1
|
Ying W, Yunqi Z, Deji L, Jian K, Fusheng Q. Follicular fluid HD-sevs-mir-128-3p is a key molecule in regulating bovine granulosa cells autophagy. Theriogenology 2024; 226:263-276. [PMID: 38954995 DOI: 10.1016/j.theriogenology.2024.06.022] [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] [Received: 03/20/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
Follicular fluid (FF) is rich in extracellular vesicles (EVs). EVs carries a variety of miRNA involved in regulating follicular development, the function of cells in follicles, primordial follicular formation, follicular recruitment and selection, follicular atresia, oocyte communication, granulosa cells (GCs) function and luteinization and other biological processes of follicular development. Previous studies in our laboratory have shown that bovine follicular fluid (bFF) high density-small extracellular vesicles (HD-sEVs)-miRNA was enriched in autophagy-related pathways. However, the mechanism of bFF EVs carrying miRNA regulating GCs autophagy is not clear. Thus, this study carried out a series of studies on the previous HD-sEVs sequencing data and miR-128-3p contained in bFF HD-sEVs. A total of 38 differentially expressed genes were detected by RNA-Seq after overexpression of miR-128-3p in bovine GCs (bGCs). Through cell transfection, Western blot (WB) and Immunofluorescence (IF), it was proved that overexpression of miR-128-3p could promote the expression of LC3 (microtubule-associated protein I light chain 3), inhibit p62, promote the number of autophagosome, promote the formation of autophagy lysosome and autophagy flow, and activate bGCs autophagy. MiR-128-3p inhibitor significantly inhibited the expression of LC3 and monodansylcadaverine (MDC) in bGCs, and promoted the expression of autophagy substrate p62, indicating that HD-sEVs-miR-128-3p could activate bGCs autophagy. In addition, through double luciferase assay, bioinformatics analysis, WB and RT-qPCR, it was concluded that bFF HD-sEVs-miR-128-3p could target TFEB (transcription factor EB) and FoxO4 (Forkhead box O4) and activate GCs autophagy.
Collapse
Affiliation(s)
- Wang Ying
- Chongqing Key Laboratory of Forage &Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhao Yunqi
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Luan Deji
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Kang Jian
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Quan Fusheng
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
2
|
Wang S, Pang Y, Wang L, Wang Q, Chen Z, Li C, Li F, Zhang G, Wang X, Gao S, Xu X. Differences in Lipid Metabolism between the Perirenal Adipose Tissue of Chinese Simmental Cattle and Angus Cattle ( Bos taurus) Based on Metabolomics Analysis. Animals (Basel) 2024; 14:2536. [PMID: 39272322 PMCID: PMC11394394 DOI: 10.3390/ani14172536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
The aim of this experiment was to investigate the differences in metabolites in perirenal fat (PF) between Chinese Simmental cattle and Angus cattle. Six healthy 18-month-old male Angus cattle and Chinese Simmental cattle were selected, and the perirenal adipose tissue was collected after slaughtering. HE staining, a triglyceride assay kit, and liquid chromatography-tandem mass spectrometry (LC-MS/MS) technology were used to compare and analyze the differences in the cell morphology, lipid accumulation, and metabolites of the two types of PF. The results showed that the PF of Angus cattle had a larger cell area and stronger lipid deposition ability than that of Simmental cattle. A total of 567 metabolites were detected by LC-MS/MS technology, of which 119 were significantly upregulated in Angus cattle PF and 129 were significantly upregulated in Simmental cattle PF. Differential metabolites were enriched in pathways such as fatty acid biosynthesis, polyunsaturated fatty acid biosynthesis, regulation of adipocyte lipolysis, and oxidative phosphorylation. Finally, 12 metabolites that may cause phenotypic differences between the two types of perirenal adipose tissue were screened out from these pathways. This study has preliminarily screened out biomarkers that may affect lipid metabolism in PF, providing basic data for the further exploration of the metabolic characteristics of PF.
