1
|
Feng X, Feng Q, Abbas Raza SH, Li F, Ma Y. Identification of key factors causing ketosis in dairy cows with low feed intake. Anim Biotechnol 2025; 36:2487089. [PMID: 40184169 DOI: 10.1080/10495398.2025.2487089] [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: 12/16/2024] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Ketosis is a common metabolic disease in high-yield dairy cows. Key genes affecting ketosis need to be further explored by new methods. The gene expression profiling and clinical data of GSE92398, GSE104079, and GSE4304 were obtained from the gene expression omnibus (GEO) database. Core modules and genes associated with RFI (residual feed intake) and ADF (alternate day fasting) were identified by weighted gene co-expression network analysis (WGCNA). Subsequently, the key genes related to ketosis and RFI were determined by protein-protein interaction (PPI) networks, ROC curves, functional enrichment, and differential expression analysis, respectively. The results showed that the genes of ACACA, ELOVL6 and XPO7 could be used as regulators of ketosis induced by low feed intake in dairy cows. At the same time, three genes (HRFI, STAT3 and IFNAR1) were retained as additional RFI biomarkers that could be considered. We identified three key factors as candidate genes and biomarkers of ketosis and RFI, respectively. These factors may provide a theoretical basis for targeted therapy of ketosis in dairy cows.
Collapse
Affiliation(s)
- Xue Feng
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Qi Feng
- School of Computer and Information Engineering, Henan University, Kaifeng, China
| | - Sayed Haidar Abbas Raza
- Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, China
| | - Fen Li
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Yun Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
2
|
Zhao C, Xu R, Xin S, Jiang B, Feng S, Wang X, Xia C. AMPKα alleviates the inhibitory effect of NEFA on the function of bovine follicular granulosa cells cultured in vitro. Anim Reprod Sci 2025; 276:107831. [PMID: 40164035 DOI: 10.1016/j.anireprosci.2025.107831] [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: 12/09/2024] [Revised: 03/16/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
High levels of non-esterified fatty acids (NEFA) in cows with subclinical ketosis (SCK) impair postpartum follicular development and disrupt estrus. The precise mechanism through which NEFA impacts the functionality of bovine follicular cells remains elusive. An in vivo experiment was conducted to compare SCK cows without estrus (SCK-E, n = 6) with healthy cows in estrus (C-E, n = 6). In the vitro test, bovine granulosa cells (GCs) were exposed to 0.4 mM NEFA. Notably, the SCK-E group exhibited an elevated ratio of phosphorylated adenosine 5'-monophosphate-activated protein kinase α (AMPKα) to total AMPKα in both liver and ovarian tissues, compared to the C-E group. NEFA treatment of GCs adversely affected steroid hormone synthesis, suppressed the expression of cyclin and proteins crucial for steroid synthesis, and triggered cell apoptosis, thereby inhibiting cell proliferation. Furthermore, it led to a decline in cell mitochondrial membrane potential and an increase in reactive oxygen species production, ultimately causing cellular damage. Subsequently, GCs were co-cultured with adenovirus (ad-AMPKα-siRNA) and NEFA (0.4 mM). Inhibiting AMPKα further exacerbated the detrimental effects of NEFA on steroid hormone synthesis, cell apoptosis, cell proliferation, and mitochondrial function in GCs. Furthermore, upon inhibiting AMPKα, a reduction was observed in both mRNA and protein levels of acetyl-CoA carboxylase 1, accompanied by an elevation in the levels of carnitine palmitoyltransferase-1. These findings suggest that AMPKα becomes activated in SCK cows experiencing elevated NEFA levels, and that AMPKα has the potential to mitigate the detrimental effects of NEFA on GCs function in vitro.
Collapse
Affiliation(s)
- Chang Zhao
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China.
| | - Ruru Xu
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China
| | - Shuzhen Xin
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China
| | - Benzheng Jiang
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China
| | - Shibin Feng
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China
| | - Xichun Wang
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China.
| | - Cheng Xia
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China.
| |
Collapse
|
3
|
Kong F, Wang S, Zhang Y, Li C, Dai D, Wang Y, Cao Z, Yang H, Shengli Li, Wei Wang. Alanine Derived from Ruminococcus_E bovis Alleviates Energy Metabolic Disorders during the Peripartum Period by Providing Glucogenic Precursors. RESEARCH (WASHINGTON, D.C.) 2025; 8:0682. [PMID: 40290137 PMCID: PMC12022398 DOI: 10.34133/research.0682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
Peripartum dairy cows commonly experience energy metabolism disorders, which lead to passive culling of postpartum cows and a decrease in milk quality. By using ketosis peripartum dairy cows as a model, this study aims to elucidate the metabolic mechanism of peripartum cows and provide a novel way for managing energy metabolic disorders. From a cohort of 211 cows, we integrated multi-omics data (metagenomics, metabolomics, and transcriptomics) to identify key microbes and then utilized an in vitro rumen fermentation simulation system and ketogenic hepatic cells to validate the potential mechanisms and the effects of postbiotics derived from key microbes. Postpartum cows with metabolic disorders compensate for glucose deficiency through mobilizing muscle proteins, which leads to marked decreases in milk protein content. Concurrently, these cows experience rumen microbiota disturbance, with marked decreases in the concentrations of volatile fatty acids and microbial protein, and the deficiency of alanine (Ala) in microbial protein is correlated with the metabolic disorder phenotype. Metagenomic binning and in vitro fermentation assays reveal that Ruminococcus_E bovis (MAG 189) is enriched in amino acid biosynthesis functions and responsible for Ala synthesis. Furthermore, transcriptomic and metabolomic analyses of the liver in metabolic disorder cows also show impaired amino acid metabolism. Supplementation with Ala can alleviate ketogenesis in liver cell models by activating the gluconeogenesis pathway. This study reveals that Ruminococcus_E bovis is associated with host energy metabolism homeostasis by supplying glucogenic precursors to the liver and suggests the use of Ala as a method for the treatment of energy metabolism disorders in peripartum cows.
Collapse
Affiliation(s)
- Fanlin Kong
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
China Agricultural University, Beijing 100193, China
| | - Shuo Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
China Agricultural University, Beijing 100193, China
| | - Yijia Zhang
- Laboratory of Animal Neurobiology, Department of Basic Veterinary Medicine, College of Veterinary Medicine,
Nanjing Agricultural University, Nanjing 210095, China
| | - Chen Li
- Department of Animal Nutrition and Feed Science, College of Animal Science,
Xinjiang Agricultural University, Urumqi 830052, China
| | - Dongwen Dai
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
Ningxia University, Yinchuan 750021, China
| | - Yajing Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
China Agricultural University, Beijing 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
China Agricultural University, Beijing 100193, China
| | - Hongjian Yang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
China Agricultural University, Beijing 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
China Agricultural University, Beijing 100193, China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology,
China Agricultural University, Beijing 100193, China
| |
Collapse
|
4
|
Chen Z, Zhang X, Teng J, Jiang L, Zhang Q. HSF1 and CPSF1 affect milk fat and protein synthesis by regulating the AKT/mTOR signaling pathway. J Anim Sci 2025; 103:skaf009. [PMID: 39932399 PMCID: PMC11897896 DOI: 10.1093/jas/skaf009] [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: 09/18/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
In our previous genome-wide association study on milk production traits in Chinese Holstein cows, HSF1 (heat shock factor 1) and CPSF1 (cleavage and polyadenylation specific factor 1) were found to be strongly associated with milk fat and protein percentages. However, their roles in milk fat and protein synthesis and the underlying mechanism are still largely unknown. In this study, we verified the effects of their expressions on milk fat and milk protein synthesis in MAC-T cells. We showed that HSF1 can participate in the AKT/mTOR signaling pathway, one of the most important pathways for fat and protein synthesis, through its interaction with the AKT protein and influence the downstream genes in this pathway to regulate milk fat and milk protein synthesis. We also found that HSF1, as a transcription factor, can bind to the promoter region of CPSF1 to regulate its transcription and expression, which in turn modulates the expression of SREBP1 and thereby influences the synthesis of milk fat.
Collapse
Affiliation(s)
- Zhujun Chen
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an, China
| | - Xinyi Zhang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an, China
| | - Jun Teng
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an, China
| | - Li Jiang
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science & Technology, China Agricultural University, Beijing, China
| | - Qin Zhang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science, Shandong Agricultural University, Tai’an, China
| |
Collapse
|
5
|
Shen T, Xia S, Usman M, Xu X, Loor JJ, Xu C. Nuclear factor erythroid 2-related factor 1 regulates the expression of proteasomal genes in ketotic cows and protects mammary cells against free fatty acid-induced endoplasmic reticulum stress. J Dairy Sci 2025; 108:1050-1061. [PMID: 39343197 DOI: 10.3168/jds.2024-25369] [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: 06/30/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
Ketosis is a common metabolic disorder in high-yielding cows and is characterized by high concentrations of BHB and free fatty acids (FFA). High concentrations of FFA induce endoplasmic reticulum (ER) stress in multiple organs including mammary tissue, and result in reduced milk production and lower milk quality. In nonruminants, loss of nuclear factor erythroid 2-related factor 1 (NFE2L1) results in ER stress. The physiological functions and molecular mechanisms controlled by NFE2L1 in bovine mammary tissue are poorly understood. Thus, the present study aimed to elucidate the role of the NFE2L1 on proteasomal homeostasis and ER stress in mammary tissue from early lactation (DIM 6-14) healthy cows (CON, blood concentration of BHB <1.2 mM, n = 10) and cows with clinical ketosis (CK blood concentration of BHB >3 mM, n = 10). Compared with CON, serum concentration of glucose was lower due to CK, while serum concentrations of BHB and FFA were greater. Protein and mRNA abundance of NFE2L1 along with abundance of proteasomal subunits (PSMD1, PSMD14, PSMA1, PSMB1, and PSMB5 genes and PSMB4 and PSMB6 proteins) were lower in cows with CK, indicating that expression of NFE2L1 and proteasomal homeostasis was impaired by ketosis. In vitro, primary bovine mammary epithelial cells were exposed to various concentrations of FFA (0, 0.3, 0.6, or 1.2 mM). Compared with the 0 mM FFA, the ratio of phosphorylated (p)-protein kinase R-like ER kinase (PERK)/PERK along with the expression of inositol-requiring enzyme 1 (IRE1) α, activating transcription factor 6 (ATF6), glucose regulated protein 78 (GRP78), and C/EBP homologous protein (CHOP) was higher with 1.2 mM FFA. A similar response was observed for ER stress-associated genes (CHOP, GRP78, and XBP1) indicating that high concentrations of FFA-induced ER stress. In line with in vivo results, 1.2 mM FFA downregulated the protein and mRNA abundance of NFE2L1, the abundance of PSMB6 protein, and proteasome subunit (PSM) genes (PSMC1, PSMC3, and PSMD1), and increased the accumulation of ubiquitin. This suggested a marked negative effect of high FFA on NFE2L1 and proteasomal homeostasis. Silencing of NFE2L1 triggered upregulation of ER stress-associated genes as well as protein abundance of GRP78 and CHOP. Further, compared with CON-siRNA, the abundance of PSM genes was downregulated in the NFE2L1-siRNA group. In contrast, abundance of markers of ER stress and PSM genes and proteins indicated that overexpression of NFE2L1 relieved the FFA-induced ER stress and improved 26S proteasome homeostasis. Our data suggested that the mammary gland experiences ER stress during ketosis partly due to disruption of proteasomal homeostasis from the excess FFA. As such, NFE2L1 could represent a target for potential therapeutic applications in the field to alleviate the accumulation of malformed proteins that may impair the long-term lactogenic capacity of the udder.
