1
|
Zhang P, Zhang C, Yao X, Xie Y, Zhang H, Shao X, Yang X, Nie Q, Ye J, Wu C, Mi H. Selenium yeast improve growth, serum biochemical indices, metabolic ability, antioxidant capacity and immunity in black carp Mylopharyngodnpiceus. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109414. [PMID: 38296006 DOI: 10.1016/j.fsi.2024.109414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/01/2024] [Accepted: 01/28/2024] [Indexed: 02/09/2024]
Abstract
This experiment was conducted to investigate the impacts of dietary selenium yeast (SeY) on the growth performance, fish body composition, metabolic ability, antioxidant capability, immunity and inflammatory responses in juvenile black carp (Mylopharyngodn piceus). The base diet was supplemented with 0.00, 0.30 and 0.60 g/kg SeY (0.04, 0.59 and 1.15 mg/kg of selenium) to form three isonitrogenous and isoenergetic diets for juvenile black carp with a 60-day. Adequate dietary SeY (0.30 and 0.60 g/kg) could significantly increase the weight gain (WG), special growth rate (SGR) compared to the SeY deficient groups (0.00 g/kg) (P < 0.05). Meanwhile, 0.30 and 0.60 g/kg SeY elevated the mRNA levels of selenoprotein T2 (SEPT2), selenoprotein H (SEPH), selenoprotein S (SEPS) and selenoprotein M (SEPM) in the liver and intestine compared with the SeY deficient groups (P < 0.05). Adequate dietary SeY could promote glucose catabolism and utilization through activating glucose transport (GLUT2), glycolysis (GCK, HK, PFK, PK, PDH), tricarboxylic acid cycle (ICDH and MDH), glycogen synthesis (LG, GCS and GBE) and IRS/PI3K/AKT signal pathway molecules (IRS2b, PI3Kc and AKT1) compared with the SeY deficient groups (P < 0.05). Similarly, adequate dietary SeY could improve lipid transport and triglycerides (TG) synthesis through increasing transcription amounts of CD36, GK, DGAT, ACC and FAS in the fish liver compared with the SeY deficient groups (P < 0.05). In addition, adequate SeY could markedly elevate activities of antioxidant enzymes (T-SOD, CAT, GR, GPX) and contents of T-AOC and GSH, while increased transcription amounts of Nrf2, Cu/Zn-SOD, CAT, and GPX in fish liver and intestine (P < 0.05). However, adequate SeY notably decreased contents of MDA, and the mRNA transcription levels of Keap1 in the intestine compared with the SeY deficient groups (P < 0.05). Adequate SeY markedly increased amounts or levels of the immune factors (ALP, ACP, LZM, C3, C4 and IgM) and the transcription levels of innate immune-related functional genes in the liver and intestine (LZM, C3 and C9) compared to the SeY deficient groups (P < 0.05). Moreover, adequate SeY could notably reduce levels of IL-8, IL-1β, and IFN-γ and elevate TGF-1β levels in fish intestine (P < 0.05). The transcription levels of MAPK13, MAPK14 and NF-κB p65 were notably reduced in fish intestine treated with 0.30 and 0.60 g/kg SeY (P < 0.05). In conclusion, these results suggested that 0.30 and 0.60 g/kg SeY could not only improve growth performance, increase Se, glucose and lipid metabolic abilities, enhance antioxidant capabilities and immune responses, but also alleviate inflammation, thereby supplying useful reference for producing artificial feeds in black carp.
Collapse
Affiliation(s)
- Penghui Zhang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Chen Zhang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Xinfeng Yao
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Yuanyuan Xie
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Hao Zhang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Xianping Shao
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Xia Yang
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Qin Nie
- The Hubei Provincial Key Laboratory of Yeast Function, Angel Yeast Co., Ltd, 168 Chengdong Avenue, Yichang, 443000, China
| | - Jinyun Ye
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China
| | - Chenglong Wu
- National-Local Joint Engineering Laboratory of Aquatic Animal Genetic Breeding and Nutrition (Zhejiang), School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou, 313000, China.
| | - Haifeng Mi
- Healthy Aquaculture Key Laboratory of Sichuan Province, Tongwei Co, Ltd, 588 Tianfu Avenue, Chengdu, 610093, China.
