s ability to properly respond to adverse environmental conditions in order to survive. Intense acute or chronic elevation of glucocorticoids, a class of stress hormone, can have deleterious neurological effects, however, including memory impairments and emotional disturbances. In recent years, the protective role of the antioxidant micronutrient selenium against the negative impact of externally applied stress has begun to come to light. In this review, we will discuss the effects of stress on the brain, with a focus on glucocorticoid action in the hippocampus and cerebral cortex, and emerging evidence of an ability of selenium to normalize neurological function in the context of various stress and glucocorticoid exposure paradigms in rodent models.
Collapse
Affiliation(s)
- Daniel J Torres
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Naghum Alfulaij
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Marla J Berry
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
14
|
Selenium and Selenoproteins in Adipose Tissue Physiology and Obesity. Biomolecules 2020; 10:biom10040658. [PMID: 32344656 PMCID: PMC7225961 DOI: 10.3390/biom10040658] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/13/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Selenium (Se) homeostasis is tightly related to carbohydrate and lipid metabolism, but its possible roles in obesity development and in adipocyte metabolism are unclear. The objective of the present study is to review the current data on Se status in obesity and to discuss the interference between Se and selenoprotein metabolism in adipocyte physiology and obesity pathogenesis. The overview and meta-analysis of the studies on blood Se and selenoprotein P (SELENOP) levels, as well as glutathione peroxidase (GPX) activity in obese subjects, have yielded heterogenous and even conflicting results. Laboratory studies demonstrate that Se may modulate preadipocyte proliferation and adipogenic differentiation, and also interfere with insulin signaling, and regulate lipolysis. Knockout models have demonstrated that the selenoprotein machinery, including endoplasmic reticulum-resident selenoproteins together with GPXs and thioredoxin reductases (TXNRDs), are tightly related to adipocyte development and functioning. In conclusion, Se and selenoproteins appear to play an essential role in adipose tissue physiology, although human data are inconsistent. Taken together, these findings do not support the utility of Se supplementation to prevent or alleviate obesity in humans. Further human and laboratory studies are required to elucidate associations between Se metabolism and obesity.
Collapse
|
15
|
Okumura F, Fujiki Y, Oki N, Osaki K, Nishikimi A, Fukui Y, Nakatsukasa K, Kamura T. Cul5-type Ubiquitin Ligase KLHDC1 Contributes to the Elimination of Truncated SELENOS Produced by Failed UGA/Sec Decoding. iScience 2020; 23:100970. [PMID: 32200094 PMCID: PMC7090344 DOI: 10.1016/j.isci.2020.100970] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/10/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
The UGA codon signals protein translation termination, but it can also be translated into selenocysteine (Sec, U) to produce selenocysteine-containing proteins (selenoproteins) by dedicated machinery. As Sec incorporation can fail, Sec-containing longer and Sec-lacking shorter proteins co-exist. Cul2-type ubiquitin ligases were recently shown to destabilize such truncated proteins; however, which ubiquitin ligase targets truncated proteins for degradation remained unclear. We report that the Cul5-type ubiquitin ligase KLHDC1 targets truncated SELENOS, a selenoprotein, for proteasomal degradation. SELENOS is involved in endoplasmic reticulum (ER)-associated degradation, which is linked to reactive oxygen species (ROS) production, and the knockdown of KLHDC1 in U2OS cells decreased ER stress-induced cell death. Knockdown of SELENOS increased the cell population with lower ROS levels. Our findings reveal that, in addition to Cul2-type ubiquitin ligases, KLHDC1 is involved in the elimination of truncated oxidoreductase-inactive SELENOS, which would be crucial for maintaining ROS levels and preventing cancer development. KLHDC1 is a Cul5-type ubiquitin ligase KLHDC1 targets immature SELENOS for proteasomal degradation KLHDC1 knockdown in U2OS cells decreases ER stress-induced cell death
Collapse
Affiliation(s)
- Fumihiko Okumura
- Department of Food and Health Sciences, International College of Arts and Sciences, Fukuoka Women's University, Fukuoka 813-8582, Japan.
| | - Yuha Fujiki
- Department of Food and Health Sciences, International College of Arts and Sciences, Fukuoka Women's University, Fukuoka 813-8582, Japan
| | - Nodoka Oki
- Department of Food and Health Sciences, International College of Arts and Sciences, Fukuoka Women's University, Fukuoka 813-8582, Japan
| | - Kana Osaki
- Department of Food and Health Sciences, International College of Arts and Sciences, Fukuoka Women's University, Fukuoka 813-8582, Japan
| | - Akihiko Nishikimi
- Laboratory of Biosafety Research, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience and Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kunio Nakatsukasa
- Graduate School of Natural Sciences, Nagoya City University, Aichi 467-8501, Japan
| | - Takumi Kamura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan.