Collapse
Affiliation(s)
- Taiyu Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shijie Xia
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Muhammad Usman
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Xinyi Xu
- College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Chuang Xu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
6
|
Wang X, Zhang C, Li R, Qiu Y, Ma Y, Wang S, Li Y, Guo S, Li C. Down-regulation of miR-29 improves lipid metabolism in fatty liver of dairy cows. Anim Biotechnol 2024; 35:2396414. [PMID: 39205627 DOI: 10.1080/10495398.2024.2396414] [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: 01/15/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
In this study, we conducted a thorough investigation into the mechanisms by which miR-29 influences lipid metabolism. Thirty-two cows were selected and categorized into distinct groups based on their liver triglyceride (TG) content: healthy, mild fatty liver, and moderate fatty liver groups. Dairy cows with moderate fatty liver showed higher levels of hepatic lipid accumulation, MDA content and serum AST, ALT and ALP contents and lower hepatic catalase CAT and SOD activities. Subsequently, hepatocytes isolated from healthy calves were exposed to sodium oleate (SO) in the presence or absence of pre-incubation with miR-29 inhibitor or inhibitor NC. Pre-transfection with miR-29 inhibitor resulted in reduced hepatocyte lipid accumulation and MDA levels, as well as decreased levels of AST, ALT, and ALP in the supernatant. In the miR-29 inhibitor + SO group, there was an increase in the expression of SREBP-1, FAS, SCD1, and Sirt1. Meanwhile, the expression of PPARα, CPT1, CPT2, PGC-1α, NRF-1, UCP2, and miR-29 were observed to be decreased. In comparison to the miR-29 inhibitor + SO group, some of the measured indicators showed partial reversal in the miR-29 inhibitor + siSirt1 + SO group. Collectively, these findings provide evidence that miR-29 may play a crucial role in the pathogenesis of fatty liver in dairy cows.
Collapse
Affiliation(s)
- Xueying Wang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Cai Zhang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Rishun Li
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Yan Qiu
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Yanbo Ma
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
- Longmen Laboratory, Innovative Research Team of Livestock Intelligent Breeding and Equipment, Luoyang, China
| | - Shuai Wang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Yuanxiao Li
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Shuai Guo
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Chenxu Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun Jilin, China
| |
Collapse
|
7
|
Yang W, Tian Y, Yang M, Mauck J, Loor JJ, Jia B, Wang S, Fan W, Li Z, Zhang B, Xu C. β-sitosterol alleviates high fatty acid-induced lipid accumulation in calf hepatocytes by regulating cholesterol metabolism. J Steroid Biochem Mol Biol 2024; 243:106543. [PMID: 38740074 DOI: 10.1016/j.jsbmb.2024.106543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
A significant reduction in plasma concentration of cholesterol during early lactation is a common occurrence in high-yielding dairy cows. An insufficient synthesis of cholesterol in the liver has been linked to lipid accumulation caused by high concentrations of fatty acids during negative energy balance (NEB). As ruminant diets do not provide quantitative amounts of cholesterol for absorption, phytosterols such as β-sitosterol may serve to mitigate the shortfall in cholesterol within the liver during NEB. To gain mechanistic insights, primary hepatocytes were isolated from healthy female 1-day old calves for in vitro studies with or without 1.2 mM fatty acids (FA) to induce metabolic stress. Furthermore, hepatocytes were treated with 50 μM β-sitosterol with or without FA. Data were analyzed by one-way ANOVA with subsequent Bonferroni correction. Results revealed that calf hepatocytes treated with FA had greater content of non-esterified fatty acids (NEFA) and triacylglycerol (TAG), and greater mRNA and protein abundance of the lipid synthesis-related SREBF1 and FASN. In contrast, mRNA and protein of CPT1A (fatty acid oxidation) and the cholesterol metabolism-related targets SREBF2, HMGCR, ACAT2, APOA1, ABCA1 and ABCG5 was lower. Content of the antioxidant-related glutathione (GSH) and activities of superoxide dismutase (SOD) also was lower. Compared with FA challenge alone, 50 μM β-sitosterol led to greater mRNA and protein abundance of SREBF2, HMGCR, ACAT2 and ABCG5, and greater content of GSH and activity of SOD. In contrast, compared with the FA group, the mRNA and protein abundance of SREBF1 and ACC1 and the content of TAG and NEFA in the β-sitosterol + FA group were lower. Overall, β-sitosterol can promote cholesterol metabolism and reduce oxidative stress while reducing lipid accumulation in hepatocytes challenged with high concentrations of fatty acids.
Collapse
Affiliation(s)
- Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yan Tian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Mingmao Yang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A & F University, Xianyang 712100, China
| | - John Mauck
- Mammalian Nutri Physio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| | - Juan J Loor
- Mammalian Nutri Physio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| | - Bin Jia
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar 163005, China
| | - Shuang Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Wenwen Fan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Zhendong Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
8
|
Ma G, Zuo Z, Li H, Zhai X, Wang T, Wang J. Comparative analysis of the liver transcriptome in the red-eared slider (Trachemys scripta elegans) post exposure to noise. PLoS One 2024; 19:e0305858. [PMID: 39088466 PMCID: PMC11293744 DOI: 10.1371/journal.pone.0305858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/05/2024] [Indexed: 08/03/2024] Open
Abstract
Exposure to noise can cause non-auditory health problems and has been widely studied in mammals such as rats and rabbits. However, the non-auditory effects of noise exposure on reptiles (such as red-eared sliders) remain unclear. In this study, we determined the noise exposure-induced transcriptomic changes in the liver of red-eared slider (Trachemys scripta elegans) using Illumina Novaseq6000 sequencing technology. The transcriptome analysis identified 176 differentially expressed genes (DEGs), which were mainly enriched in lipid metabolism. KEGG analysis showed that by affecting the peroxisome proliferator activated receptor (PPAR) signaling pathway these DEGs increased lipid synthesis and decreased lipid oxidation. The Oil Red O staining results validated our data that noise exposure increased hepatic lipid deposition. Thus, noise exposure may lead to lipid accumulation and toxicity, mitochondrial damage, and accelerated oxidative stress. Our findings provide insights into the molecular process underlying non-auditory damage caused by noise exposure in T. scripta elegans.
Collapse
Affiliation(s)
- Guangwei Ma
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Ziye Zuo
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Handong Li
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Xiaofei Zhai
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Tongliang Wang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Jichao Wang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| |
Collapse
|
9
|
Mohsin MA, Zhou X, Huiru Y, Shen W, He B, Sobiech P, Pierzchała M, Ogłuszka M, Starzyński R, Kalra G, Deshmukh B, Thangarasu R, Kashyap N, Czarnik U, Lepczyński A, Woźniakowski G, Pareek CS. Effect of β-hydroxybutyrate acid on gene expression levels of antioxidant biomarkers and growth hormone-related genes in liver cell culture. J Vet Res 2024; 68:313-324. [PMID: 38947149 PMCID: PMC11210367 DOI: 10.2478/jvetres-2024-0037] [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: 11/01/2023] [Accepted: 06/17/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction In dairy cattle, oxidative stress is a predominant problem associated with diseases and reproductive health issues. This study aimed to detect the variation in the antioxidant biomarkers by adding different concentrations of β-hydroxybutyric acid (BHBA) and sought to elucidate its effects on the gene expression levels of growth hormone (GH) and antioxidant biomarkers in bovine hepatocytes. Material and Methods Four antioxidant biomarkers, namely malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH Px) were evaluated using commercially available bovine ELISA kits. The expression levels of the bovine GH, its receptor (GHR), insulin-like growth factor (IGF), IGF-1, IGF-1 receptor, CAT, SOD, GSH-Px and β-actin (as a reference) genes in liver cell culture were determined by reverse transcriptase-PCR assay. Results With the increase of BHBA concentration and culture time, the activities of SOD, CAT, and GSH Px biomarkers in hepatocytes decreased. However, the content of MDA in hepatocytes increased gradually with the increase of hepatocyte culture time and BHBA concentration. The qPCR results revealed that after adding BHBA, gene expression levels of GSH-Px, SOD and IGF biomarkers in hepatocytes began to differ in the culture groups at 12 h, whereas the gene expression level of the CAT and GHR biomarkers in hepatocytes began to differ at 6 h. Conclusion Quantitative PCR results showed that the BHBA significantly downregulated the expression levels of the GHR gene and CAT, GSH Px and SOD antioxidant biomarker genes.
Collapse
Affiliation(s)
- Muhammad Ali Mohsin
- Department of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
| | - Xiaojing Zhou
- Department of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
| | - Yu Huiru
- Department of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
- Shanghai Animal Disease Prevention and Control Center, Changning District, 201103, Shanghai, China
| | - Wenxiang Shen
- Department of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
| | - Baoxiang He
- Department of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi, China
| | - Przemysław Sobiech
- Department of Clinical Sciences, Internal Disease Unit, Faculty of Veterinary Medicine, Utrecht, Netherlands
| | - Mariusz Pierzchała
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552Magdalenka, Poland
| | - Magdalena Ogłuszka
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552Magdalenka, Poland
| | - Rafał Starzyński
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552Magdalenka, Poland
| | - Garima Kalra
- Department of Animal Genetics and Breeding, Ludhiana, Punjab, India
| | - Bharti Deshmukh
- Department of Animal Genetics and Breeding, Ludhiana, Punjab, India
| | - Revathy Thangarasu
- College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, 141004, Punjab, India
| | - Neeraj Kashyap
- College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, 141004, Punjab, India
| | - Urszula Czarnik
- Department of Pig Breeding, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, 10-719Olsztyn, Poland
| | - Adam Lepczyński
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology, 71-270Szczecin, Poland
| | - Grzegorz Woźniakowski
- Department of Infectious and Invasive Diseases and Veterinary Administration, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100Toruń, Poland
- Division of Functional Genomics in Biological and Biomedical Research, Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100Toruń, Poland
| | - Chandra S. Pareek
- Department of Infectious and Invasive Diseases and Veterinary Administration, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100Toruń, Poland
- Division of Functional Genomics in Biological and Biomedical Research, Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87-100Toruń, Poland
| |
Collapse
|
10
|
Danielewski M, Rapak A, Kruszyńska A, Małodobra-Mazur M, Oleszkiewicz P, Dzimira S, Kucharska AZ, Słupski W, Matuszewska A, Nowak B, Szeląg A, Piórecki N, Zaleska-Dorobisz U, Sozański T. Cornelian Cherry ( Cornus mas L.) Fruit Extract Lowers SREBP-1c and C/EBPα in Liver and Alters Various PPAR-α, PPAR-γ, LXR-α Target Genes in Cholesterol-Rich Diet Rabbit Model. Int J Mol Sci 2024; 25:1199. [PMID: 38256272 PMCID: PMC10816641 DOI: 10.3390/ijms25021199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Cornelian cherry (Cornus mas L.) fruits, abundant in iridoids and anthocyanins, are natural products with proven beneficial impacts on the functions of the cardiovascular system and the liver. This study aims to assess and compare whether and to what extent two different doses of resin-purified cornelian cherry extract (10 mg/kg b.w. or 50 mg/kg b.w.) applied in a cholesterol-rich diet rabbit model affect the levels of sterol regulatory element-binding protein 1c (SREBP-1c) and CCAAT/enhancer binding protein α (C/EBPα), and various liver X receptor-α (LXR-α), peroxisome proliferator-activated receptor-α (PPAR-α), and peroxisome proliferator-activated receptor-γ (PPAR-γ) target genes. Moreover, the aim is to evaluate the resistive index (RI) of common carotid arteries (CCAs) and aortas, and histopathological changes in CCAs. For this purpose, the levels of SREBP-1c, C/EBPα, ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette transporter G1 (ABCG1), fatty acid synthase (FAS), endothelial lipase (LIPG), carnitine palmitoyltransferase 1A (CPT1A), and adiponectin receptor 2 (AdipoR2) in liver tissue were measured. Also, the levels of lipoprotein lipase (LPL), visceral adipose tissue-derived serine protease inhibitor (Vaspin), and retinol-binding protein 4 (RBP4) in visceral adipose tissue were measured. The RI of CCAs and aortas, and histopathological changes in CCAs, were indicated. The oral administration of the cornelian cherry extract decreased the SREBP-1c and C/EBPα in both doses. The dose of 10 mg/kg b.w. increased ABCA1 and decreased FAS, CPT1A, and RBP4, and the dose of 50 mg/kg b.w. enhanced ABCG1 and AdipoR2. Mitigations in atheromatous changes in rabbits' CCAs were also observed. The obtained outcomes were compared to the results of our previous works. The beneficial results confirm that cornelian cherry fruit extract may constitute a potentially effective product in the prevention and treatment of obesity-related disorders.