Collapse
Affiliation(s)
- Siyuan Wang
- Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot 137400, China
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Yue Pang
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Lixiang Wang
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Qi Wang
- Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot 137400, China
| | - Zhongling Chen
- Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot 137400, China
| | - Chengjiao Li
- Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot 137400, China
| | - Fengjiao Li
- Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot 137400, China
| | - Guoxi Zhang
- Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot 137400, China
| | - Xiaoying Wang
- Tongliao Animal Husbandry Development Center, Tongliao 028000, China
| | - Shuxin Gao
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Xingjian Xu
- Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot 137400, China
| |
Collapse
|
3
|
Ru M, Liang H, Ruan J, Haji RA, Cui Y, Yin C, Wei Q, Huang J. Chicken ovarian follicular atresia: interaction network at organic, cellular, and molecular levels. Poult Sci 2024; 103:103893. [PMID: 38870615 PMCID: PMC11225904 DOI: 10.1016/j.psj.2024.103893] [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] [Received: 01/26/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Most of follicles undergo a degenerative process called follicular atresia. This process directly affects the egg production of laying hens and is regulated by external and internal factors. External factors primarily include nutrition and environmental factors. In follicular atresia, internal factors are predominantly regulated at 3 levels; organic, cellular and molecular levels. At the organic level, the hypothalamic-pituitary-ovary (HPO) axis plays an essential role in controlling follicular development. At the cellular level, gonadotropins and cytokines, as well as estrogens, bind to their receptors and activate different signaling pathways, thereby suppressing follicular atresia. By contrast, oxidative stress induces follicular atresia by increasing ROS levels. At the molecular level, granulosa cell (GC) apoptosis is not the only factor triggering follicular atresia. Autophagy is also known to give rise to atresia. Epigenetics also plays a pivotal role in regulating gene expression in processes that seem to be related to follicular atresia, such as apoptosis, autophagy, proliferation, and steroidogenesis. Among these processes, the miRNA regulation mechanism is well-studied. The current review focuses on factors that regulate follicular atresia at organic, cellular and molecular levels and evaluates the interaction network among these levels. Additionally, this review summarizes atretic follicle characteristics, in vitro modeling methods, and factors preventing follicular atresia in laying hens.
Collapse
Affiliation(s)
- Meng Ru
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Haiping Liang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Jiming Ruan
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Ramlat Ali Haji
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Yong Cui
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Chao Yin
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Qing Wei
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Jianzhen Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China.
| |
Collapse
|
4
|
Li XW, Guo K, Wang CC, Yang Y, Li W, Talukder M, Li XN, Li JL. The Nrf2/ARE pathway as a potential target to ameliorate atrazine-induced endocrine disruption in granulosa cells. Poult Sci 2024; 103:103730. [PMID: 38631229 PMCID: PMC11040167 DOI: 10.1016/j.psj.2024.103730] [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] [Received: 01/18/2024] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/19/2024] Open
Abstract
Atrazine (ATR) is widely used worldwide as a commercial herbicide, Diaminochlorotriazine (DACT) is the main metabolite of ATR in the organism. Both of them disrupt the production of steroids and induce abnormal reproductive development. The granulosa cells (GCs) are important for growth and reproduction of animals. However, the toxicity of ATR on the GCs of birds is not well clarified. To evaluate the effect of the environmental pollutant ATR on bird GCs. The quail GCs were allotted into 7 groups, C (The medium of M199), A20 (20 µM ATR), A100 (100 µM ATR), A250 (250 µM ATR), D20 (20 µM DACT), D100 (100 µM DACT) and D200 (200 µM DACT). The results demonstrated that ATR reduced the viability of GCs, disrupted mitochondrial structure (including mitochondrial cristae fragmentation and the mitochondrial morphology disappearance) and decreased mitochondrial membrane potential. Meanwhile, ATR interfered with the expression of key factors in the steroid synthesis pathway, inducing the secretion of the sex hormones E2 and P in GCs. which in turn induced apoptosis. Furthermore, the Nrf2/ARE pathway as a potential target to ameliorate ATR-induced endocrine disruption in GCs for proper reproductive functions. Our research provides a new perspective for understanding the effects of ATR on reproductive functions in birds.