| |
Collapse
|
2
|
Li F, Shi Z, Cheng M, Zhou Z, Chu M, Sun L, Zhou JC. Biology and Roles in Diseases of Selenoprotein I Characterized by Ethanolamine Phosphotransferase Activity and Antioxidant Potential. J Nutr 2023; 153:3164-3172. [PMID: 36963501 DOI: 10.1016/j.tjnut.2023.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 03/26/2023] Open
Abstract
Selenoprotein I (SELENOI) has been demonstrated to be an ethanolamine phosphotransferase (EPT) characterized by a nonselenoenzymatic domain and to be involved in the main synthetic branch of phosphatidylethanolamine (PE) in the endoplasmic reticulum. Therefore, defects of SELENOI may affect the health status through the multiple functions of PE. On the other hand, selenium (Se) is covalently incorporated into SELENOI as selenocysteine (Sec) in its peptide, which forms a Sec-centered domain as in the other members of the selenoprotein family. Unlike other selenoproteins, Sec-containing SELENOI was formed at a later stage of animal evolution, and the high conservation of the structural domain for PE synthesis across a wide range of species suggests the importance of EPT activity in supporting the survival and evolution of organisms. A variety of factors, such as species characteristics (age and sex), diet and nutrition (dietary Se and fat intakes), SELENOI-specific properties (tissue distribution and rank in the selenoproteome), etc., synergistically regulate the expression of SELENOI in a tentatively unclear interaction. The N- and C-terminal domains confer 2 distinct biochemical functions to SELENOI, namely PE regulation and antioxidant potential, which may allow it to be involved in numerous physiological processes, including neurological diseases (especially hereditary spastic paraplegia), T cell activation, tumorigenesis, and adipocyte differentiation. In this review, we summarize advances in the biology and roles of SELENOI, shedding light on the precise regulation of SELENOI expression and PE homeostasis by dietary Se intake and pharmaceutical or transgenic approaches to modulate the corresponding pathological status.
Collapse
Affiliation(s)
- Fengna Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhan Shi
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Minning Cheng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhongwei Zhou
- School of Medical, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ming Chu
- Department of Neurosurgery, The Third People's Hospital of Shenzhen, Shenzhen 518112, China
| | - Litao Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China.
| | - Ji-Chang Zhou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China; Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, China.
| |
Collapse
|
3
|
Mo L, Chen J, Cai C, Guo Y, Zeng LH, Li S, Tan J. The Amphiphilic Property and Structure of β-Amyloid Peptide Contribute to Its Impacts on the Activities of Horseradish Peroxidase and Alkaline Phosphatase. ACS Chem Neurosci 2023; 14:3019-3024. [PMID: 37607046 DOI: 10.1021/acschemneuro.3c00391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Recent studies have found that β-amyloid (Aβ) oligomers may play much more important roles than amyloid plaques in the pathogenesis of Alzheimer's disease (AD). However, due to the complexity of Aβ, studying the structural basis of Aβ oligomer toxicity is challenging. Here, we assessed the amphiphilic property and β-hairpin structure of Aβ monomer. The potential impacts of Aβ oligomers and three sequence-modifying peptides on the enzyme activities of horseradish peroxidase (HRP) and alkaline phosphatase (ALP) were further evaluated. We demonstrated that Aβ oligomer possesses the ability to alter the activity of two enzymes. Moreover, modifications on the hydrophobic region and β-turn structure of Aβ monomer significantly alter its impacts on the enzyme activities. In addition, these modifications also change the bonding modes of Aβ monomers or oligomers binding to HRP, as assessed by molecular docking. All of these findings provide direct experimental evidence to reveal the critical roles of the amphiphilic property and β-sheet structure of Aβ monomer in its impacts on protein activity.