Collapse
Affiliation(s)
- Maciej Danielewski
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Andrzej Rapak
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (A.K.)
| | - Angelika Kruszyńska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (A.K.)
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland;
| | - Paweł Oleszkiewicz
- Department of Radiology and Imaging Diagnostics II, Lower Silesian Center of Oncology, Pulmonology and Hematology, Grabiszynska 105, 53-439 Wroclaw, Poland;
| | - Stanisław Dzimira
- Department of Pathology, Wroclaw University of Environmental and Life Sciences, C. K. Norwida 31, 50-375 Wroclaw, Poland;
| | - Alicja Z. Kucharska
- Department of Fruit, Vegetable, and Plant Nutraceutical Technology, Wroclaw University of Environmental and Life Sciences, J. Chelmonskiego 37, 51-630 Wroclaw, Poland;
| | - Wojciech Słupski
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Agnieszka Matuszewska
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Beata Nowak
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Adam Szeląg
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Narcyz Piórecki
- Bolestraszyce Arboretum and Institute of Physiography, Bolestraszyce 130, 37-722 Wyszatyce, Poland;
- Institute of Physical Culture Sciences, Medical College, University of Rzeszow, Cicha 2A, 35-326 Rzeszow, Poland
| | - Urszula Zaleska-Dorobisz
- Department of General and Pediatric Radiology, Wroclaw Medical University, M. Sklodowskiej-Curie 50/52, 50-369 Wroclaw, Poland;
| | - Tomasz Sozański
- Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Faculty of Medicine, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| |
Collapse
|
11
|
Xia M, Wu Z, Wang J, Buist-Homan M, Moshage H. The Coumarin-Derivative Esculetin Protects against Lipotoxicity in Primary Rat Hepatocytes via Attenuating JNK-Mediated Oxidative Stress and Attenuates Free Fatty Acid-Induced Lipid Accumulation. Antioxidants (Basel) 2023; 12:1922. [PMID: 38001774 PMCID: PMC10669015 DOI: 10.3390/antiox12111922] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Coumarin derivates have been proposed as a potential treatment for metabolic-dysfunction-associated fatty liver disease (MAFLD). However, the mechanisms underlying their beneficial effects remain unclear. In the present study, we explored the potential of the coumarin derivate esculetin in MAFLD, focusing on hepatocyte lipotoxicity and lipid accumulation. Primary cultures of rat hepatocytes were exposed to palmitic acid (PA) and palmitic acid plus oleic acid (OA/PA) as models of lipotoxicity and lipid accumulation, respectively. Esculetin significantly reduced oxidative stress in PA-treated hepatocytes, as shown by decreased total reactive oxygen species (ROS) and mitochondrial superoxide production and elevated expression of antioxidant genes, including Nrf2 and Gpx1. In addition, esculetin protects against PA-induced necrosis. Esculetin also improved lipid metabolism in primary hepatocytes exposed to nonlipotoxic OA/PA by decreasing the expression of the lipogenesis-related gene Srebp1c and increasing the expression of the fatty acid β-oxidation-related gene Ppar-α. Moreover, esculetin attenuated lipid accumulation in OA/PA-treated hepatocytes. The protective effects of esculetin against lipotoxicity and lipid accumulation were shown to be dependent on the inhibition of JNK and the activation of AMPK, respectively. We conclude that esculetin is a promising compound to target lipotoxicity and lipid accumulation in the treatment of MAFLD.
Collapse
Affiliation(s)
- Mengmeng Xia
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (M.X.); (Z.W.); (J.W.); (M.B.-H.)
| | - Zongmei Wu
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (M.X.); (Z.W.); (J.W.); (M.B.-H.)
| | - Junyu Wang
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (M.X.); (Z.W.); (J.W.); (M.B.-H.)
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (M.X.); (Z.W.); (J.W.); (M.B.-H.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (M.X.); (Z.W.); (J.W.); (M.B.-H.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
12
|
Lisuzzo A, Alterisio MC, Mazzotta E, Ciaramella P, Guccione J, Gianesella M, Badon T, Fiore E. Metabolic Changes Associated with Different Levels of Energy Deficits in Mediterranean Buffaloes during the Early Lactation Stage: Type and Role of the Main Lipid Fractions Involved. Animals (Basel) 2023; 13:2333. [PMID: 37508110 PMCID: PMC10376174 DOI: 10.3390/ani13142333] [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: 06/06/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Cell function and energy redistribution are influenced by lipid classes (phospholipids (PLs), free fatty acids (FFAs), triglycerides (TGs), and cholesterol esters (CEs)). The aim of this study was to investigate metabolic alterations that are related to changes in lipid classes according to different levels of energy deficits in early lactating Mediterranean buffaloes (MBs). Sixty-three MBs were enrolled at the beginning of lactation using an observational study with a cross-sectional experimental design. Serum β-hydroxybutyrate (BHB) levels were used to group the animals into a healthy group (Group H; n = 38; BHB < 0.70 mmol/L) and hyperketonemia risk group (Group K; n = 25; BHB ≥ 0.70 mmol/L). Statistical analysis was performed using a linear model that included the effect of the group and body condition score to assess differences in fatty acid (FA) concentrations. A total of 40 plasma FAs were assessed in each lipid class. Among the FAs, eight PLs, seven FFAs, four TGs, and four CEs increased according to BHB levels, while three FFAs, three TGs, and one CE decreased. The changes among lipid class profiles suggested the influence of inflammatory response, liver metabolism, and the state of body lipid reserves. In addition, the possible similarities of buffaloes at risk of hyperketonemia with ketotic cows suggest the necessity of further investigations in these ruminants.
Collapse
Affiliation(s)
- Anastasia Lisuzzo
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Legnaro, Italy
| | - Maria Chiara Alterisio
- Department of Veterinary Medicine and Animal Productions, University of Napoli "Federico II", 80137 Napoli, Italy
| | - Elisa Mazzotta
- Istituto Zooprofilattico Sperimentale delle Venezie (IZSVE), 35020 Legnaro, Italy
| | - Paolo Ciaramella
- Department of Veterinary Medicine and Animal Productions, University of Napoli "Federico II", 80137 Napoli, Italy
| | - Jacopo Guccione
- Department of Veterinary Medicine and Animal Productions, University of Napoli "Federico II", 80137 Napoli, Italy
| | - Matteo Gianesella
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Legnaro, Italy
| | - Tamara Badon
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Legnaro, Italy
| | - Enrico Fiore
- Department of Animal Medicine, Production and Health, University of Padua, 35020 Legnaro, Italy
| |
Collapse
|
13
|
Zhao C, Wu B, Li J, Jiang Q, Loor JJ, Liu M, Chen L, Zhu Y, Gao W, Du X, Song Y, Liu G, Lei L, Li X. AdipoRon alleviates fatty acid-induced lipid accumulation and mitochondrial dysfunction in bovine hepatocytes by promoting autophagy. J Dairy Sci 2023:S0022-0302(23)00295-3. [PMID: 37268562 DOI: 10.3168/jds.2022-22723] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/28/2023] [Indexed: 06/04/2023]
Abstract
During the transition period in dairy cows, high circulating concentrations of nonesterified fatty acids (NEFA) increase hepatic lipid deposits and are considered a major pathological factor for liver damage. We investigated whether AdipoRon, a synthetic small-molecule agonist of adiponectin receptors 1 and 2 shown to prevent liver lipid accumulation in nonruminants, could alleviate NEFA-induced lipid accumulation and mitochondrial dysfunction. Bovine hepatocytes were isolated from 5 healthy Holstein female newborn calves (1 d of age, 30-40 kg, fasting), and independently isolated hepatocytes from at least 3 different calves were used for each subsequent experiment. The composition and concentration of NEFA used in this study were selected according to hematological criteria of dairy cows with fatty liver or ketosis. First, hepatocytes were cultured with various concentrations of NEFA (0, 0.6, 1.2, or 2.4 mM) for 12 h. In a second experiment, hepatocytes were treated with AdipoRon at different concentrations (0, 5, 25, or 50 μM for 12 h) and times (25 μM for 0, 6, 12, or 24 h) with or without NEFA (1.2 mM) treatment. In the last experiment, hepatocytes were treated with AdipoRon (25 μM), NEFA (1.2 mM), or both for 12 h after treatment with or without the autophagy inhibitor chloroquine. Hepatocytes treated with NEFA had increased protein abundance of sterol regulatory element-binding protein 1c (SREBP-1c) and mRNA abundance of acetyl-CoA carboxylase 1 (ACACA), and decreased protein abundance of peroxisome proliferator-activated receptor α (PPARA), proliferator-activated receptor gamma coactivator-1 α (PGC-1α), mitofusin 2 (MFN2), cytochrome c oxidase subunit IV (COX IV), and mRNA abundance of carnitine palmitoyltransferase 1A (CPT1A), along with lower ATP concentrations. AdipoRon treatment reversed these effects, suggesting this compound had a positive effect on lipid metabolism and mitochondrial dysfunction during the NEFA challenge. In addition, upregulated expression of microtubule-associated protein 1 light chain 3-II (LC3-II, encoded by MAP1LC3) and downregulated expression of sequestosome-1 (SQSTM1, also called p62) indicated that AdipoRon enhanced autophagic activity in hepatocytes. The fact that chloroquine impeded the beneficial effects of AdipoRon on lipid accumulation and mitochondrial dysfunction suggested a direct role for autophagy during NEFA challenge. Our results suggest that autophagy is an important cellular mechanism to prevent NEFA-induced lipid accumulation and mitochondrial dysfunction in bovine hepatocytes, which is consistent with other studies. Overall, AdipoRon may represent a promising therapeutic agent to maintain hepatic lipid homeostasis and mitochondrial function in dairy cows during the transition period.