Collapse
Affiliation(s)
- Xiao-Wei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Kai Guo
- Chifeng Agriculture and Animal Husbandry Comprehensive Administrative Law Enforcement Detachment, Chifeng City, Inner Mongolia, 024000, China
| | - Chi-Chiu Wang
- Department of Obstetrics & Gynaecology; Li Ka Shing Institute of Health Sciences; School of Biomedical Sciences; and The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Wei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh
| | - Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Department of Obstetrics & Gynaecology; Li Ka Shing Institute of Health Sciences; School of Biomedical Sciences; and The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
5
|
Liu H, Ryu D, Hwang S, Lee SS. Therapies for Cirrhotic Cardiomyopathy: Current Perspectives and Future Possibilities. Int J Mol Sci 2024; 25:5849. [PMID: 38892040 PMCID: PMC11173074 DOI: 10.3390/ijms25115849] [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] [Received: 05/03/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Cirrhotic cardiomyopathy (CCM) is defined as cardiac dysfunction associated with cirrhosis in the absence of pre-existing heart disease. CCM manifests as the enlargement of cardiac chambers, attenuated systolic and diastolic contractile responses to stress stimuli, and repolarization changes. CCM significantly contributes to mortality and morbidity in patients who undergo liver transplantation and contributes to the pathogenesis of hepatorenal syndrome/acute kidney injury. There is currently no specific treatment. The traditional management for non-cirrhotic cardiomyopathies, such as vasodilators or diuretics, is not applicable because an important feature of cirrhosis is decreased systemic vascular resistance; therefore, vasodilators further worsen the peripheral vasodilatation and hypotension. Long-term diuretic use may cause electrolyte imbalances and potentially renal injury. The heart of the cirrhotic patient is insensitive to cardiac glycosides. Therefore, these types of medications are not useful in patients with CCM. Exploring the therapeutic strategies of CCM is of the utmost importance. The present review summarizes the possible treatment of CCM. We detail the current status of non-selective beta-blockers (NSBBs) in the management of cirrhotic patients and discuss the controversies surrounding NSBBs in clinical practice. Other possible therapeutic agents include drugs with antioxidant, anti-inflammatory, and anti-apoptotic functions; such effects may have potential clinical application. These drugs currently are mainly based on animal studies and include statins, taurine, spermidine, galectin inhibitors, albumin, and direct antioxidants. We conclude with speculations on the future research directions in CCM treatment.
Collapse
Affiliation(s)
- Hongqun Liu
- Liver Unit, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (H.L.); (D.R.); (S.H.)
| | - Daegon Ryu
- Liver Unit, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (H.L.); (D.R.); (S.H.)
- Division of Gastroenterology, Yangsan Hospital, Pusan National University School of Medicine, Pusan 46033, Republic of Korea
| | - Sangyoun Hwang
- Liver Unit, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (H.L.); (D.R.); (S.H.)
- Department of Internal Medicine, Dongnam Institute of Radiological and Medical Sciences, Pusan 46033, Republic of Korea
| | - Samuel S. Lee
- Liver Unit, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (H.L.); (D.R.); (S.H.)
| |
Collapse
|
6
|
Gao Z, He W, Liu Y, Gao Y, Fan W, Luo Y, Shi X, Song S. Perinatal bisphenol S exposure exacerbates the oxidative burden and apoptosis in neonatal ovaries by suppressing the mTOR/autophagy axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 349:123939. [PMID: 38593938 DOI: 10.1016/j.envpol.2024.123939] [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: 02/03/2024] [Revised: 04/05/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
Bisphenol S (BPS) is an emerging environmental endocrine disruptor capable of crossing the placental barrier, resulting in widespread exposure to pregnant women due to its extensive usage. However, the impact of perinatal maternal exposure to BPS on reproductive health in offspring and the underlying molecular mechanism remain underexplored. In this study, gestational ICR mice were provided with drinking water containing 3.33 mg/L BPS to mimic possible human exposure in some countries. Results demonstrated that BPS accelerated the breakdown of germ-cell cysts and the assembly of primordial follicles in neonates, leading to oocyte over-loss. Furthermore, the expression levels of folliculogenesis-related genes (Kit, Nobox, Gdf9, Sohlh2, Kitl, Bmp15, Lhx8, Figla, and Tgfb1) decreased, thus compromising oocyte quality and disrupting early folliculogenesis dynamics. BPS also disrupted other aspects of offspring reproduction, including advancing puberty onset, disrupting the estrus cycle, and impairing fertility. Further investigation found that BPS exposure inhibited the activities and expression levels of antioxidant-related enzymes in neonatal ovaries, leading to the substantial accumulation of MDA and ROS. The increased oxidative burden exacerbated the intracellular apoptotic signaling, manifested by increased expression levels of pro-apoptotic markers (Bax, Caspase 3, and Caspase 9) and decreased expression levels of anti-apoptotic marker (Bcl2). Concurrently, BPS inhibited autophagy by increasing p-mTOR/mTOR and decreasing p-ULK1/ULK1, subsequently down-regulating autophagy flux-related biomarkers (LC3b/LC3a and Beclin-1) and impeding the degradation of autophagy substrate p62. However, the imbalanced crosstalk between autophagy, apoptosis and oxidative stress homeostasis was restored after rapamycin treatment. Collectively, the findings demonstrated that BPS exposure induced reproductive disorders in offspring by perturbing the mTOR/autophagy axis, and such autophagic dysfunction exacerbated redox imbalance and promoted excessive apoptosis. These results provide novel mechanistic insights into the role of autophagy in mitigating BPS-induced intergenerational reproductive dysfunction.