Collapse
Affiliation(s)
- Ling Mo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang 550004, China
| | - Jiang Chen
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang 550004, China
| | - Chuanbin Cai
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang 550004, China
| | - Yi Guo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang 550004, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Song Li
- The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Jun Tan
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang 550004, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| |
Collapse
|
4
|
Abstract
Significance: Thioredoxin (Trx) is a powerful antioxidant that reduces protein disulfides to maintain redox stability in cells and is involved in regulating multiple redox-dependent signaling pathways. Recent Advance: The current accumulation of findings suggests that Trx participates in signaling pathways that interact with various proteins to manipulate their dynamic regulation of structure and function. These network pathways are critical for cancer pathogenesis and therapy. Promising clinical advances have been presented by most anticancer agents targeting such signaling pathways. Critical Issues: We herein link the signaling pathways regulated by the Trx system to potential cancer therapeutic opportunities, focusing on the coordination and strengths of the Trx signaling pathways in apoptosis, ferroptosis, immunomodulation, and drug resistance. We also provide a mechanistic network for the exploitation of therapeutic small molecules targeting the Trx signaling pathways. Future Directions: As research data accumulate, future complex networks of Trx-related signaling pathways will gain in detail. In-depth exploration and establishment of these signaling pathways, including Trx upstream and downstream regulatory proteins, will be critical to advancing novel cancer therapeutics. Antioxid. Redox Signal. 38, 403-424.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zhengjia Zhao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | | | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, China
| |
Collapse
|
5
|
Takata N, Myburgh J, Botha A, Nomngongo PN. The importance and status of the micronutrient selenium in South Africa: a review. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2022; 44:3703-3723. [PMID: 34708333 DOI: 10.1007/s10653-021-01126-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 10/09/2021] [Indexed: 06/13/2023]
Abstract
Selenium (Se) is a vital micronutrient with widespread biological action but leads to toxicity when taken in excessive amounts. The biological benefits of Se are mainly derived from its presence in active sites of selenoproteins such as glutathione peroxidase (GPx). An enzyme whose role is to protect tissues against oxidative stress by catalysing the reduction of peroxidase responsible for various forms of cellular damage. The benefits of Se can be harvested when proper regulations of its intake are used. In South Africa, Se distribution in people's diets and animals are low with socio-economic factors and heterogeneous spread of Se in soil throughout the country playing a significant role. The possible causes of low Se in soils may be influenced by underlying geological material, climatic conditions, and anthropogenic activities. Sedimentary rock formations show higher Se concentrations compared to igneous and metamorphic rock formations. Higher Se concentrations in soils dominates in humid and sub-humid areas of South Africa. Furthermore, atmospheric acid deposition dramatically influences the availability of Se to plants. The studies reviewed in this article have shown that atomic absorption spectroscopy (AAS) is the most utilised analytical technique for total Se concentration determination in environmental samples and there is a lack of speciation data for Se concentrations. Shortcomings in Se studies have been identified, and the future research directions of Se in South Africa have been discussed.
Collapse
Affiliation(s)
- Nwabisa Takata
- Department of Chemical Sciences, University of Johannesburg, Doornfontein Campus, P.O. Box 17011, Doornfontein, 2028, South Africa
- National Metrology Institute of South Africa, CSIR Campus, Building 5, Meiring Naude Road, Brummeria, Pretoria, 0182, South Africa
| | - Jan Myburgh
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, 0110, South Africa
| | - Angelique Botha
- National Metrology Institute of South Africa, CSIR Campus, Building 5, Meiring Naude Road, Brummeria, Pretoria, 0182, South Africa
| | - Philiswa Nosizo Nomngongo
- Department of Chemical Sciences, University of Johannesburg, Doornfontein Campus, P.O. Box 17011, Doornfontein, 2028, South Africa.
- Department of Science and Innovation (DSI)/National Research Foundation (NRF) South African Research Chair (SARChI): Nanotechnology for Water, University of Johannesburg, Doornfontein, 2028, South Africa.
| |
Collapse
|
6
|
Quantitative Proteomic Analysis Reveals Yeast Cell Wall Products Influence the Serum Proteome Composition of Broiler Chickens. Int J Mol Sci 2022; 23:ijms231911844. [PMID: 36233150 PMCID: PMC9569515 DOI: 10.3390/ijms231911844] [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: 08/26/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
With an ever-growing market and continual financial pressures associated with the prohibition of antibiotic growth promoters, the poultry industry has had to rapidly develop non-antibiotic alternatives to increase production yields. A possible alternative is yeast and its derivatives, such as the yeast cell wall (YCW), which have been proposed to confer selected beneficial effects on the host animal. Here, the effect of YCW supplementation on the broiler chicken was investigated using a quantitative proteomic strategy, whereby serum was obtained from three groups of broilers fed with distinct YCW-based Gut Health Products (GHP) or a control basal diet. Development of a novel reagent enabled application of ProteoMiner™ technology for sample preparation and subsequent comparative quantitative proteomic analysis revealed proteins which showed a significant change in abundance (n = 167 individual proteins; p < 0.05); as well as proteins which were uniquely identified (n = 52) in, or absent (n = 37) from, GHP-fed treatment groups versus controls. An average of 7.1% of proteins showed changes in abundance with GHP supplementation. Several effects of these GHPs including immunostimulation (via elevated complement protein detection), potential alterations in the oxidative status of the animal (e.g., glutathione peroxidase and catalase), stimulation of metabolic processes (e.g., differential abundance of glyceraldehyde-3-phosphate dehydrogenase), as well as evidence of a possible hepatoprotective effect (attenuated levels of serum α-glutathione s-transferase) by one GHP feed supplement, were observed. It is proposed that specific protein detection may be indicative of GHP efficacy to stimulate broiler immune status, i.e., may be biomarkers of GHP efficacy. In summary, this work has developed a novel technology for the preparation of high dynamic range proteomic samples for LC-MS/MS analysis, is part of the growing area of livestock proteomics and, importantly, provides evidential support for beneficial effects that GHP supplementation has on the broiler chicken.