Collapse
Affiliation(s)
- Chenchen Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bing Wu
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Jinxia Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Menglin Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Linfang Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yiwei Zhu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiliang Du
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuxiang Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guowen Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
14
|
Pregnancy Toxemia in Ewes: A Review of Molecular Metabolic Mechanisms and Management Strategies. Metabolites 2023; 13:metabo13020149. [PMID: 36837768 PMCID: PMC9961611 DOI: 10.3390/metabo13020149] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Pregnancy toxemia is a nutritional metabolic disease during late gestation in small ruminants. The condition is characterized by disorders in carbohydrate and fat metabolism. Obese and multiparous ewes are particularly susceptible to pregnancy toxemia, which may lead to maternal death, abortion, or premature birth. Highly productive multiparous meat ewes are major breeding animals, which has led to an increased incidence of the disease. However, the pathogenesis of pregnancy toxemia remains unclear and adequate disease prevention and treatment strategies are absent. Investigating the pathogenesis of pregnancy toxemia, especially the metabolic pathways of hepatic lipids, is key to an improved understanding of the condition. This review provides a snapshot of the genes that are associated with lipid metabolism in the ovine liver, including genes involved in fatty acid oxidation, acetyl coenzyme metabolism, and triglyceride synthesis; describes the interrelationships between these genes; and summarizes the diagnosis, prevention, and treatment of pregnancy toxemia.
Collapse
|
15
|
The E3 ubiquitin ligase NEDD4-1 protects against acetaminophen-induced liver injury by targeting VDAC1 for degradation. Acta Pharm Sin B 2023; 13:1616-1630. [PMID: 37139424 PMCID: PMC10150139 DOI: 10.1016/j.apsb.2023.01.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/27/2022] [Accepted: 12/15/2022] [Indexed: 01/30/2023] Open
Abstract
Acetaminophen (APAP) overdose is a major cause of liver injury. Neural precursor cell expressed developmentally downregulated 4-1 (NEDD4-1) is an E3 ubiquitin ligase that has been implicated in the pathogenesis of numerous liver diseases; however, its role in APAP-induced liver injury (AILI) is unclear. Thus, this study aimed to investigate the role of NEDD4-1 in the pathogenesis of AILI. We found that NEDD4-1 was dramatically downregulated in response to APAP treatment in mouse livers and isolated mouse hepatocytes. Hepatocyte-specific NEDD4-1 knockout exacerbated APAP-induced mitochondrial damage and the resultant hepatocyte necrosis and liver injury, while hepatocyte-specific NEDD4-1 overexpression mitigated these pathological events both in vivo and in vitro. Additionally, hepatocyte NEDD4-1 deficiency led to marked accumulation of voltage-dependent anion channel 1 (VDAC1) and increased VDAC1 oligomerization. Furthermore, VDAC1 knockdown alleviated AILI and weakened the exacerbation of AILI caused by hepatocyte NEDD4-1 deficiency. Mechanistically, NEDD4-1 was found to interact with the PPTY motif of VDAC1 through its WW domain and regulate K48-linked ubiquitination and degradation of VDAC1. Our present study indicates that NEDD4-1 is a suppressor of AILI and functions by regulating the degradation of VDAC1.
Collapse
|
16
|
Acetate-Induced Milk Fat Synthesis Is Associated with Activation of the mTOR Signaling Pathway in Bovine Mammary Epithelial Cells. Animals (Basel) 2022; 12:ani12192616. [PMID: 36230357 PMCID: PMC9558539 DOI: 10.3390/ani12192616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Acetate is a precursor substance for fatty acid synthesis in bovine mammary epithelial cells (BMECs), and the mTOR signaling pathway plays an important role in milk fat synthesis. However, the mechanism of the regulatory effects of acetate on lipogenic genes via the mTOR signaling pathway in BMEC remains unknown. We hypothesized that acetate can enhance the expression of lipogenic genes and triglyceride (TG) production by activating the mTOR signaling pathway in BMECs. Therefore, the aim of this study was to investigate the network of acetate-regulated lipid metabolism by the mTOR signaling pathway in BMECs. These results showed that TG synthesis was elevated (p < 0.01) in BMECs with acetate treatment. The lipid droplets were increased in the acetate-treated groups compared with those in the control group through the Bodipy staining of the lipids. In addition, the fatty acid profile in BMECs treated with acetate was affected, with an elevation in the proportions of C14:0, C16:0, and C18:0. The mRNA levels of the sterol-response-element-binding protein 1 (SREBP1), stearoyl-CoA desaturase 1 (SCD1), and fatty acid synthase (FAS) genes involved in the lipogenesis and transcriptional factors were upregulated (p < 0.05) in BMECs with acetate treatment. Remarkably, the expression of acetyl-CoA carboxylase α (ACCα) and FAS rate-limiting enzymes involved in lipogenesis was upregulated in BMECs with acetate treatment. Moreover, the addition of acetate enhanced the key protein expression of S6K1, which is related to the mTOR signaling pathway. Taken together, our data suggest that TG accumulation and expression of lipogenic genes induced by acetate are associated with the activation of the mTOR signaling pathway, which provides new insights into the understanding of the molecular mechanism in the expression of mTOR-signaling-pathway-regulated lipogenic genes.
Collapse
|
17
|
Reichelt K, Niebisch AM, Kacza J, Schoeniger A, Fuhrmann H. The Bovine Hepatic Cell Line BFH12 as a Possible Model for Hepatosteatosis in Dairy Cows. Front Vet Sci 2022; 9:840202. [PMID: 35359674 PMCID: PMC8963807 DOI: 10.3389/fvets.2022.840202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatosteatosis is a common metabolic disorder of dairy cows, especially during early lactation. Currently, there are a few models of bovine hepatic steatosis available, including primary hepatocytes, liver slices, and animal models. Studies that elucidate the influence of single fatty acids on lipid classes, fatty acid pattern, gene expression, and phenotypic changes are still limited. Hence, we investigated the suitability of the fetal bovine hepatocyte-derived cell line BFH12 as a model for hepatosteatosis. To create a steatotic environment, we treated BFH12 with stearic acid, palmitic acid, or oleic acid in non-toxic doses. Thin-layer chromatography and gas chromatography were used to analyze lipid classes and fatty acid pattern, and qPCR was used to quantify gene expression of relevant target genes. Lipid droplets were visualized with confocal laser scanning microscopy and evaluated for number and size. Treatment with oleic acid increased triglycerides, as well as lipid droplet count per cell and upregulated carnitine palmitoyl transferase 1, which correlates with findings of in vivo models. Oleic acid was largely incorporated into triglycerides, phospholipids, and non-esterified fatty acids. Stearic acid was found mainly in non-esterified fatty acids and triglycerides, whereas palmitic acid was mainly desaturated to palmitoleic acid. All three fatty acids downregulated stearyl-CoA-desaturase 1. In conclusion, BFH12 can acquire a steatotic phenotype by incorporating and accumulating fatty acids. Oleic acid is particularly suitable to produce hepatosteatosis. Therefore, BFH12 may be a useful in vitro model to study bovine hepatosteatosis and its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Kristin Reichelt
- Faculty of Veterinary Medicine, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
- *Correspondence: Kristin Reichelt
| | - Anna M. Niebisch
- Faculty of Veterinary Medicine, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Johannes Kacza
- BioImaging Core Facility, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Axel Schoeniger
- Faculty of Veterinary Medicine, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Herbert Fuhrmann
- Faculty of Veterinary Medicine, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| |
Collapse
|
18
|
Fang Z, Li X, Wang S, Jiang Q, Loor JJ, Jiang X, Ju L, Yu H, Shen T, Chen M, Song Y, Wang Z, Du X, Liu G. Overactivation of hepatic mechanistic target of rapamycin kinase complex 1 (mTORC1) is associated with low transcriptional activity of transcription factor EB and lysosomal dysfunction in dairy cows with clinical ketosis. J Dairy Sci 2022; 105:4520-4533. [DOI: 10.3168/jds.2021-20892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 01/14/2022] [Indexed: 11/19/2022]
|
19
|
Yang W, Wang S, Loor JJ, Lopes MG, Zhao Y, Ma X, Li M, Zhang B, Xu C. Role of diacylglycerol O-acyltransferase (DGAT) isoforms in bovine hepatic fatty acid metabolism. J Dairy Sci 2022; 105:3588-3600. [PMID: 35181144 DOI: 10.3168/jds.2021-21140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022]
Abstract
Fatty acid accumulation in hepatocytes induced by high concentrations of fatty acids due to lipolysis and the associated oxidative damage they cause occur most frequently after calving. Because of their role in esterification of fatty acids, diacylglycerol acyltransferase isoforms (DGAT1 and DGAT2) could play a role in the susceptibility of dairy cows to develop fatty liver. To gain mechanistic insights, we performed in vivo and in vitro analyses using liver biopsies or isolated primary hepatocytes. The in vivo study (n = 5 cows/group) involved healthy cows [average liver triacylglycerol (TAG) = 0.78%; 0.58 to 0.93%, ratio of triglyceride weight to wet liver weight] or cows diagnosed with fatty liver (average TAG = 7.60%; 5.31 to 10.54%). In vitro, hepatocytes isolated from 3 healthy female calves (1 d old, 44 to 53 kg) were challenged with (fatty acids) or without (control) a 1.2 mM mixture of fatty acids in an attempt to induce metabolic stress. Furthermore, hepatocytes were treated with DGAT1 inhibitor or DGAT2 inhibitor for 2 h followed by a challenge with (DGAT1 inhibitor + fatty acids or DGAT2 inhibitor + fatty acids) or without (DGAT1 inhibitor or DGAT2 inhibitor) the 1.2 mM mixture of fatty acids for 12 h. Data analysis of liver biopsies was compared using a 2-tailed unpaired Student's t-test. Data from calf hepatocyte treatment comparisons were assessed by one-way ANOVA, and multiplicity for each experiment was adjusted by the Holm's procedure. Data indicated that both fatty liver and in vitro challenge with fatty acids were associated with greater mRNA and protein abundance of SREBF1, FASN, DGAT1, and DGAT2. In contrast, mRNA and protein abundance of CPT1A and very low-density lipoprotein synthesis-related proteins MTTP and APOB were markedly lower. However, compared with fatty acid challenge alone, DGAT1 inhibitor + fatty acids led to greater mRNA and protein abundance of CPT1A and APOB, and greater mRNA abundance of SREBF1 and MTTP. Furthermore, this treatment led to lower mRNA abundance of FASN and DGAT2 and TAG concentrations. Compared with fatty acid challenge alone, DGAT2 inhibitor + fatty acids led to greater mRNA and protein abundance of CPT1A, MTTP, and APOB, and lower mRNA and protein abundance of SREBF1 and FASN. In addition, compared with control and fatty acids, there was greater protein abundance of GRP78 and PERK in both DGAT1 and DGAT2 inhibitor with or without fatty acids. Furthermore, compared with control and fatty acids, reactive oxygen species concentrations in the DGAT1 inhibitor with or without fatty acid group was greater. Overall, data suggested that DGAT1 is particularly relevant in the context of hepatocyte TAG synthesis from exogenous fatty acids. Disruption of both DGAT1 and DGAT2 altered lipid homeostasis, channeling fatty acids toward oxidation and generation of reactive oxygen species. Both DGAT isoforms play a role in promoting fatty acid storage into TAG and lipid droplets to protect hepatocytes from oxidative damage.