Collapse
Affiliation(s)
- Zhangshan Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Wanqiu He
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Yapei Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Yixin Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Yan Luo
- Administration for Market Regulation of Guangdong Province Key Laboratory of Supervision for Edible Agricultural Products, Shenzhen Centre of Inspection and Testing for Agricultural Products, Shenzhen, 518000, China
| | - Xizhi Shi
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
7
|
Han T, Sun Z, Zhang H, Zhao Y, Jiao A, Gao Q. Ursolic acid alleviates meiotic abnormalities induced by 3-nitropropionic acid in mouse oocytes. Toxicol Appl Pharmacol 2024; 485:116910. [PMID: 38521372 DOI: 10.1016/j.taap.2024.116910] [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] [Received: 12/12/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
3-nitropropionic acid (3-NPA), a toxic metabolite produced by mold, is mainly found in moldy sugarcane. 3-NPA inhibits the activity of succinate dehydrogenase that can induce oxidative stress injury in cells, reduce ATP production and induce oxidative stress in mouse ovaries to cause reproductive disorders. Ursolic acid (UA) has a variety of biological activities and is a pentacyclic triterpene compound found in many plants. This experiment aimed to investigate the cytotoxicity of 3-NPA during mouse oocyte in vitro maturation and the protective effects of UA on oocytes challenged with 3-NPA. The results showed that UA could alleviate 3-NPA-induced oocyte meiotic maturation failure. Specifically, 3-NPA induced a decrease in the first polar body extrusion rate of oocytes, abnormal distribution of cortical granules, and an increase in the proportion of spindle abnormalities. In addition, 3-NPA caused mitochondrial dysfunction and induced oxidative stress, including decreases in the GSH, mitochondrial membrane potential and ATP levels, and increases in the ROS levels, and these effects led to apoptosis and autophagy. The addition of UA could significantly improve the adverse effects caused by 3-NPA. In general, our data show that 3-NPA affects the normal development of oocytes during the in vitro culture, and the addition of UA can effectively repair the damage caused by 3-NPA to oocytes.
Collapse
Affiliation(s)
- Tiancang Han
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Engineering Research Center of Yanbian Yellow Cattle Resources Reservation, China; Yanbian University, Yanji 133002, China
| | - Zhaoyang Sun
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Engineering Research Center of Yanbian Yellow Cattle Resources Reservation, China; Yanbian University, Yanji 133002, China
| | - Hongbo Zhang
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Engineering Research Center of Yanbian Yellow Cattle Resources Reservation, China; Yanbian University, Yanji 133002, China
| | - Yuhan Zhao
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Engineering Research Center of Yanbian Yellow Cattle Resources Reservation, China; Yanbian University, Yanji 133002, China
| | - Anhui Jiao
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Engineering Research Center of Yanbian Yellow Cattle Resources Reservation, China; Yanbian University, Yanji 133002, China
| | - Qingshan Gao
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji 133002, China; Jilin Engineering Research Center of Yanbian Yellow Cattle Resources Reservation, China; Yanbian University, Yanji 133002, China.