| |
Collapse
|
16
|
Leonardi A, Evke S, Lee M, Melendez JA, Begley TJ. Epitranscriptomic systems regulate the translation of reactive oxygen species detoxifying and disease linked selenoproteins. Free Radic Biol Med 2019; 143:573-593. [PMID: 31476365 PMCID: PMC7650020 DOI: 10.1016/j.freeradbiomed.2019.08.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Here we highlight the role of epitranscriptomic systems in post-transcriptional regulation, with a specific focus on RNA modifying writers required for the incorporation of the 21st amino acid selenocysteine during translation, and the pathologies linked to epitranscriptomic and selenoprotein defects. Epitranscriptomic marks in the form of enzyme-catalyzed modifications to RNA have been shown to be important signals regulating translation, with defects linked to altered development, intellectual impairment, and cancer. Modifications to rRNA, mRNA and tRNA can affect their structure and function, while the levels of these dynamic tRNA-specific epitranscriptomic marks are stress-regulated to control translation. The tRNA for selenocysteine contains five distinct epitranscriptomic marks and the ALKBH8 writer for the wobble uridine (U) has been shown to be vital for the translation of the glutathione peroxidase (GPX) and thioredoxin reductase (TRXR) family of selenoproteins. The reactive oxygen species (ROS) detoxifying selenocysteine containing proteins are a prime examples of how specialized translation can be regulated by specific tRNA modifications working in conjunction with distinct codon usage patterns, RNA binding proteins and specific 3' untranslated region (UTR) signals. We highlight the important role of selenoproteins in detoxifying ROS and provide details on how epitranscriptomic marks and selenoproteins can play key roles in and maintaining mitochondrial function and preventing disease.
Collapse
Affiliation(s)
- Andrea Leonardi
- Colleges of Nanoscale Science and Engineering, University at Albany, State University of New York, Albany, NY, USA
| | - Sara Evke
- Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, USA
| | - May Lee
- Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, USA
| | - J Andres Melendez
- Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, USA.
| | - Thomas J Begley
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA; RNA Institute, University at Albany, State University of New York, Albany, NY, USA.
| |
Collapse
|
17
|
Ha HY, Alfulaij N, Berry MJ, Seale LA. From Selenium Absorption to Selenoprotein Degradation. Biol Trace Elem Res 2019; 192:26-37. [PMID: 31222623 PMCID: PMC6801053 DOI: 10.1007/s12011-019-01771-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022]
Abstract
Selenium is an essential dietary micronutrient. Ingested selenium is absorbed by the intestines and transported to the liver where it is mostly metabolized to selenocysteine (Sec). Sec is then incorporated into selenoproteins, including selenoprotein P (SELENOP), which is secreted into plasma and serves as a source of selenium to other tissues of the body. Herein, we provide an overview of the biology of selenium from its absorption and distribution to selenoprotein uptake and degradation, with a particular focus on the latter. Molecular mechanisms of selenoprotein degradation include the lysosome-mediated pathway for SELENOP and endoplasmic reticulum-mediated degradation of selenoproteins via ubiquitin-activated proteasomal pathways. Ubiquitin-activated pathways targeting full-length selenoproteins include the peroxisome proliferator-activated receptor gamma-dependent pathway and substrate-dependent ubiquitination. An alternate mechanism is utilized for truncated selenoproteins, in which cullin-RING E3 ubiquitin ligase 2 targets the defective proteins for ubiquitin-proteasomal degradation. Selenoproteins, particularly SELENOP, may have their Sec residues reutilized for new selenoprotein synthesis via Sec decomposition. This review will explore these aspects in selenium biology, providing insights to knowledge gaps that remain to be uncovered.
Collapse
Affiliation(s)
- Herena Y Ha
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI, 96813, USA
| | - Naghum Alfulaij
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI, 96813, USA
| | - Marla J Berry
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI, 96813, USA
| | - Lucia A Seale
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI, 96813, USA.