Collapse
Affiliation(s)
- Wei Yang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shuang Wang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Matheus G Lopes
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Yingying Zhao
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Xinru Ma
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ming Li
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Chuang Xu
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| |
Collapse
|
20
|
Mohsin MA, Yu H, He R, Wang P, Gan L, Du Y, Huang Y, Abro MB, Sohaib S, Pierzchala M, Sobiech P, Miętkiewska K, Pareek CS, He BX. Differentiation of Subclinical Ketosis and Liver Function Test Indices in Adipose Tissues Associated With Hyperketonemia in Postpartum Dairy Cattle. Front Vet Sci 2022; 8:796494. [PMID: 35187139 PMCID: PMC8850981 DOI: 10.3389/fvets.2021.796494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022] Open
Abstract
Past studies suggested that during early lactation and the transition period, higher plasma growth hormone (GH) levels in subclinical ketosis (SCK) might involve the initiation of body adipose tissues mobilization, resulting in metabolic disorders in ruminants particularly hyperketonemia. The upregulated GH mRNA expression in adipose tissue may take part in the adipolysis process in SCK-affected cows that paves a way for study further. This study aimed to characterize the plasma levels of GH, β-hydroxybutyrate acid (BHBA) and non-esterified fatty acid (NEFA) and glucose (GLu) in ketotic cows and healthy control (CON) cows; to measure the liver function test (LFT) indices in ketotic and healthy CON cows, and finally the quantitative real-time PCR (qRT-PCR) assay of candidate genes expressed in adipose tissues of ketotic and healthy CON cows during 0 to 7 week postpartum. Three experiments were conducted. Experiment-1 involved 21 Holstein cows weighing 500–600 kg with 2–5 parities. Results showed that GH, BHBA, and NEFA levels in ketotic cows were significantly higher and the GLu level significantly lower. Pearson's correlation analysis revealed a significant positive correlation of GH with BHBA, NEFA, and GLu in ketotic and healthy CON cows. In experiment-2, dynamic monitoring of LFT indices namely, alanine aminotransferase (ALT), aspartate aminotransferase (AST), γ-glutamyl transpeptidase (GGT), total bilirubin (TBIL), direct bilirubin (DBIL), total protein (TP), albumin (ALB), globulin (GLOB) and albumin/globulin (A/G) were examined. The TBIL, DBIL, and GGT indices were significantly higher in ketotic cows and TP was significantly lower. In experiment-3, mRNA expression levels of GHR and peroxisome-proliferator-activated receptor alpha (PPARα) genes in adipose tissue were significantly upregulated in ketotic cows. However, the mRNA expression of insulin-like growth factor-I (IGF-1), insulin-like growth factor-I receptor (IGF-1R), and sterol regulatory element-binding protein-1c (SREBP-1c) genes in adipose tissue were downregulated in ketotic cows. Our study concluded that during postpartum, higher plasma GH levels in SCK cows might involve the initiation of body adipose tissue mobilization, resulting in hyperketonemia.
Collapse
Affiliation(s)
- Muhammad Ali Mohsin
- Laboratory of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Huiru Yu
- Laboratory of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, China
- Shanghai Animal Disease Prevention and Control Center, Shanghai, China
| | - Rongze He
- Laboratory of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Peng Wang
- Laboratory of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Linli Gan
- Laboratory of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yulan Du
- Laboratory of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yunfei Huang
- School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Muhammad Bakhsh Abro
- Department of Veterinary Medicine, Faculty of Veterinary and Animal Science, Lasbela University of Agriculture, Water, and Marine Sciences, Uthal, Pakistan
| | - Sarmad Sohaib
- Department of Veterinary Medicine, Faculty of Veterinary and Animal Science, Lasbela University of Agriculture, Water, and Marine Sciences, Uthal, Pakistan
| | - Mariusz Pierzchala
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Przemysław Sobiech
- Internal Disease Unit, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Klaudia Miętkiewska
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland
- Division of Functional Genomics in Biological and Biomedical Research, Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Torun, Poland
| | - Chandra S. Pareek
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland
- Division of Functional Genomics in Biological and Biomedical Research, Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Torun, Poland
| | - Bao Xiang He
- Laboratory of Clinical Veterinary Medicine, College of Animal Science and Technology, Guangxi University, Nanning, China
- *Correspondence: Bao Xiang He
| |
Collapse
|
21
|
Liu S, Li X, Zhou X, Loor JJ, Jiang Q, Feng X, Yang Y, Lei L, Du X, Li X, Zhe W, Song Y, Liu G. β-Hydroxybutyrate impairs the release of bovine neutrophil extracellular traps through inhibiting phosphoinositide 3-kinase–mediated nicotinamide adenine dinucleotide phosphate oxidase reactive oxygen species production. J Dairy Sci 2022; 105:3405-3415. [DOI: 10.3168/jds.2021-21174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
|
22
|
Yang T, Ma X, Jiang M, Cheng Z, Datsomor O, Zhao G, Zhan K. The Role of Tea Tree Oil in Alleviating Palmitic Acid-Induced Lipid Accumulation in Bovine Hepatocytes. Front Vet Sci 2022; 8:814840. [PMID: 35127885 PMCID: PMC8814581 DOI: 10.3389/fvets.2021.814840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Tea tree oil (TTO) plays an important role in lipid metabolism, alleviating the inflammatory responses. Fatty liver is associated with lipid accumulation in hepatocytes, leading to inflammation. However, there is very limited information on the effects of TTO on lipid accumulation, and inflammation in bovine hepatocytes. This study aimed to evaluate whether TTO alleviates palmitic acid (PA)-induced lipid accumulation in bovine hepatocytes. Hepatocytes isolated from mid-lactating Holstein cows were pretreated with 100 μM PA for 72 h. Cells were either pretreated with PA alone (PA group) or with PA followed by 0.00625% TTO treatment for 12 h (PT group). Expression of fatty acid oxidant genes increased (P < 0.05) while fatty acid synthesis genes decreased (P < 0.05) in the PT group compared with the PA group. PA treatment resulted in increased (P < 0.05) expression of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), but these increases were less in the PT group (P < 0.05). Compared to the PA group, expression of phosphorylated (p)-p65 and p-inhibitor κBα (p-IκBα) was suppressed (P < 0.05) by TTO treatment. TTO treatment limited (P < 0.05) the increase in intracellular reactive oxygen species (ROS) and prevented (P < 0.05) a reduction in mitochondrial membrane potential observed in response to PA treatment. Expression of endoplasmic reticulum (ER) stress genes was reduced (P < 0.05) in the PT group compared with the PA group. Our results suggest that TTO treatment attenuates the effects of PA in hepatocytes, leading to fatty acid oxidation, decreased fatty acid synthesis, suppressed inflammatory response, and reduced ER stress. Taken together, the results of this study suggest that TTO treatment may be a promising therapeutic approach to imbalanced lipid homeostasis, inflammation and ER stress in dairy cows shortly before and after calving.
Collapse
|
23
|
Tsuji A, Ikeda Y, Murakami M, Kitagishi Y, Matsuda S. Reduction of oocyte lipid droplets and meiotic failure due to biotin deficiency was not rescued by restoring the biotin nutritional status. Nutr Res Pract 2022; 16:314-329. [PMID: 35663441 PMCID: PMC9149319 DOI: 10.4162/nrp.2022.16.3.314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/29/2021] [Accepted: 09/09/2021] [Indexed: 11/04/2022] Open
Affiliation(s)
- Ai Tsuji
- Department of Food Science and Nutrition, Faculty of Human Life and Environment, Nara Women's University, Nara 630-8506, Japan
| | - Yuka Ikeda
- Department of Food Science and Nutrition, Faculty of Human Life and Environment, Nara Women's University, Nara 630-8506, Japan
| | - Mutsumi Murakami
- Department of Food Science and Nutrition, Faculty of Human Life and Environment, Nara Women's University, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Faculty of Human Life and Environment, Nara Women's University, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Faculty of Human Life and Environment, Nara Women's University, Nara 630-8506, Japan
| |
Collapse
|
24
|
Song Y, Yang Y, Zeng W, Loor JJ, Jiang Q, Peng Z, Li Y, Jiang S, Feng X, Du X, Li X, Liu G. β-Hydroxybutyrate impairs neutrophil migration distance through activation of a protein kinase C and myosin light chain 2 signaling pathway in ketotic cows. J Dairy Sci 2021; 105:761-771. [PMID: 34635355 DOI: 10.3168/jds.2021-20875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/24/2021] [Indexed: 11/19/2022]
Abstract
Ketosis in dairy cows often occurs in the peripartal period and is accompanied by immune dysfunction. High concentrations of β-hydroxybutyrate (BHB) in peripheral blood during ketosis are closely related to the impairment of polymorphonuclear neutrophil (PMN) chemotaxis and contribute to immune dysfunction. The specific effect of BHB on PMN chemotaxis in dairy cows and the underlying molecular mechanisms are unclear. Here, 30 multiparous cows (within 3 wk postpartum) classified based on serum BHB as control (n = 15, BHB <0.6 mM) or clinically ketotic (n = 15, BHB >3.0 mM) were used. Blood samples were collected before feeding, and the isolated PMN were treated with platelet-activating factor for 0.5 h to activate their migration. Scanning electron microscopy revealed a longer tail in PMN of ketotic cows. In addition, the phosphorylation and transcription levels of myosin light chain 2 (MLC2) increased in PMN of ketotic cows. Polymorphonuclear neutrophils from control dairy cows were incubated with 3.0 mM BHB for different times in vitro, and 6 h was selected as the proper duration of BHB stimulation according to its inhibition effect on PMN migration using an under-agarose PMN chemotaxis model. Similarly, BHB stimulation in vitro resulted in inhibition of migration distance and deviation of migration direction of PMN, as well as a longer tail in morphology in the scanning electron microscope data, suggesting that BHB-induced PMN migration inhibition may be mediated by impairing the trailing edge contraction. To confirm this hypothesis, sotrastaurin (Sotra)-a specific inhibitor of protein kinase C (PKC), which is the core regulator of cell contraction-was used with or without BHB treatment in vitro. Sotra was pretreated 0.5 h before BHB treatment. Accordingly, BHB treatment increased the phosphorylation level of PKC and MLC2, the protein abundance of RhoA and rho-kinase 1 (ROCK1), and the mRNA abundance of PRKCA, MYL2, RHOA, and ROCK1 in PMN. In contrast, these effects of BHB on PMN were dampened by Sotra. As demonstrated by immunofluorescence experiments in vitro, the BHB-induced inhibition of trailing edge contraction of PMN was relieved by Sotra. In addition, Sotra also dampened the effects of BHB on PMN migration in vitro. Furthermore, as verified by in vivo experiments, compared with the control cows, both abundance and activation of PKC signaling were enhanced in PMN of ketotic cows. Overall, the present study revealed that high concentrations of blood BHB impaired PMN migration distance through inhibition of the trailing edge contraction, mediated by enhancing the activation of PKC-MLC2 signaling. These findings help explain the dysfunctional immune state in ketotic cows and provide information on the pathogenesis of infectious diseases secondary to ketosis.