Collapse
|
7
|
Morão IFC, Lemos MFL, Félix R, Vieira S, Barata C, Novais SC. Stress response markers in the blood of São Tomé green sea turtles (Chelonia mydas) and their relation with accumulated metal levels. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 293:118490. [PMID: 34780755 DOI: 10.1016/j.envpol.2021.118490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 06/13/2023]
Abstract
Metals are persistent worldwide being harmful for diverse organisms and having complex and combined effects with other contaminants in the environment. Sea turtles accumulate these contaminants being considered good bioindicator species for marine pollution. However, very little is known on how this is affecting these charismatic animals. São Tomé and Príncipe archipelago harbours important green sea turtle (Chelonia mydas) nesting and feeding grounds. The main goal of this study was to determine metal and metalloid accumulation in the blood of females C. mydas nesting in São Tomé Island, and evaluate the possible impacts of this contamination by addressing molecular stress responses. Gene expression analysis was performed in blood targeting genes involved in detoxification/sequestration and metal transport (mt, mtf and fer), and in antioxidant and oxidative stress responses (cat, sod, gr, tdx, txrd, selp and gclc). Micronuclei analysis in blood was also addressed as a biomarker of genotoxicity. Present results showed significant correlations between different gene expressions with the metals evaluated. The best GLM models and significant relationships were found for mt expression, for which 78% of the variability was attributed to metal levels (Al, Cu, Fe, Hg, Pb and Zn), followed by micronuclei count (65% - Cr, Cu, Fe, Hg, Mn and Zn), tdx expression (52% - Cd, Fe, Mn, Pb and Se), and cat expression (52% - As, Fe, Se and Cd x Hg). Overall, this study demonstrates that these green sea turtles are trying to adapt to the oxidative stress and damage produced by metals through the increased expression of antioxidants and other protectors, which raises concerns about the impacts on these endangered organisms' fitness. Furthermore, promising biomarker candidates associated to metal stress were identified in this species that may be used in future biomonitoring studies using C. mydas' blood, allowing for a temporal follow-up of the organisms.
Collapse
Affiliation(s)
- Inês F C Morão
- MARE - Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, Portugal
| | - Marco F L Lemos
- MARE - Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, Portugal
| | - Rafael Félix
- MARE - Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, Portugal
| | - Sara Vieira
- Associação Programa Tatô, Avenida Marginal 12 de Julho, Cidade de São Tomé, São Tomé e Príncipe, Portugal
| | - Carlos Barata
- Environmental Chemistry Department, IDAEA-CSIC, Jordi Girona 18-26, 08034, Barcelona, Spain
| | - Sara C Novais
- MARE - Marine and Environmental Sciences Centre, ESTM, Politécnico de Leiria, Portugal.