| |
Collapse
|
18
|
Yu S, Liu X, Men L, Yao J, Xing Q, Du J. Selenoprotein S protects against high glucose-induced vascular endothelial apoptosis through the PKCβII/JNK/Bcl-2 pathway. J Cell Biochem 2019; 120:8661-8675. [PMID: 30485531 DOI: 10.1002/jcb.28154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 11/05/2018] [Indexed: 01/24/2023]
Abstract
Vascular endothelial apoptosis is closely associated with the pathogenesis and progression of diabetic macrovascular diseases. Selenoprotein S (SelS) participates in the protection of vascular endothelial and smooth muscle cells from oxidative and endoplasmic reticulum stress-induced injury. However, whether SelS can protect vascular endothelium from high glucose (HG)-induced apoptosis and the underlying mechanism remains unclear. The present study preliminarily analyzed aortic endothelial apoptosis and SelS expression in diabetic rats in vivo and the effects of HG on human umbilical vein endothelial cell (HUVEC) apoptosis and SelS expression in vitro. Subsequently, SelS expression was up- or downregulated in HUVECs using the pcDNA3.1-SelS recombinant plasmid and SelS-specific small interfering RNAs, and the effects of high/low SelS expression on HG-induced HUVEC apoptosis and a possible molecular mechanism were analyzed. As expected, HG induced vascular endothelial apoptosis and upregulated endothelial SelS expression in vivo and in vitro. SelS overexpression in HUVECs suppressed HG-induced increase in apoptosis and cleaved caspase3 level, accompanied by reduced protein kinase CβII (PKCβII), c-JUN N-terminal kinase (JNK), and B-cell lymphoma/leukemia-2 (Bcl-2) phosphorylation. In contrast, inhibiting SelS expression in HUVECs further aggravated HG-induced increase in apoptosis and cleaved caspase3 level, which was accompanied by increased PKCβII, JNK, and Bcl-2 phosphorylation. Pretreatment with PKC activators blocked the protective effects of SelS and increased the apoptosis and cleaved caspase3 level in HUVECs. In summary, SelS protects vascular endothelium from HG-induced apoptosis, and this was achieved through the inhibition of PKCβII/JNK/Bcl-2 pathway to eventually inhibit caspase3 activation. SelS may be a promising target for the prevention and treatment of diabetic macrovascular complications.
Collapse
Affiliation(s)
- Shanshan Yu
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaoying Liu
- Department of General Practice, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lili Men
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Junjie Yao
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qian Xing
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianling Du
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
Abstract
Cultured meat, or tissue engineered meat, is a promising alternative to conventional meat production. In order to realistically mimic the multiple tissue types found in beef, food-compatible methods for bovine fat tissue engineering must be developed. We present a protocol for the isolation of adipose tissue-derived preadipocytes and subsequent adipogenic differentiation through free fatty acid stimulation. Differentiating preadipocytes can be either grown in 2D culture conditions or seeded in 3D alginate scaffolds. Differentiation is visually confirmed through lipophilic staining.
Collapse
Affiliation(s)
- Frea Mehta
- Department of Physiology, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Ruud Theunissen
- Department of Physiology, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Mark J Post
- Department of Physiology, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
20
|
Addinsall AB, Martin SD, Collier F, Conlan XA, Foletta VC, Stupka N. Differential regulation of cellular stress responses by the endoplasmic reticulum-resident Selenoprotein S (Seps1) in proliferating myoblasts versus myotubes. Physiol Rep 2018; 6:e13926. [PMID: 30557449 PMCID: PMC6296459 DOI: 10.14814/phy2.13926] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 01/03/2023] Open
Abstract
The antioxidant Selenoprotein S (Seps1, Selenos) is an endoplasmic reticulum (ER)-resident protein associated with metabolic and inflammatory disease. While Seps1 is highly expressed in skeletal muscle, its mechanistic role as an antioxidant in skeletal muscle cells is not well characterized. In C2C12 myotubes treated with palmitate for 24 h, endogenous Seps1 protein expression was upregulated twofold. Two different siRNA constructs were used to investigate whether decreased levels of Seps1 exacerbated lipid-induced oxidative and ER stress in C2C12 myotubes and myoblasts, which differ with regards to cell cycle state and metabolic phenotype. In myoblasts, Seps1 protein knockdown of ~50% or ~75% exacerbated cellular stress responses in the presence of palmitate; as indicated by decreased cell viability and proliferation, higher H2 O2 levels, a lower reduced to oxidized glutathione (GSH:GSSG) ratio, and enhanced gene expression of ER and oxidative stress markers. Even in the absence of palmitate, Seps1 knockdown increased oxidative stress in myoblasts. Whereas, in myotubes in the presence of palmitate, a ~50% knockdown of Seps1 was associated with a trend toward a marginal (3-5%) decrease in viability (P = 0.05), decreased cellular ROS levels, and a reduced mRNA transcript abundance of the cellular stress marker thioredoxin inhibitory binding protein (Txnip). Furthermore, no enhancement of gene markers of ER stress was observed in palmitate-treated myotubes in response to Seps1 knockdown. In conclusion, reduced Seps1 levels exacerbate nutrient-induced cellular stress responses to a greater extent in glycolytic, proliferating myoblasts than in oxidative, differentiated myotubes, thus demonstrating the importance of cell phenotype to Seps1 function.