Collapse
Affiliation(s)
- Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yuchen Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Wen Zeng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Zhicheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yunfei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Shang Jiang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xiancheng Feng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
25
|
Xu W, Grindler S, Dänicke S, Frahm J, Kenéz Á, Huber K. Increased plasma and milk short-chain acylcarnitine concentrations reflect systemic LPS response in mid-lactation dairy cows. Am J Physiol Regul Integr Comp Physiol 2021; 321:R429-R440. [PMID: 34318701 DOI: 10.1152/ajpregu.00072.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lipopolysaccharides (LPS) challenge the metabolic integrity of high-yielding dairy cows, activating the immune system and altering energy metabolism. Fatty acid oxidation, a major energy-gaining pathway, can be improved by supplementary carnitine, facilitating the transport of fatty acids into mitochondria. The metabolic response to the LPS challenge could alter both the plasma and the milk metabolome. Plasma and milk samples collected from cows treated with (n = 27) or without (n = 27) dietary carnitine, before and after intravenous administration of LPS, were subjected to a targeted metabolomics analysis. Multivariate statistical analyses revealed that both plasma and milk metabolome changed in response to the LPS challenge in both the carnitine-supplemented and the control cows. Short-chain acylcarnitines (carbon chain length C2, C3, C4, and C5) and long-chain acylcarnitines (C14, C16, and C18) had the highest performance to indicate LPS response when testing the predictive power of single metabolites using receiver-operator characteristics (ROC) analysis. The maximum area under a ROC curve (AUC) was 0.93. Biogenic amines, including sarcosine, and amino acids such as glutamine and isoleucine had AUC > 0.80 indicating metabolic changes due to the LPS challenge. In summary, the metabolites involved in the LPS response were acylcarnitines C2 and C5, sarcosine, glutamine, and isoleucine in plasma, and acylcarnitines C4 and C5 in milk. The interrelationship of plasma and milk metabolome included correlation of acylcarnitines C2, C4, and C5 between plasma and milk.
Collapse
Affiliation(s)
- Wei Xu
- Beijing Research Center of Intelligent Equipment for Agriculture, Beijing, People's Republic of China
| | - Sandra Grindler
- Faculty of Agricultural Sciences, Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | - Sven Dänicke
- Institute of Animal Nutrition, Federal Research Institute for Animal Health (Friedrich-Loeffler-Institute), Braunschweig, Germany
| | - Jana Frahm
- Institute of Animal Nutrition, Federal Research Institute for Animal Health (Friedrich-Loeffler-Institute), Braunschweig, Germany
| | - Ákos Kenéz
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, Hong Kong
| | - Korinna Huber
- Faculty of Agricultural Sciences, Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
26
|
Chen F, Sheng L, Xu C, Li J, Ali I, Li H, Cai Y. Ufbp1, a Key Player of Ufm1 Conjugation System, Protects Against Ketosis-Induced Liver Injury via Suppressing Smad3 Activation. Front Cell Dev Biol 2021; 9:676789. [PMID: 34307359 PMCID: PMC8297976 DOI: 10.3389/fcell.2021.676789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/02/2021] [Indexed: 01/06/2023] Open
Abstract
The dairy cattle suffer from severe liver dysfunction during the pathogenesis of ketosis. The Ufm1 conjugation system is crucial for liver development and homeostasis. Ufm1 binding protein (Ufbp1) is a putative Ufm1 target and an integral component, but its role in ketosis-induced liver injury is unclear so far. The purpose of this study is to explore the key role of Ufbp1 in liver fibrosis caused by ketosis in vivo and in vitro. Liver tissues were collected from ketotic cows and Ufbp1 conditional knockout (CKO) mice in vivo. However, Ufbp1–/– mouse embryonic fibroblast cells and Hela cells were used for in vitro validation. Subsequently, various assays were performed to reveal the underlying molecular mechanisms of the Ufbp1 protective effect. In this study, hepatic fibrosis, endoplasmic reticulum (ER) stress, and apoptosis were reported in the liver of ketotic cows, fibrotic markers (alpha-smooth muscle actin, Collagen1) and ER stress markers (glucose-regulated protein 78, CEBP homologous protein) were upregulated remarkably, and the apoptosis-related genes (Bcl2, Bax) were in line with expectations. Interestingly, Ufbp1 expression was almost disappeared, and Smad2/Smad3 protein was largely phosphorylated in the liver of ketotic cows, but Ufbp1 deletion caused Smad3 phosphorylation apparently, rather than Smad2, and elevated ER stress was observed in the CKO mice model. At the cellular level, Ufbp1 deficiency led to serious fibrotic and ER stress response, Smad3 was activated by phosphorylation significantly and then was translocated into the nucleus, whereas p-Smad2 was largely unaffected in embryonic fibroblast cells. Ufbp1 overexpression obviously suppressed Smad3 phosphorylation in Hela cells. Ufbp1 was found to be in full combination with Smad3 using endogenous immunoprecipitation. Taken together, our findings suggest that downregulation or ablation of Ufbp1 leads to Smad3 activation, elevated ER stress, and hepatocyte apoptosis, which in turn causes liver fibrosis. Ufbp1 plays a protective role in ketosis-induced liver injury.
Collapse
Affiliation(s)
- Fanghui Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Le Sheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chenjie Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jun Li
- College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Ilyas Ali
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Honglin Li
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
27
|
Li M, Yang W, Wen J, Loor JJ, Aboragah A, Wang J, Wang S, Li M, Yu L, Hou X, Xu C, Zhang B. Intracellular Ca2+ signaling and ORAI calcium release-activated calcium modulator 1 are associated with hepatic lipidosis in dairy cattle. J Anim Sci 2021; 99:skab184. [PMID: 34100951 PMCID: PMC8280943 DOI: 10.1093/jas/skab184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
Fatty liver is a common metabolic disorder afflicting dairy cows during the periparturient period and is closely associated with endoplasmic reticulum (ER) stress. The onset of ER stress in humans and mice alters hepatic lipid metabolism, but it is unknown if such event contributes to fatty liver in dairy cows soon after parturition. ORAI calcium release-activated calcium modulator 1 (ORAI1) is a key component of the store-operated Ca2+ entry mechanism regulating cellular Ca2+ balance. The purpose of this study was to investigate the role of ORAI1 on hepatic lipidosis via ER stress in dairy cows. Liver tissue biopsies were collected from Holstein cows diagnosed as healthy (n = 6) or with hepatic lipidosis (n = 6). Protein and mRNA abundance of ER stress-related targets, lipogenic targets, or the transcription regulator SREBP1 and ORAI1 were greater in cows with lipidosis. In vitro, hepatocytes were isolated from four healthy female calves and used for culture with a 1.2 mM mixture of fatty acids (oleic, linoleic, palmitic, stearic, and palmitoleic acid) for various times (0, 3, 6, 9, or 12 h). As incubation time progressed, increases in concentration of Ca2+ and abundance of protein kinase RNA-like ER kinase (PERK), inositol-requiring protein 1α (IRE1α), and activating transcription factor-6 (ATF6) protein in response to exogenous fatty acids underscored a mechanistic link among Ca2+, fatty acids, and ER stress. In a subsequent study, hepatocytes were transfected with small interfering RNA (siORAI1) or the ORAI1 inhibitor BTP2 for 48 h or 2 h followed by a challenge with the 1.2 mM mixture of fatty acids for 6 h. Compared with control group, silencing or inhibition of ORAI1 led to decreased abundance of fatty acid synthesis (FASN, SREBP1, and ACACA) and ER stress-related proteins in bovine hepatocytes. Overall, data suggested that NEFA through ORAI1 regulate intracellular Ca2+ signaling, induce ER stress, and lead to lipidosis in isolated hepatocytes.
Collapse
Affiliation(s)
- Ming Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Jianan Wen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Juan J Loor
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Ahmad Aboragah
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Jingjing Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Shuang Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Mingyang Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Liyun Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Xilin Hou
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| |
Collapse
|
28
|
Ghaffari MH, Alaedin MT, Sadri H, Hofs I, Koch C, Sauerwein H. Longitudinal changes in fatty acid metabolism and in the mitochondrial protein import system in overconditioned and normal conditioned cows: A transcriptional study using microfluidic quantitative PCR. J Dairy Sci 2021; 104:10338-10354. [PMID: 34147221 DOI: 10.3168/jds.2021-20237] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/03/2021] [Indexed: 12/22/2022]
Abstract
This study investigated the effect of body condition around calving on the hepatic mRNA expression of genes involved in fatty acid (FA) metabolism and mitochondrial protein import system of dairy cows during the transition period. Fifteen weeks before their anticipated calving date, 38 multiparous Holstein cows were selected based on their current and previous body condition scores (BCS) and allocated to either a high or a normal BCS group (19 cows each). They received different diets to reach targeted differences in BCS and backfat thickness (BFT) until dry-off. At dry-off, normal BCS (NBCS) cows had a BCS <3.5 and BFT <1.2 cm, and the high BCS (HBCS) cows had a BCS >3.75 and BFT >1.4 cm. The expression of targeted genes in the liver was assayed by reverse-transcription quantitative real-time PCR using microfluidics integrated fluidic circuit chips on a subset of 5 cows from each group. Liver biopsies were collected at d -49, +3, +21, and +84 relative to parturition. The mRNA abundance of 47 genes related to lipid metabolism including carnitine metabolism, FA uptake and transport, lipoprotein export, carnitine metabolism, mitochondrial and proximal FA oxidation, ketogenesis, AMP-activated protein kinase/mammalian target of rapamycin pathway, and mitochondrial protein import system was assessed in liver tissue. The mRNA abundances of FA binding protein (FABP)6 (in both groups), and FABP1 and solute carrier family 22 member 5 (SLC22A5) in HBCS were upregulated (>1.5-fold change, FC) in early lactation (at d +3 and +21 postpartum) compared with antepartum (d -49), indicating promoted FA uptake and intracellular transport in the liver due to the metabolic adaptations of elevated lipo-mobilization after parturition. The upregulation of SLC22A5 and SLC25A20 after parturition was more pronounced in HBCS than in NBCS cows, suggesting a need for increasing the capacity of FA uptake, and FA transport into the hepatocyte. The increased mRNA abundance of carnitine palmitoyltransferase 1A, after parturition and to a greater extent in HBCS (FC = 4.1) versus NBCS (FC = 2.1) indicates a physiological increase in the capacity of long-chain fatty acyl-CoA entry into the liver mitochondria compared with antepartum (ap; d -49 relative to calving). The greater hepatic mRNA abundance of genes encoding enzymes involved in mitochondrial FA oxidation in HBCS than in NBCS points to an increased rate of mitochondrial β-oxidation. The hepatic mRNA abundance of 3-hydroxy-3-methylglutaryl-CoA synthase 2 and 3-hydroxy-3-methylglutaryl-CoA were upregulated after parturition (d +21/d +3 pp) to a greater extent in HBCS than in NBCS cows, indicating that excess acetyl-CoA generated via β-oxidation was increasingly used for ketogenesis. We observed for the first time that the mRNA abundance of genes involved in the translocase of the inner membrane (TIM) complex (TIM22 and TIM23) in the hepatic mitochondrial protein import system were undergoing distinct changes during the transition from late pregnancy to early lactation in dairy cows. Even though sample size in this study was relatively small, the results support that overconditioning around calving may contribute to mitochondrial FA overload and greater ketogenesis at the level of transcription in the liver of early lactation cows.