| |
Collapse
|
8
|
Xu Y, Zuo W, Wang X, Zhang Q, Gan X, Tan N, Jia W, Liu J, Li Z, Zhou B, Zhao D, Xie Z, Tan Y, Zheng S, Liu C, Li H, Chen Z, Yang X, Huang Z. Deciphering the effects of PYCR1 on cell function and its associated mechanism in hepatocellular carcinoma. Int J Biol Sci 2021; 17:2223-2239. [PMID: 34239351 PMCID: PMC8241733 DOI: 10.7150/ijbs.58026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/16/2021] [Indexed: 12/28/2022] Open
Abstract
Overexpression of pyrroline-5-carboxylate reductase 1 (PYCR1) has been associated with the development of certain cancers; however, no studies have specifically examined the role of PYCR1 in hepatocellular carcinoma (HCC). Based on The Cancer Genome Atlas expression array and meta-analysis conducted using the Gene Expression Omnibus database, we determined that PYCR1 was upregulated in HCC compared to adjacent nontumor tissues (P < 0.05). These data were verified using quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry analysis. Additionally, patients with low PYCR1 expression showed a higher overall survival rate than patients with high PYCR1 expression. Furthermore, PYCR1 overexpression was associated with the female sex, higher levels of alpha-fetoprotein, advanced clinical stages (III and IV), and a younger age (< 45 years old). Silencing of PYCR1 inhibited cell proliferation, invasive migration, epithelial-mesenchymal transition, and metastatic properties in HCC in vitro and in vivo. Using RNA sequencing and bioinformatics tools for data-dependent network analysis, we found binary relationships among PYCR1 and its interacting proteins in defined pathway modules. These findings indicated that PYCR1 played a multifunctional role in coordinating a variety of biological pathways involved in cell communication, cell proliferation and growth, cell migration, a mitogen-activated protein kinase cascade, ion binding, etc. The structural characteristics of key pathway components and PYCR1-interacting proteins were evaluated by molecular docking, and hotspot analysis showed that better affinities between PYCR1 and its interacting molecules were associated with the presence of arginine in the binding site. Finally, a candidate regulatory microRNA, miR-2355-5p, for PYCR1 mRNA was discovered in HCC. Overall, our study suggests that PYCR1 plays a vital role in HCC pathogenesis and may potentially serve as a molecular target for HCC treatment.
Collapse
Affiliation(s)
- Yanzhen Xu
- Department of pathology, Affiliated hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Department of Pathology, Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, China
| | - Wenpu Zuo
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Medical Scientific Research Center, Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Xiao Wang
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Qinle Zhang
- Genetic and metabolic central laboratory, the maternal and children's health hospital of Guangxi, Nanning, 530000, Guangxi, China
| | - Xiang Gan
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Ning Tan
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Wenxian Jia
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Jiayi Liu
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhouquan Li
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Bo Zhou
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Dong Zhao
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhibin Xie
- Department of Urology, the Five Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Yanjun Tan
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Shengfeng Zheng
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Chengwu Liu
- Department of Pathophysiology, Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Hongtao Li
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhijian Chen
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Xiaoli Yang
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhaoquan Huang
- Department of pathology, Affiliated hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
- Department of Pathology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| |
Collapse
|
9
|
Ge J, Liu LL, Cui ZG, Talukder M, Lv MW, Li JY, Li JL. Comparative study on protective effect of different selenium sources against cadmium-induced nephrotoxicity via regulating the transcriptions of selenoproteome. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 215:112135. [PMID: 33780782 DOI: 10.1016/j.ecoenv.2021.112135] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
Cadmium (Cd) is a ubiquitous environmental pollutant, which mainly input to the aquatic environment through discharge of industrial and agricultural waste, can be a threat to human and animal health. Selenium (Se) possesses a beneficial role in protecting animals and ameliorating the toxic effects of Cd. However, the comparative antagonistic effects of different Se sources such as inorganic, organic Se and nano-form Se on Cd toxicity are still under-investigated. Hence, the purpose of this study was to evaluate the comparative of Se sources antagonism on Cd-induced nephrotoxicity via oxidative stress and selenoproteome transcription. In the present study, Cd-diet disturbed in the system balance of 5 trace elements (Zinc (Zn), copper (Cu), Iron (Fe), Se, Cd) and impaired renal function. Se sources, including nano- Se (NS), Se- yeast (SY), sodium selenite (SS) and mixed selenium (MS) significantly recovered the balance of 4 trace elements (Zn, Cu, Cd, Se) and renal impaired indexes (blood urea nitrogen (BUN) and creatinine (CREA)). Histological appearance of Cd-treated kidney indicated renal tubular epithelial vacuoles, particle degeneration and enlarged capsular space. Ultrastructure observation results illustrated that Cd-induced mitochondrial cristae reduction, membrane disappearance, and nuclear deformation. Treatment with Se sources, NS appeared a better impact on improving kidney tissues against the pathological alterations resulting from Cd administration. Meanwhile, NS reflected a significant impact on relieving Cd-induced kidney oxidative damage, and significantly restored the antioxidant defense system of the body. Our findings also showed NS ameliorated the Cd-induced downtrends expression of selenoproteome and selenoprotein synthesis related transcription factors. Overall, NS was the most effective Se source in avoiding of Cd cumulative toxicity, improving antioxidant capacity and regulating of selenoproteome transcriptome and selenoprotein synthesis related transcription factors expression, which contributes to ameliorate Cd-induced nephrotoxicity in chickens. These results demonstrated diet supplement with NS may prove to be an effective approach for alleviating Cd toxicity and minimizing Cd -induced health risk.