Collapse
Affiliation(s)
- Alex B. Addinsall
- Centre for Molecular and Medical ResearchSchool of MedicineDeakin UniversityGeelongAustralia
| | - Sheree D. Martin
- Centre for Molecular and Medical ResearchSchool of MedicineDeakin UniversityGeelongAustralia
| | - Fiona Collier
- GCEID, University HospitalBarwon HealthGeelongAustralia
- School of MedicineDeakin UniversityGeelongAustralia
| | - Xavier A. Conlan
- Centre for Chemistry and BiotechnologySchool of Life and Environmental SciencesFaculty of Science, Engineering and Built EnvironmentDeakin UniversityGeelongAustralia
| | - Victoria C. Foletta
- Institute for Physical Activity and Nutrition (IPAN)School of Exercise and Nutrition SciencesDeakin UniversityGeelongAustralia
| | - Nicole Stupka
- Centre for Molecular and Medical ResearchSchool of MedicineDeakin UniversityGeelongAustralia
| |
Collapse
|
21
|
Kim CY, Kim KH. Selenate Prevents Adipogenesis through Induction of Selenoprotein S and Attenuation of Endoplasmic Reticulum Stress. Molecules 2018; 23:molecules23112882. [PMID: 30400605 PMCID: PMC6278472 DOI: 10.3390/molecules23112882] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 12/05/2022] Open
Abstract
The conversion of preadipocytes to adipocytes (adipogenesis) is a potential target to treat or prevent obesity. Selenate, an inorganic form of selenium, elicits diverse health benefits, mainly through its incorporation into selenoproteins. The individual roles of selenium and certain selenoproteins have been reported. However, the effects of selenate treatment on selenoproteins in adipocytes are unclear. In this study, the effects of selenate pretreatment on selenoprotein and endoplasmic reticulum (ER) stress during adipogenesis were examined in vitro. The selenate pretreatment dose-dependently suppressed the adipogenesis of 3T3-L1 preadipocytes. The selenate pretreatment at 50 μM for 24 h almost completely suppressed adipogenesis without cytotoxic effects. The expression of the adipogenic genes peroxisome proliferator-activated receptor gamma, CCAAT-enhancer binding protein alpha, and leptin was suppressed by selenate. This pretreatment also upregulated selenoprotein S (SEPS1), an ER resident selenoprotein that reduces ER stress, and prevented dexamethasone-induced SEPS1 degradation during the early stage of adipogenesis. The selenate-inhibited adipogenesis was associated with an attenuation of ER stress. The expression of the ER stress marker genes was upregulated during the early stage of differentiation, whereas the selenate pretreatment suppressed the mRNA expression of the XBP1 and C/EBP homologous protein. The collective data suggest a preventive role of selenate and SEPS1 in adipogenesis, and support a novel dietary approach to prevent obesity.
Collapse
Affiliation(s)
- Choon Young Kim
- Department of Food and Nutrition, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Kee-Hong Kim
- Department of Food Science, Purdue University, West Lafayette, IN 47897, USA.
| |
Collapse
|
22
|
Degradation of selenoprotein S and selenoprotein K through PPARγ-mediated ubiquitination is required for adipocyte differentiation. Cell Death Differ 2018; 26:1007-1023. [PMID: 30082770 DOI: 10.1038/s41418-018-0180-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 07/06/2018] [Accepted: 07/18/2018] [Indexed: 01/15/2023] Open
Abstract
Adipocyte differentiation is known to be related with endoplasmic reticulum (ER) stress. We have reported that selenoprotein S (SelS) and selenoprotein K (SelK) have a function in the regulation of ER stress and ER-associated degradation. However, the association between adipocyte differentiation and the ER-resident selenoproteins, SelS and SelK, is unclear. In this study, we found that the levels of SelS and SelK were decreased during adipocyte differentiation and were inversely related to the levels of peroxisome proliferator-activated receptor γ (PPARγ), a central regulator of adipogenesis. It has been recently reported that PPARγ has E3 ubiquitin ligase activity. Here, we report that PPARγ directly interacts with both SelS and SelK via its ligand-binding domain to induce ubiquitination and degradation of the selenoproteins. Lysine residues at the 150th position of SelS and the 47th and 48th positions of SelK were the target sites for ubiquitination by PPARγ. We also found that adipocyte differentiation was inhibited when either SelS or SelK was not degraded by PPARγ. Thus, these data indicate that PPARγ-mediated ubiquitination and degradation of SelS and SelK is required for adipocyte differentiation.
Collapse
|
23
|
Selenoprotein S silencing triggers mouse hepatoma cells apoptosis and necrosis involving in intracellular calcium imbalance and ROS-mPTP-ATP. Biochim Biophys Acta Gen Subj 2018; 1862:2113-2123. [PMID: 30017912 DOI: 10.1016/j.bbagen.2018.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/22/2018] [Accepted: 07/04/2018] [Indexed: 12/26/2022]
Abstract
Selenoprotein S (SelenoS) is one of the cellular endoplasmic reticulum (ER) and membrane located selenoproteins, and it has the main functions of anti-oxidation, anti-apoptosis and anti-ER stress. To investigate the effect of SelenoS silencing on mouse hepatoma cell death and the intracellular biological function of SelenoS, we knocked down SelenoS in Hepa1-6 cells, and detected ER stress, intracellular calcium homeostasis, mitochondrial dynamics, apoptosis and necrosis. To further explore whether reactive oxygen species (ROS) has an effect on apoptosis and necrosis under SelenoS silencing, we used NAC (2.5 mM) to pretreat cells, and detected ΔΨm, ATP, and apoptosis and necrosis rates. SelenoS silencing broke the intracellular calcium homeostasis, induced mitochondrial dynamic disorder, ROS accumulation, loss of ΔΨm and ATP, and triggered apoptosis and necrosis in mouse hepatoma cells. The clearance of ROS alleviated the loss of ΔΨm and ATP caused by silencing of SelenoS, reduced cell necrosis and increased apoptosis. However, SelenoS silencing did not cause ER stress in Hepa1-6 cells. These results indicate that SelenoS silencing triggers mouse hepatoma cells apoptosis and necrosis through affecting intracellular calcium homeostasis and ROS-mPTP-ATP participates in cell death transformation from apoptosis to necrosis to rise damage.