| |
Collapse
|
8
|
Jiang D, Ji C, Zhou X, Wang Z, Sun Q, Wang X, An X, Ling W, Kang B. Pathway analysis of spermidine anti-oxidative stress and inducing autophagy in granulosa cells of Sichuan white geese. Theriogenology 2024; 215:290-301. [PMID: 38118229 DOI: 10.1016/j.theriogenology.2023.12.020] [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] [Received: 08/25/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/22/2023]
Abstract
Spermidine, a natural polyamine, has been proven antioxidant function, but its pathway and mechanism of action remain unclear. Based on the oxidative stress model by 3-nitropropionic acid (3-NPA), the study explored the pathways by spermidine to rescue oxidative stress via autophagic process in goose granulosa cells by RNA-seq and RNA interference. In transcriptional regulation, in addition to KEGG pathways related to cell proliferation and differentiation, lots of KEGG pathways associated with inflammation, metabolism, and signaling were also significantly enriched in 3-NPA vs. 3-NPA + spermidine treatments. Six key genes (JUN, CD44, KITLG, RND2, BMP4 and KALRN) involved in spermidine-mediated anti-oxidative stress were screened. Furthermore, the experimental results showed that spermidine (80 μmol/L) significantly increased autophagic gene expression in goose granulosa cells, while EP300-siRNA or MAP1S-siRNA also significantly increased autophagic process. The autophagic gene expressions were no difference between EP300-siRNA and EP300-siRNA + spermidine treatments, although spermidine significantly increased autophagic process of granulosa cells compared to MAP1S-siRNA alone. In addition, inhibition of mTOR pathway significantly increased autophagic gene expression, which was further enhanced by spermidine in combined with mTOR inhibitor. These results suggest that spermidine can alleviate oxidative stress by inducing autophagy regulated by EP300, MAP1S and mTOR as well as regulating other independent gene expressions in goose granulosa cells.
Collapse
Affiliation(s)
- Dongmei Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Chengweng Ji
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Xuemin Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Zelong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Qian Sun
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Xin Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Xiaoguang An
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Weikang Ling
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Bo Kang
- State Key Laboratory of Swine and Poultry Breeding Industry, Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
9
|
Liu H, Nguyen HH, Hwang SY, Lee SS. Oxidative Mechanisms and Cardiovascular Abnormalities of Cirrhosis and Portal Hypertension. Int J Mol Sci 2023; 24:16805. [PMID: 38069125 PMCID: PMC10706054 DOI: 10.3390/ijms242316805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/19/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
In patients with portal hypertension, there are many complications including cardiovascular abnormalities, hepatorenal syndrome, ascites, variceal bleeding, and hepatic encephalopathy. The underlying mechanisms are not yet completely clarified. It is well known that portal hypertension causes mesenteric congestion which produces reactive oxygen species (ROS). ROS has been associated with intestinal mucosal injury, increased intestinal permeability, enhanced gut bacterial overgrowth, and translocation; all these changes result in increased endotoxin and inflammation. Portal hypertension also results in the development of collateral circulation and reduces liver mass resulting in an overall increase in endotoxin/bacteria bypassing detoxication and immune clearance in the liver. Endotoxemia can in turn aggravate oxidative stress and inflammation, leading to a cycle of gut barrier dysfunction → endotoxemia → organ injury. The phenotype of cardiovascular abnormalities includes hyperdynamic circulation and cirrhotic cardiomyopathy. Oxidative stress is often accompanied by inflammation; thus, blocking oxidative stress can minimize the systemic inflammatory response and alleviate the severity of cardiovascular diseases. The present review aims to elucidate the role of oxidative stress in cirrhosis-associated cardiovascular abnormalities and discusses possible therapeutic effects of antioxidants on cardiovascular complications of cirrhosis including hyperdynamic circulation, cirrhotic cardiomyopathy, and hepatorenal syndrome.
Collapse
Affiliation(s)
| | | | | | - Samuel S. Lee
- Liver Unit, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada (H.H.N.); (S.Y.H.)
| |
Collapse
|
10
|
Kang B, Wang X, An X, Ji C, Ling W, Qi Y, Li S, Jiang D. Polyamines in Ovarian Aging and Disease. Int J Mol Sci 2023; 24:15330. [PMID: 37895010 PMCID: PMC10607840 DOI: 10.3390/ijms242015330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Ovarian aging and disease-related decline in fertility are challenging medical and economic issues with an increasing prevalence. Polyamines are a class of polycationic alkylamines widely distributed in mammals. They are small molecules essential for cell growth and development. Polyamines alleviate ovarian aging through various biological processes, including reproductive hormone synthesis, cell metabolism, programmed cell death, etc. However, an abnormal increase in polyamine levels can lead to ovarian damage and promote the development of ovarian disease. Therefore, polyamines have long been considered potential therapeutic targets for aging and disease, but their regulatory roles in the ovary deserve further investigation. This review discusses the mechanisms by which polyamines ameliorate human ovarian aging and disease through different biological processes, such as autophagy and oxidative stress, to develop safe and effective polyamine targeted therapy strategies for ovarian aging and the diseases.
Collapse
Affiliation(s)
- Bo Kang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoguang An
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Chengweng Ji
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Weikang Ling
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuxin Qi
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shuo Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Dongmei Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.W.); (X.A.); (C.J.); (W.L.); (Y.Q.); (S.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|