Collapse
Affiliation(s)
- Morteza H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - Mohamad Taher Alaedin
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Hassan Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 516616471 Tabriz, Iran
| | - Inga Hofs
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Christian Koch
- Educational and Research Center for Animal Husbandry, Hofgut Neumuehle, 67728 Muenchweiler an der Alsenz, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
29
|
Soares RAN, Vargas G, Duffield T, Schenkel F, Squires EJ. Genome-wide association study and functional analyses for clinical and subclinical ketosis in Holstein cattle. J Dairy Sci 2021; 104:10076-10089. [PMID: 34099305 DOI: 10.3168/jds.2020-20101] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/26/2021] [Indexed: 01/01/2023]
Abstract
Ketosis is one of the most frequent metabolic diseases in high-yielding dairy cows and is characterized by high concentrations of ketone bodies in blood, urine, and milk, causing high economic losses. The search for polymorphic genes, whose alleles have different effects on resistance to developing the disease, is of extreme importance to help select less susceptible animals. The aims of this study were to identify genomic regions associated with clinical and subclinical ketosis (β-hydroxybutyrate concentration) in North American Holstein dairy cattle and to investigate these regions to identify candidate genes and metabolic pathways associated with these traits. To achieve this, a GWAS was performed for 4 traits: clinical ketosis lactation 1, clinical ketosis lactation 2 to 5, subclinical ketosis lactation 1, and subclinical ketosis lactation 2 to 5. The estimated breeding values from 77,277 cows and 7,704 bulls were deregressed and used as pseudophenotypes in the GWAS. The top-20 genomic regions explaining the largest proportion of the genetic variance were investigated for putative genes associated with the traits through functional analyses. Regions of interest were identified on chromosomes 2, 5, and 6 for clinical ketosis lactation 1; 3, 6, and 7 for clinical ketosis lactation 2 to 5; 1, 2, and 12 for subclinical ketosis lactation 1; and 20, 11, and 25 for subclinical ketosis lactation 2 to 5. The highlighted genes potentially related to clinical and subclinical ketosis included ACAT2 and IGF1. Enrichment analysis of the list of candidate genes for clinical and subclinical ketosis showed molecular functions and biological processes involved in fatty acid metabolism, lipid metabolism, and inflammatory response in dairy cattle. Several genomic regions and SNPs related to susceptibility to ketosis in dairy cattle that were previously described in other studies were confirmed. The novel genomic regions identified in this study aid to characterize the most important genes and pathways that explain the susceptibility to clinical and subclinical ketosis in dairy cattle.
Collapse
Affiliation(s)
- R A N Soares
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada N1G 2W1.
| | - G Vargas
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - T Duffield
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - F Schenkel
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - E J Squires
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| |
Collapse
|
30
|
Shi Z, Song Y, Gao X, Loor JJ, Aboragah A, Yu H, Fang Z, Zhu Y, Du X, Li X, Gao W, Liu G. Disruption of endoplasmic reticulum homeostasis exacerbates liver injury in clinically ketotic cows. J Dairy Sci 2021; 104:9130-9141. [PMID: 34001360 DOI: 10.3168/jds.2021-20238] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/10/2021] [Indexed: 12/19/2022]
Abstract
Disruption of endoplasmic reticulum (ER) homeostasis, a condition termed "ER stress," contributes to the development of liver injury in nonruminants. Because liver injury is a prominent pathological feature associated with overproduction of ketone bodies in dairy cows with ketosis, understanding the ER stress state and its functional consequences on liver injury is of particular interest. Here, 30 multiparous cows (within 3 wk postpartum) classified based on blood β-hydroxybutyrate (BHB) as healthy (n = 15, BHB <0.6 mM) or clinically ketotic (n = 15, BHB >3.0 mM) were used. Compared with healthy cows, ketotic cows had greater levels of serum fatty acids and activities of serum aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, γ-glutamyl transferase, and glutamate dehydrogenase but lower serum glucose. Furthermore, dairy cows with ketosis had greater protein abundance of ER stress markers in liver tissue, including protein kinase RNA-like ER kinase (PERK), inositol-requiring protein-1α (IRE1α), and cleaved activating transcription factor-6 (ATF6). Cows with ketosis also had higher mRNA levels of hepatic 78-kDa glucose-regulated protein (GRP78) and spliced X-box binding protein 1 (sXBP1). These data confirmed an enhanced ER stress state in clinically ketotic cows. To explore whether enhanced hepatic ER stress was induced by elevated ketone bodies and the possible contribution of ER stress to liver injury, in vitro experiments were then performed using isolated primary calf hepatocytes treated with incremental concentrations of BHB (0, 0.6, 1.2, 3.0, and 4.8 mM) for 12 h with or without overexpression of GRP78 (the master regulator of unfolded protein response). Phosphorylation levels of PERK and IRE1α proteins, level of cleaved ATF6 protein, and mRNA abundance of GRP78 and sXBP1 in hepatocytes increased after treatment with high (3.0 and 4.8 mM) BHB, indicating a mechanistic link between excessive BHB and enhanced hepatic ER stress. Furthermore, treatment with 3.0 and 4.8 mM BHB markedly elevated activities of aspartate aminotransferase and alanine aminotransferase in cell supernatant, indicating exacerbated hepatocyte damage after ER stress was enhanced. Overexpression of GRP78 attenuated both BHB-induced ER stress and the ensuing cellular damage, suggesting that hepatocyte damage caused by excessive BHB can be mediated via enhanced ER stress. Overall, the present study revealed that ER stress may exacerbate liver injury development in clinically ketotic cows, underscoring the biological relevance of this pathway in the context of liver injury.
Collapse
Affiliation(s)
- Zhen Shi
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Xinxing Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Ahmad Aboragah
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Hao Yu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Zhiyuan Fang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Yiwei Zhu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Wenwen Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China.
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
31
|
Liu Y, Wang H, Zhang M, Wang J, Zhang Z, Wang Y, Sun Y, Zhang Z. Protective effect of selenomethionine on T-2 toxin-induced liver injury in New Zealand rabbits. BMC Vet Res 2021; 17:153. [PMID: 33836763 PMCID: PMC8033731 DOI: 10.1186/s12917-021-02866-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Background T-2 toxin is a mycotoxin produced by Fusarium species that is highly toxic to animals. Recent studies have indicated that Selenomethionine (SeMet) have protective effect against mycotoxins-induced toxicity. The aim of the present study was to investigate the protective effect of SeMet on T-2-toxin-induced liver injury in rabbit and explore its molecular mechanism. Fifty rabbits (30 d, 0.5 ± 0.1 kg) were randomly divided into 5 groups: control group, T-2 toxin group, low, medium and high dose SeMet treatment group. The SeMet-treated group was orally pretreated with SeMet (containing selenium 0.2 mg/kg, 0.4 mg/kg and 0.6 mg/kg) for 21 days. On the 17th day, T-2 toxin group and SeMet-treated group were orally administered with T-2 toxin (0.4 mg/kg body weight) for 5 consecutive days. Results The results showed that low-dose SeMet significantly improved T-2 toxin-induced liver injury. We found that low-dose SeMet can reduce the level of oxidative stress and the number of hepatocyte apoptosis. Moreover, the levels of Bax, caspase-3 and caspase-9 were significantly reduced and the levels of Bcl-2 were increased. Conclusions Therefore, we confirmed that low-dose SeMet may protect rabbit hepatocytes from T-2 toxin by inhibiting the mitochondrial-caspase apoptosis pathway.
Collapse
Affiliation(s)
- Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Haojie Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Mengyu Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Jiajia Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Zhixiang Zhang
- College of Life Science, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Yuqin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China.,Engineering Research Center for Mutton Sheep Breeding of Henan Province, Luoyang, 471000, Henan, China
| | - Yingying Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China.
| |
Collapse
|
32
|
Shen T, Xu F, Fang Z, Loor JJ, Ouyang H, Chen M, Jin B, Wang X, Shi Z, Zhu Y, Liang Y, Ju L, Song Y, Wang Z, Li X, Du X, Liu G. Hepatic autophagy and mitophagy status in dairy cows with subclinical and clinical ketosis. J Dairy Sci 2021; 104:4847-4857. [PMID: 33551163 DOI: 10.3168/jds.2020-19150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/26/2020] [Indexed: 12/25/2022]
Abstract
Severe negative energy balance around parturition is an important contributor to ketosis, a metabolic disorder that occurs most frequently in the peripartal period. Autophagy and mitophagy are important processes responsible for breaking down useless or toxic cellular material, and in particular damaged mitochondria. However, the role of autophagy and mitophagy during the occurrence and development of ketosis is unclear. The objective of this study was to investigate autophagy and mitophagy in the livers of cows with subclinical ketosis (SCK) and clinical ketosis (CK). We assessed autophagy by measuring the protein abundance of microtubule-associated protein 1 light chain 3-II (LC3-II; encoded by MAP1LC3) and sequestosome-1 (p62, encoded by SQSTM1), as well as the mRNA abundance of autophagy-related genes 5 (ATG5), 7 (ATG7), and 12 (ATG12), beclin1 (BECN1), and phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3). Mitophagy was evaluated by measuring the protein abundance of the mitophagy upstream regulators PTEN-induced putative kinase 1 (PINK1) and Parkin. Liver and blood samples were collected from healthy cows [n = 15; blood β-hydroxybutyrate (BHB) concentration <1.2 mM], cows with SCK (n = 15; blood BHB concentration 1.2 to 3.0 mM) and cows with CK (n = 15; blood BHB concentration >3.0 mM with clinical signs) with similar lactation numbers (median = 3, range = 2 to 4) and days in milk (median = 6, range = 3 to 9). The serum activity of aspartate aminotransferase and alanine aminotransferase was greater in cows with CK than in healthy cows. Levels of oxidative stress biomarkers malondialdehyde and hydrogen peroxide were also higher in liver tissue from ketotic cows (SCK and CK) than from healthy cows. Compared with cows with CK and healthy cows, the hepatic mRNA abundance of MAP1LC3, SQSTM1, ATG5, ATG7, ATG12, and PIK3C3 was upregulated in cows with SCK. Compared with healthy cows, cows with SCK had a lower abundance of p62 and a greater abundance of LC3-II, but levels of both were higher in cows with CK. The mRNA abundance of ATG12 was lower in cows with CK than in healthy cows. Furthermore, the hepatic protein abundance of PINK1 and Parkin was greater in cows with SCK and slightly lower in cows with CK than in healthy cows. These data demonstrated differences in the hepatic activities of autophagy and mitophagy in cows with SCK compared with cows with CK. Although the precise mechanisms for these differences could not be discerned, autophagy and mitophagy seem to be involved in ketosis.