Collapse
|
24
|
Gao Y, Zhu H, Yang F, Wang Q, Feng Y, Zhang C. Glucocorticoid-activated IRE1α/XBP-1s signaling: an autophagy-associated protective pathway against endotheliocyte damage. Am J Physiol Cell Physiol 2018; 315:C300-C309. [PMID: 29768047 DOI: 10.1152/ajpcell.00009.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Glucocorticoid-induced endothelial injury has been reported in several diseases. Although there are several theories, the exact mechanism underlying the role of glucocorticoids in this process remains unclear. Autophagy has been reported to occur as a response to different stimuli and can affect cell survival and function. In this study, we found that glucocorticoids induced apoptosis and endoplasmic reticulum (ER) stress in endotheliocytes. Furthermore, we discovered that glucocorticoids induced autophagy in these cells and the inositol requiring protein 1 (IRE1α)/X-box binding protein 1s (XBP-1s) axis, one of the downstream signaling pathways of ER stress, was associated with the glucocorticoid-induced autophagy. The autophagy partly protected endotheliocytes from glucocorticoid-induced apoptosis and inhibition of proliferation. In conclusion, glucocorticoid-induced endoplasmic reticulum stress activated the IRE1α/XBP-1s signaling and induced autophagy, which, in turn, played a protective role in endotheliocyte survival and proliferation, avoiding further cellular damage caused by glucocorticoids.
Collapse
Affiliation(s)
- Yanchun Gao
- Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Hongyi Zhu
- Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Fan Yang
- Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Qiyang Wang
- Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Yong Feng
- Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital , Shanghai , China
| |
Collapse
|
25
|
Addinsall AB, Wright CR, Shaw CS, McRae NL, Forgan LG, Weng CH, Conlan XA, Francis PS, Smith ZM, Andrikopoulos S, Stupka N. Deficiency of selenoprotein S, an endoplasmic reticulum resident oxidoreductase, impairs the contractile function of fast-twitch hindlimb muscles. Am J Physiol Regul Integr Comp Physiol 2018; 315:R380-R396. [PMID: 29668323 DOI: 10.1152/ajpregu.00244.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Selenoprotein S (Seps1) is an endoplasmic reticulum (ER) resident antioxidant implicated in ER stress and inflammation. In human vastus lateralis and mouse hindlimb muscles, Seps1 localization and expression were fiber-type specific. In male Seps1+/- heterozygous mice, spontaneous physical activity was reduced compared with wild-type littermates ( d = 1.10, P = 0.029). A similar trend was also observed in Seps1-/- knockout mice ( d = 1.12, P = 0.051). Whole body metabolism, body composition, extensor digitorum longus (EDL), and soleus mass and myofiber diameter were unaffected by genotype. However, in isolated fast EDL muscles from Seps1-/- knockout mice, the force frequency curve (FFC; 1-120 Hz) was shifted downward versus EDL muscles from wild-type littermates ( d = 0.55, P = 0.002), suggestive of reduced strength. During 4 min of intermittent, submaximal (60 Hz) stimulation, the genetic deletion or reduction of Seps1 decreased EDL force production ( d = 0.52, P < 0.001). Furthermore, at the start of the intermittent stimulation protocol, when compared with the 60-Hz stimulation of the FFC, EDL muscles from Seps1-/- knockout or Seps1+/- heterozygous mice produced 10% less force than those from wild-type littermates ( d = 0.31, P < 0.001 and d = 0.39, P = 0.015). This functional impairment was associated with reduced mRNA transcript abundance of thioredoxin-1 ( Trx1), thioredoxin interacting protein ( Txnip), and the ER stress markers Chop and Grp94, whereas, in slow soleus muscles, Seps1 deletion did not compromise contractile function and Trx1 ( d = 1.38, P = 0.012) and Txnip ( d = 1.27, P = 0.025) gene expression was increased. Seps1 is a novel regulator of contractile function and cellular stress responses in fast-twitch muscles.