Collapse
Affiliation(s)
- Jing Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Li-Li Liu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, PR China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Science, University of Fukui, Fukui 910-1193, Japan
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Mei-Wei Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Yang Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
10
|
Zhang Z, Liu Q, Yang J, Yao H, Fan R, Cao C, Liu C, Zhang S, Lei X, Xu S. The proteomic profiling of multiple tissue damage in chickens for a selenium deficiency biomarker discovery. Food Funct 2020; 11:1312-1321. [PMID: 32022057 DOI: 10.1039/c9fo02861g] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past decades, substantial advances have been made in both the early diagnosis and accurate prognosis of numerous cancers because of the impressive development of novel proteomic strategies. Selenium (Se) is an essential trace element in humans and animals. Se deficiency could lead to Keshan disease in humans, mulberry heart disease in pigs and damage of tissues including cardiac injury, apoptosis in the liver, reduction in the immune responses in spleen and cerebral lesions in chickens. However, it is well know that plasma biomarkers are not specific and also show alterations in various diseases including those caused by Se deficiency. Therefore, new definition biomarkers are needed to improve disease surveillance and reduce unnecessary chicken losses due to Se deficiency. To identify new biomarkers for Se deficiency, we performed exploratory heart, liver, spleen, muscle, vein, and artery proteomic screens to further validate the biomarkers using Venn analysis, GO enrichment, heatmap analysis, and IPA analysis. Based on the bioinformatics methods mentioned above, we found that differentially expressed genes and proteins are enriched to the PI3K/AKT/mTOR signal pathway and insulin pathway. We further used western blot to detect the expression of proteins related to the two pathways. Results showed that the components of the PI3K/AKT/mTOR signal pathway were definitely decreased in heart, liver, spleen, muscle, vein and artery tissues in the Se deficient group. Expression IGF and IGFBP2 of the insulin pathway were differentially increased in the heart, liver, and spleen in Se deficient group samples and decreased in muscle and artery. In conclusion, 5 proteins, namely PI3K, AKT, mTOR, IGF, and IGFBP2, were differentially expressed, which could be potentially useful Se deficient biomarkers. In the present study, proteomic profiling was used to elucidate protein biomarkers that distinguished Se deficient samples from the controls, which might provide a new direction for the diagnosis and targeted treatment induced by Se deficiency in chickens.
Collapse
Affiliation(s)
- Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China. and Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Haidong Yao
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Ruifeng Fan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, P. R. China
| | - Changyu Cao
- College of Life and Science, Foshan University, Foshan, 528000, P. R. China
| | - Ci Liu
- College of Animal Technology, Shanxi Agricultural University, Jinzhong, 030600, P. R. China
| | - Sheng Zhang
- Institute of Biotechnology, Cornell University, Ithaca, NY 14583, USA
| | - Xingen Lei
- Department of Animal Science, Cornell University, Ithaca, NY 14583, USA
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China. and Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| |
Collapse
|
11
|
Yang J, Gong Y, Cai J, Liu Q, Zhang Y, Zheng Y, Yu D, Zhang Z. Dysfunction of thioredoxin triggers inflammation through activation of autophagy in chicken cardiomyocytes. Biofactors 2020; 46:579-590. [PMID: 32031748 DOI: 10.1002/biof.1625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022]
Abstract
Thioredoxin (Txn) is a hydrogen carrier protein and exists widely in organism. Txn deficiency implicates cardiomyocytes injury has been proven. However, the exact mechanism remains unclear. To understand the mechanistic response of cardiomyocytes subsequent to Txn suppression, we established the model of Txn dysfunction by employing gene interference technology (siRNA) and Txn inhibitor (PX-12) in cardiomyocytes. We detected the ROS levels, inflammation factors, and key proteins in the autophagy and apoptosis. In addition, heat map was used for further analysis. Our results revealed that Txn dysfunction increased the release of ROS and induced activation of autophagy via upregulation of Becline-1, LC3-1, 2, which further regulated the inflammatory response, meanwhile, Txn silence inhibited apoptosis in chicken cardiomyocytes through Caspase-3 inhibition. Altogether we concluded that Txn-deficient chicken cardiomyocytes experienced autophagy, which caused severe inflammatory reactions and resulting in damage to cardiomyocytes.