Collapse
Affiliation(s)
- Alex B Addinsall
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Waurn Ponds, Victoria , Australia
| | - Craig R Wright
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Waurn Ponds, Victoria , Australia
| | - Chris S Shaw
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Waurn Ponds, Victoria , Australia
| | - Natasha L McRae
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Waurn Ponds, Victoria , Australia
| | - Leonard G Forgan
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Waurn Ponds, Victoria , Australia
| | - Chia-Heng Weng
- Department of Medicine-Austin Health, The University of Melbourne , Heidelberg, Victoria , Australia
| | - Xavier A Conlan
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, Waurn Ponds, Victoria , Australia
| | - Paul S Francis
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, Waurn Ponds, Victoria , Australia
| | - Zoe M Smith
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, Waurn Ponds, Victoria , Australia
| | - Sofianos Andrikopoulos
- Department of Medicine-Austin Health, The University of Melbourne , Heidelberg, Victoria , Australia
| | - Nicole Stupka
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Waurn Ponds, Victoria , Australia
| |
Collapse
|
26
|
Yu SS, Du JL. Selenoprotein S: a therapeutic target for diabetes and macroangiopathy? Cardiovasc Diabetol 2017; 16:101. [PMID: 28797256 PMCID: PMC5553675 DOI: 10.1186/s12933-017-0585-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022] Open
Abstract
Inflammatory response, oxidative stress, and endoplasmic reticulum (ER) stress are important pathophysiological bases of the occurrence and development of diabetes mellitus (DM) and macroangiopathy complications. Selenoprotein S (SELENOS) is involved in the regulation of these mechanisms; therefore, its association with DM and macroangiopathy has gradually received attention from scholars worldwide. SELENOS has different biological functions in different tissues and organs: it exerts antioxidant protection and has anti-ER stress effects in the pancreas and blood vessels, while it promotes the occurrence and development of insulin resistance in the liver, adipose tissue, and skeletal muscle. In addition, studies have confirmed that some SELENOS gene polymorphisms can influence the inflammatory response and are closely associated with the risk for developing DM and macroangiopathy. Therefore, comprehensive understanding of the association between SELENOS and inflammation, oxidative stress, and ER stress may better elucidate and supplement the pathogenic mechanisms of DM and macroangiopathy complications. Furthermore, in-depth investigation of the association of SELENOS function in different tissues and organs with DM and macroangiopathy may facilitate the development of new strategies for the prevention and treatment of DM and macrovascular complications. Here, we summarize the consensus and controversy regarding functions of SELENOS on currently available evidence.
Collapse
Affiliation(s)
- Shan-Shan Yu
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Jian-Ling Du
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| |
Collapse
|
27
|
Wright CR, Allsopp GL, Addinsall AB, McRae NL, Andrikopoulos S, Stupka N. A Reduction in Selenoprotein S Amplifies the Inflammatory Profile of Fast-Twitch Skeletal Muscle in the mdx Dystrophic Mouse. Mediators Inflamm 2017; 2017:7043429. [PMID: 28592916 PMCID: PMC5448157 DOI: 10.1155/2017/7043429] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/23/2017] [Accepted: 03/07/2017] [Indexed: 12/20/2022] Open
Abstract
Excessive inflammation is a hallmark of muscle myopathies, including Duchenne muscular dystrophy (DMD). There is interest in characterising novel genes that regulate inflammation due to their potential to modify disease progression. Gene polymorphisms in Selenoprotein S (Seps1) are associated with elevated proinflammatory cytokines, and in vitro SEPS1 is protective against inflammatory stress. Given that SEPS1 is highly expressed in skeletal muscle, we investigated whether the genetic reduction of Seps1 exacerbated inflammation in the mdx mouse. F1 male mdx mice with a heterozygous Seps1 deletion (mdx:Seps1-/+) were generated. The mdx:Seps1-/+ mice had a 50% reduction in SEPS1 protein expression in hindlimb muscles. In the extensor digitorum longus (EDL) muscles, mRNA expression of monocyte chemoattractant protein 1 (Mcp-1) (P = 0.034), macrophage marker F4/80 (P = 0.030), and transforming growth factor-β1 (Tgf-β1) (P = 0.056) were increased in mdx:Seps1-/+ mice. This was associated with a reduction in muscle fibre size; however, ex vivo EDL muscle strength and endurance were unaltered. In dystrophic slow twitch soleus muscles, SEPS1 reduction had no effect on the inflammatory profile nor function. In conclusion, the genetic reduction of Seps1 appears to specifically exacerbate the inflammatory profile of fast-twitch muscle fibres, which are typically more vulnerable to degeneration in dystrophy.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Body Composition/genetics
- Body Composition/physiology
- Female
- Immunohistochemistry
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle Contraction/physiology
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Fast-Twitch/physiology
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/physiology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Muscular Dystrophy, Duchenne/metabolism
- Real-Time Polymerase Chain Reaction
- Selenoproteins/genetics
- Selenoproteins/metabolism
Collapse
Affiliation(s)
- Craig Robert Wright
- Institute for Physical Activity and Nutrition Research (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Giselle Larissa Allsopp
- Institute for Physical Activity and Nutrition Research (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Alex Bernard Addinsall
- Molecular Medical Research SRC, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Natasha Lee McRae
- Molecular Medical Research SRC, School of Medicine, Deakin University, Geelong, VIC, Australia
| | | | - Nicole Stupka
- Molecular Medical Research SRC, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
28
|
Selenoproteins: Antioxidant selenoenzymes and beyond. Arch Biochem Biophys 2016; 595:113-9. [PMID: 27095226 DOI: 10.1016/j.abb.2015.06.024] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/08/2015] [Indexed: 11/21/2022]
Abstract
Adequate intake of the essential trace element and micronutrient selenium is thought to be beneficial for maintaining human health. Selenium may modulate a broad spectrum of key biological processes, including the cellular response to oxidative stress, redox signalling, cellular differentiation, the immune response, and protein folding. Biochemical and cellular effects of selenium are achieved through activities of selenocysteine-containing selenoproteins. This small yet essential group comprises proteins encoded by 25 genes in humans, e.g. oxidoreductases such as glutathione peroxidases (GPx) and thioredoxin reductases (TrxR), as well as the iodothyronine deiodinases (DIO) and the plasma selenium transport protein, selenoprotein P (SePP1). Synthetic selenoorganic compounds, including the GPx mimetic ebselen, have also been applied in biological systems in vitro and in vivo; antioxidant and anti-inflammatory actions of ebselen and its history as a drug candidate are summarised here. Furthermore, we discuss several aspects of selenoprotein biochemistry, ranging from their well-known importance for cellular protection against oxidative damage to more recent data that link selenoprotein expression/activity to enterocyte and adipocyte differentiation and function and to (dys)regulation of insulin action and secretion.
Collapse
|
29
|
Castro JP, Grune T, Speckmann B. The two faces of reactive oxygen species (ROS) in adipocyte function and dysfunction. Biol Chem 2016; 397:709-24. [DOI: 10.1515/hsz-2015-0305] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/08/2016] [Indexed: 12/11/2022]
Abstract
Abstract
White adipose tissue (WAT) is actively involved in the regulation of whole-body energy homeostasis via storage/release of lipids and adipokine secretion. Current research links WAT dysfunction to the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). The expansion of WAT during oversupply of nutrients prevents ectopic fat accumulation and requires proper preadipocyte-to-adipocyte differentiation. An assumed link between excess levels of reactive oxygen species (ROS), WAT dysfunction and T2D has been discussed controversially. While oxidative stress conditions have conclusively been detected in WAT of T2D patients and related animal models, clinical trials with antioxidants failed to prevent T2D or to improve glucose homeostasis. Furthermore, animal studies yielded inconsistent results regarding the role of oxidative stress in the development of diabetes. Here, we discuss the contribution of ROS to the (patho)physiology of adipocyte function and differentiation, with particular emphasis on sources and nutritional modulators of adipocyte ROS and their functions in signaling mechanisms controlling adipogenesis and functions of mature fat cells. We propose a concept of ROS balance that is required for normal functioning of WAT. We explain how both excessive and diminished levels of ROS, e.g. resulting from over supplementation with antioxidants, contribute to WAT dysfunction and subsequently insulin resistance.
Collapse
|
30
|
Lee JH, Park KJ, Jang JK, Jeon YH, Ko KY, Kwon JH, Lee SR, Kim IY. Selenoprotein S-dependent Selenoprotein K Binding to p97(VCP) Protein Is Essential for Endoplasmic Reticulum-associated Degradation. J Biol Chem 2015; 290:29941-52. [PMID: 26504085 DOI: 10.1074/jbc.m115.680215] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Indexed: 01/24/2023] Open
Abstract
Cytosolic valosin-containing protein (p97(VCP)) is translocated to the ER membrane by binding to selenoprotein S (SelS), which is an ER membrane protein, during endoplasmic reticulum-associated degradation (ERAD). Selenoprotein K (SelK) is another known p97(VCP)-binding selenoprotein, and the expression of both SelS and SelK is increased under ER stress. To understand the regulatory mechanisms of SelS, SelK, and p97(VCP) during ERAD, the interaction of the selenoproteins with p97(VCP) was investigated using N2a cells and HEK293 cells. Both SelS and SelK co-precipitated with p97(VCP). However, the association between SelS and SelK did not occur in the absence of p97(VCP). SelS had the ability to recruit p97(VCP) to the ER membrane but SelK did not. The interaction between SelK and p97(VCP) did not occur in SelS knockdown cells, whereas SelS interacted with p97(VCP) in the presence or absence of SelK. These results suggest that p97(VCP) is first translocated to the ER membrane via its interaction with SelS, and then SelK associates with the complex on the ER membrane. Therefore, the interaction between SelK and p97(VCP) is SelS-dependent, and the resulting ERAD complex (SelS-p97(VCP)-SelK) plays an important role in ERAD and ER stress.