Collapse
Affiliation(s)
- Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yafan Gong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Jingzeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yuan Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yingying Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Dahai Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
12
|
Li P, Xu Y, Zhang Q, Li Y, Jia W, Wang X, Xie Z, Liu J, Zhao D, Shao M, Chen S, Mo N, Jiang Z, Li L, Liu R, Huang W, Chang L, Chen S, Li H, Zuo W, Li J, Zhang R, Yang X. Evaluating the role of RAD52 and its interactors as novel potential molecular targets for hepatocellular carcinoma. Cancer Cell Int 2019; 19:279. [PMID: 31719794 PMCID: PMC6836504 DOI: 10.1186/s12935-019-0996-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Background Radiation sensitive 52 (RAD52) is an important protein that mediates DNA repair in tumors. However, little is known about the impact of RAD52 on hepatocellular carcinoma (HCC). We investigated the expression of RAD52 and its values in HCC. Some proteins that might be coordinated with RAD52 in HCC were also analyzed. Methods Global RAD52 mRNA levels in HCC were assessed using The Cancer Genome Atlas (TCGA) database. RAD52 expression was analyzed in 70 HCC tissues and adjacent tissues by quantitative real-time PCR (qRT-PCR), Western blotting and immunohistochemistry. The effect of over-expressed RAD52 in Huh7 HCC cells was investigated. The String database was then used to perform enrichment and functional analysis of RAD52 and its interactome. Cytoscape software was used to create a protein–protein interaction network. Molecular interaction studies with RAD52 and its interactome were performed using the molecular docking tools in Hex8.0.0. Finally, these DNA repair proteins, which interact with RAD52, were also analyzed using the TCGA dataset and were detected by qRT-PCR. Based on the TCGA database, algorithms combining ROC between RAD52 and RAD52 interactors were used to diagnose HCC by binary logistic regression. Results In TCGA, upregulated RAD52 related to gender was obtained in HCC. The area under the receiver operating characteristic curve (AUC) of RAD52 was 0.704. The results of overall survival (OS) and recurrence-free survival (RFS) indicated no difference in the prognosis between patients with high and low RAD52 gene expression. We validated that RAD52 expression was increased at the mRNA and protein levels in Chinese HCC tissues compared with adjacent tissues. Higher RAD52 was associated with older age, without correlation with other clinicopathological factors. In vitro, over-expressed RAD52 significantly promoted the proliferation and migration of Huh7 cells. Furthermore, RAD52 interactors (radiation sensitive 51, RAD51; X-ray repair cross complementing 6, XRCC6; Cofilin, CFL1) were also increased in HCC and participated in some biological processes with RAD52. Protein structure analysis showed that RAD52–RAD51 had the firmest binding structure with the lowest E-total energy (− 1120.5 kcal/mol) among the RAD52–RAD51, RAD52–CFL1, and RAD52–XRCC6 complexes. An algorithm combining ROC between RAD52 and its interactome indicated a greater specificity and sensitivity for HCC screening. Conclusions Overall, our study suggested that RAD52 plays a vital role in HCC pathogenesis and serves as a potential molecular target for HCC diagnosis and treatment. This study’s findings regarding the multigene prediction and diagnosis of HCC are valuable.