Collapse
Affiliation(s)
- Jea Hwang Lee
- From the Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, 1, 5-Ka, Anam-Dong, Sungbuk-Ku, Seoul 136-713, Republic of Korea and
| | - Ki Jun Park
- From the Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, 1, 5-Ka, Anam-Dong, Sungbuk-Ku, Seoul 136-713, Republic of Korea and
| | - Jun Ki Jang
- From the Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, 1, 5-Ka, Anam-Dong, Sungbuk-Ku, Seoul 136-713, Republic of Korea and
| | - Yeong Ha Jeon
- From the Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, 1, 5-Ka, Anam-Dong, Sungbuk-Ku, Seoul 136-713, Republic of Korea and
| | - Kwan Young Ko
- From the Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, 1, 5-Ka, Anam-Dong, Sungbuk-Ku, Seoul 136-713, Republic of Korea and
| | - Joon Hyun Kwon
- From the Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, 1, 5-Ka, Anam-Dong, Sungbuk-Ku, Seoul 136-713, Republic of Korea and
| | - Seung-Rock Lee
- the Departments of Biochemistry and Biomedical Science, Research Center for Aging and Geriatrics, Research Institute of Medical Science, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Ick Young Kim
- From the Laboratory of Cellular and Molecular Biochemistry, Division of Life Sciences, Korea University, 1, 5-Ka, Anam-Dong, Sungbuk-Ku, Seoul 136-713, Republic of Korea and
| |
Collapse
|
31
|
Abstract
SIGNIFICANCE Selenoproteins employ selenium to supplement the chemistry available through the common 20 amino acids. These powerful enzymes are affiliated with redox biology, often in connection with the detection, management, and signaling of oxidative stress. Among them, membrane-bound selenoproteins play prominent roles in signaling pathways, Ca(2+) regulation, membrane complexes integrity, and biosynthesis of lipophilic molecules. RECENT ADVANCES The number of selenoproteins whose physiological roles, protein partners, expression, evolution, and biosynthesis are characterized is steadily increasing, thus offering a more nuanced view of this specialized family. This review focuses on human membrane selenoproteins, particularly the five least characterized ones: selenoproteins I, K, N, S, and T. CRITICAL ISSUES Membrane-bound selenoproteins are the least understood, as it is challenging to provide the membrane-like environment required for their biochemical and biophysical characterization. Hence, their studies rely mostly on biological rather than structural and biochemical assays. Another aspect that has not received much attention is the particular role that their membrane association plays in their physiological function. FUTURE DIRECTIONS Findings cited in this review show that it is possible to infer the structure and the membrane-binding mode of these lesser-studied selenoproteins and design experiments to examine the role of the rare amino acid selenocysteine.
Collapse
Affiliation(s)
- Jun Liu
- Department of Chemistry and Biochemistry, University of Delaware , Newark, Delaware
| | - Sharon Rozovsky
- Department of Chemistry and Biochemistry, University of Delaware , Newark, Delaware
| |
Collapse
|
32
|
Kim CY, Zhu Y, Buhman KK, Kim KH. Dietary selenate attenuates adiposity and improves insulin sensitivity in high-fat diet-induced obese mice. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
33
|
Rajalin AM, Micoogullari M, Sies H, Steinbrenner H. Upregulation of the thioredoxin-dependent redox system during differentiation of 3T3-L1 cells to adipocytes. Biol Chem 2014; 395:667-77. [DOI: 10.1515/hsz-2014-0102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/05/2014] [Indexed: 11/15/2022]
Abstract
Abstract
Hydrogen peroxide acts as a signaling molecule in early adipogenesis. In differentiating adipocytes, elevated hydrogen peroxide generation is balanced through induction of antioxidant enzymes such as catalase and peroxiredoxins. Thioredoxin reductases (TrxR) and glutathione peroxidases (GPx) are selenoenzymes that constitute part of the major thiol-dependent antioxidant systems in cells. Here we show that the protein levels of cytoplasmic/nuclear TrxR1 and mitochondrial TrxR2 increase in the course of adipocyte differentiation of 3T3-L1 cells together with the TrxR2 substrate thioredoxin 2 (Trx2), resulting in elevated TrxR activity in mature adipocytes. Gene and protein expression of the GPx isoenzyme GPx4 was also stimulated during adipogenesis. Chronic exposure of 3T3-L1 cells to the anti-adipogenic factors tumor necrosis factor α (TNF-α) or rapamycin during differentiation suppressed TrxR1 and Trx2 upregulation, concomitantly with inhibition of adipogenesis and lipogenesis. In contrast, TNF-α or rapamycin did not affect expression of TrxRs and their Trx substrates in mature adipocytes. These results indicate that upregulation of the thioredoxin-dependent redox system is linked to the development of an adipocyte phenotype.
Collapse
|