Collapse
Affiliation(s)
- Ping Li
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,4College & Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi China.,5Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - YanZhen Xu
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,8Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi China
| | - Qinle Zhang
- Genetic and Metabolic Central Laboratory, The Maternal and Children Health Hospital of Guangxi, Guangxi, China
| | - Yu Li
- Medical Science Laboratory at Liuzhou Worker's Hospital, Liuzhou, Guangxi China
| | - Wenxian Jia
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,6College of Pharmacy, Guangxi Medical University, Nanning, Guangxi China
| | - Xiao Wang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Zhibin Xie
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China
| | - Jiayi Liu
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,4College & Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi China.,5Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - Dong Zhao
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China
| | - Mengnan Shao
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China
| | - Suixia Chen
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,8Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi China
| | - Nanfang Mo
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Zhiwen Jiang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Liuyan Li
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Run Liu
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Wanying Huang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Li Chang
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Siyu Chen
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Hongtao Li
- 2Scientific Research Center, Guilin Medical University, Guilin, Guangxi China
| | - Wenpu Zuo
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | - Jiaquan Li
- 3Medical Scientific Research Center, Guangxi Medical University, Nanning, Guangxi China
| | | | - Xiaoli Yang
- 1Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi China.,2Scientific Research Center, Guilin Medical University, Guilin, Guangxi China
| |
Collapse
|
13
|
Sattar H, Yang J, Zhao X, Cai J, Liu Q, Ishfaq M, Yang Z, Chen M, Zhang Z, Xu S. Selenoprotein-U (SelU) knockdown triggers autophagy through PI3K-Akt-mTOR pathway inhibition in rooster Sertoli cells. Metallomics 2019; 10:929-940. [PMID: 29961786 DOI: 10.1039/c8mt00090e] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Selenium (Se) is a major component of male reproduction which exerts its effects via selenoproteins. Selenoprotein U (SelU), a newly identified protein, is expressed highly in eukaryotes and possesses a conserved motif similar to that existing in other thiol-dependent redox regulating selenoproteins; however its function is unknown. To investigate the role of SelU in testis autophagic and/or apoptosis cell death mechanisms, we established a Sertoli cell (SC) model isolated from 45 day old layer roosters. Small interfering RNA (siRNA) technology was used to develop SelU-knockdown (SelU-KD) and normal (N) SC models. Consequent to transfection, electron microscopy, qPCR, and western blot were performed. The results show that the mRNA and proteins of autophagy and anti-apoptosis genes increased while that of anti-autophagic mammalian target of rapamycin (mTOR) and pro-apoptosis genes decreased significantly in SelU-KD in contrast to N cells. Simultaneously, in contrast to N cells the expression of phosphoinositide-3-kinase (PI3K) and protein kinase B (PKB/Akt) both at the mRNA and protein levels decreased significantly in SelU-KD cells. In-addition, SelU depletion altered the expression of regulatory factors and increased the mRNA of TSC (tuberous sclerosis complex) genes as compared to N cells. Extensive autophagosome formation and lysosome degradation with an intact cytoskeleton were observed in SelU-KD cells. Our data indicate that SelU deprivation elicits autophagy and reduces the expression of important growth factors in SCs by disrupting the PI3K-Akt-mTOR signaling pathway. However SelU attenuation did not induce apoptosis in rooster SCs. Taken together, we conclude that SelU is essential for the survival and normal functioning of SCs.
Collapse
Affiliation(s)
- Hamid Sattar
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Regulation and function of avian selenogenome. Biochim Biophys Acta Gen Subj 2018; 1862:2473-2479. [PMID: 29627451 DOI: 10.1016/j.bbagen.2018.03.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Selenium (Se) is an essential micronutrient required by avian species. Dietary Se/vitamin E deficiency induces three classical diseases in chicks: exudative diathesis, nutritional pancreatic atrophy, and nutritional muscular dystrophy. SCOPE OF REVIEW This review is to summarize and analyze the evolution, regulation, and function of avian selenogenome and selenoproteome and their relationship with the three classical Se/vitamin E deficiency diseases. MAJOR CONCLUSIONS There are 24 selenoproteins confirmed in chicks, with two avian-specific members (SELENOU and SELENOP2) and two missing mammalian members (GPX6 and SELENOV). There are two forms of SELENOP containing 1 or 13 selenocysteine residues. In addition, a Gallus gallus gene was conjectured to be the counterpart of the human SEPHS2. Expression of selenoprotein genes in the liver, pancreas, and muscle of chicks seemed to be highly responsive to dietary Se changes. Pathogeneses of the Se/vitamin E deficient diseases in the chicks were likely produced by missing functions of selected selenoproteins in regulating cellular and tissue redox balance and inhibiting oxidative/reductive stress-induced cell death. GENERAL SIGNIFICANCE Gene knockout models, similar to those of rodents, will help characterize the precise functions of avian selenoproteins and their comparisons with those of mammalian species.
Collapse
|