Collapse
Affiliation(s)
- Taiyu Shen
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Feng Xu
- Department of Renal Medicine, Second Affiliated Hospital of Jilin University, Changchun, Jilin Province, 130041, China
| | - Zhiyuan Fang
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Hongsheng Ouyang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Meng Chen
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Bo Jin
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xinghui Wang
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Zhen Shi
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yiwei Zhu
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yusheng Liang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Lingxue Ju
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yuxiang Song
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Zhe Wang
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xiliang Du
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| | - Guowen Liu
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
33
|
Soares R, Vargas G, Muniz M, Soares M, Cánovas A, Schenkel F, Squires E. Differential gene expression in dairy cows under negative energy balance and ketosis: A systematic review and meta-analysis. J Dairy Sci 2021; 104:602-615. [DOI: 10.3168/jds.2020-18883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/06/2020] [Indexed: 01/11/2023]
|
34
|
Sun X, Tang Y, Jiang C, Luo S, Jia H, Xu Q, Zhao C, Liang Y, Cao Z, Shao G, Loor JJ, Xu C. Oxidative stress, NF-κB signaling, NLRP3 inflammasome, and caspase apoptotic pathways are activated in mammary gland of ketotic Holstein cows. J Dairy Sci 2020; 104:849-861. [PMID: 33131808 DOI: 10.3168/jds.2020-18788] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022]
Abstract
Ketosis is a serious metabolic disorder characterized by systemic and hepatic oxidative stress, inflammation, and apoptosis, as well as reduced milk yield. Because of the paucity of data on mammary responses during ketosis, the aim of this study was to evaluate alterations in oxidative stress, NF-κB signaling, NLRP3 inflammasome, and caspase apoptotic pathways in mammary gland of dairy cows with ketosis. Blood, mammary gland tissue, and milk samples were collected from healthy cows [Control, blood concentration of β-hydroxybutyrate (BHB) <0.6 mM, n = 10] and cows with subclinical ketosis (SCK, blood concentration of BHB >1.2 mM and <3 mM, n = 10) or clinical ketosis (CK, blood concentration of BHB >3 mM, n = 10) at median 8 d in milk (range = 6-12). Compared with Control, serum concentration of glucose was lower (3.91 vs. 2.86 or 2.12 mM) in cows with SCK or CK, whereas concentrations of fatty acids (0.25 vs. 0.57 or 1.09 mM) and BHB (0.42 vs. 1.81 or 3.85 mM) were greater. Compared with Control, the percentage of milk fat was greater in cows with SCK or CK. In contrast, the percentage of milk protein was lower in cows with SCK or CK. We detected no differences in milk lactose content across groups. Compared with Control, activities of glutathione peroxidase, superoxide dismutase, and catalase were lower in mammary gland tissue of cows with SCK or CK. In contrast, concentrations of hydrogen peroxide and malondialdehyde were greater in cows with SCK or CK. Compared with Control, mRNA abundances of TNFA, IL6, and IL1B were greater in mammary tissues of cows with SCK or CK. In addition, activity of IKKβ and the ratio of phosphorylated inhibitor of κBα to IκBα, and of phosphorylated NF-κB p65 to NF-κB p65, were also greater in mammary tissues of cows with SCK or CK. Subclinical or clinical ketosis also led to greater activity of caspase 1 and protein abundance of caspase 1, NLRP3, Bax, caspase 3, and caspase 9. In contrast, abundance of the antiapoptotic protein was lower in SCK or CK cows. The data indicate that the mammary gland of SKC or CK cows undergoes severe oxidative stress, inflammation, and cell death.
Collapse
Affiliation(s)
- Xudong Sun
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China
| | - Yan Tang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China
| | - Chunhui Jiang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China
| | - Shengbin Luo
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China
| | - Hongdou Jia
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China
| | - Qiushi Xu
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China
| | - Chenxu Zhao
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China
| | - Yusheng Liang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guang Shao
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar, Heilongjiang Province 161000, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Chuang Xu
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, China.
| |
Collapse
|
35
|
Yang Q, Han B, Xue J, Lv Y, Li S, Liu Y, Wu P, Wang X, Zhang Z. Hexavalent chromium induces mitochondrial dynamics disorder in rat liver by inhibiting AMPK/PGC-1α signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114855. [PMID: 32474337 DOI: 10.1016/j.envpol.2020.114855] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
Occupational exposure to hexavalent chromium (Cr(VI)) can cause cytotoxicity and carcinogenicity. In this study, we established a liver injury model in rats via intraperitoneal injection of potassium dichromate (0, 2, 4, and 6 mg/kg body weight) for 35 d to investigate the mechanism of Cr(VI)-induced liver injury. We found that Cr(VI) induced hepatic histopathological lesions, oxidative stress, and apoptosis and reduced the expression of mitochondrial-related regulatory factors such as adenosine 5'-monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in a dose-dependent manner. Furthermore, Cr(VI) promoted mitochondrial division and inhibited fusion, leading to increased expression of caspase-3 and production of mitochondrial reactive oxygen species. Our study demonstrates that long-term exposure to Cr(VI) induces mitochondrial dynamics disorder by inhibiting AMPK/PGC-1α signaling pathway in rat liver.
Collapse
Affiliation(s)
- Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China
| | - Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Jiangdong Xue
- College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, 028000, China
| | - Yueying Lv
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Yan Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Xiaoqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China.
| |
Collapse
|
36
|
Shen J, Sun B, Yu C, Cao Y, Cai C, Yao J. Choline and methionine regulate lipid metabolism via the AMPK signaling pathway in hepatocytes exposed to high concentrations of nonesterified fatty acids. J Cell Biochem 2020; 121:3667-3678. [PMID: 31680310 DOI: 10.1002/jcb.29494] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/08/2019] [Indexed: 01/24/2023]
Abstract
High concentrations of nonesterified fatty acids (NEFAs) and β-hydroxybutyric acid (BHBA) induce lipid peroxidation, resulting in liver damage. Choline and methionine (Met) can promote energy balance and benefit liver health in transition dairy cows; however, the regulating mechanism remains unclear. In the present study, we established the hepatocyte damage model by 1.5 mM NEFAs or BHBA treatment, and examined lipid metabolism in hepatocytes. The results showed that 1.5 mM NEFAs and 1.5 mM BHBA significantly decreased the messenger RNA (mRNA) expression of AMP-activated protein kinase (AMPK)-α as well as its target genes carnitine palmitoyltransferase-1α (CPT-1α), acetyl-CoA carboxylase, fatty acid synthetase, and Apolipoprotein B100 (ApoB100). Choline and Met upregulated the phosphorylation level of AMPK-α, which was blocked by BML (an AMPK-α inhibitor). The mRNA expression level of peroxisome proliferator-activated receptor-α (PPAR-α), CPT-1α, and ApoB100 showed a similar trend. The expressions of liver X recptoer α (LXR-α) and sterol regulatory element-binding protein 1c (SREBP-1c) were decreased by choline and Met, while only the decrease of LXR-α was blocked by BML. These findings indicate that the high-level NEFAs and BHBA weaken the lipid metabolism by impairing the fatty acid oxidation, synthesis, and transport proteins. Choline and Met regulate PPAR-α and LXR-α transcriptional activity through AMPK-α phosphorylation and regulate SREBP-1c independently of AMPK-α to promote lipid oxidation and transport in NEFAs-treated hepatocytes.
Collapse
Affiliation(s)
- Jing Shen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Bofei Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chao Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.,The Development Centre of Animal Husbandry, Shangluo, Shaanxi, China
| | - Yangchun Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chuanjiang Cai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
37
|
Han B, Lv Z, Zhang X, Lv Y, Li S, Wu P, Yang Q, Li J, Qu B, Zhang Z. Deltamethrin induces liver fibrosis in quails via activation of the TGF-β1/Smad signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 259:113870. [PMID: 31918140 DOI: 10.1016/j.envpol.2019.113870] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 06/10/2023]
Abstract
Deltamethrin (DLM) is an important member of the pyrethroid pesticide family, and its widespread use has led to serious environmental and health problems. Exposure to DLM causes pathological changes in the liver of animals and humans and can lead to liver fibrosis. However, the mechanism of DLM-induced liver fibrosis remains unclear. Therefore, to address its potential molecular mechanisms, we used both in vivo and in vitro methods. Quails were treated in vivo by intragastric administration of different concentrations of DLM (0, 15, 30, or 45 mg kg-1), and the chicken liver cancer cell line LMH was treated in vitro with various doses of DLM (0, 50, 200, or 800 μg mL-1). We found that DLM treatment in vivo induced liver fibrosis in a dose-dependent manner through the promotion of oxidative stress, activation of transforming growth factor-β1 (TGF-β1) and phosphorylation of Smad2/3. Treatment of LMH cells with different concentrations of DLM similarly induced oxidative stress and also decreased cell viability. Collectively, our study demonstrates that DLM-induced liver fibrosis in quails occurs via activation of the TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, 150030, China
| | - Xiaoya Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yueying Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bing Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, 150030, China.
| |
Collapse
|
38
|
Li X, Li G, Du X, Sun X, Peng Z, Zhao C, Xu Q, Abdelatty A, Mohamed F, Wang Z, Liu G. Increased autophagy mediates the adaptive mechanism of the mammary gland in dairy cows with hyperketonemia. J Dairy Sci 2020; 103:2545-2555. [DOI: 10.3168/jds.2019-16910] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 10/16/2019] [Indexed: 12/16/2022]
|
39
|
Baimishev M, Eremin S, Plemyashov K, Baimishev H, Konopeltsev I, Yesengaliev K. The effect of metabolism on the course of labor and the postpartum period in highly productive cows. BIO WEB OF CONFERENCES 2020. [DOI: 10.1051/bioconf/20201700256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The purpose of the research is to determine the etiopathogenesis of reproductive dysfunction in highly productive cows. For this, one group of cows was formed on the principle of paranalogs in the amount of 37 animals inseminated in the first sexual hunt after calving, followed by taking blood samples from them using the Monovet system, considering the duration of pregnancy. During the start-up period, blood was taken 1–4 days before calving and on the first day after calving. A total of 253 blood samples were examined. Subsequently, depending on the effectiveness of insemination, animals were divided into two groups. The first group included inseminated cows after the first insemination (20 animals), the second group included 17 unfertilized cows after the first insemination. Subsequently, blood was taken from animals considering the course of childbirth and the postpartum period. Blood counts were studied according to generally accepted methods using certified equipment. The study found that at an early stage of pregnancy, cows have a significant difference in lipid metabolism and in their peroxidation, in the state of antioxidant systems compared to unstable animals. In the process of pregnancy development in cows, there is a decrease in the level of total lipids and their class, and the accumulation of products of transoxidation of lipids is reduced. In animals with retention of the placenta, a low lipid metabolism and a higher level of peroxidation were established already in the dry period. After calving, this difference increases. The obtained data can be used to develop an algorithm for the prevention of postpartum complications in cows by using substances with antioxidant properties.
Collapse
|
40
|
Li T, Li X, Meng H, Chen L, Meng F. ACSL1 affects Triglyceride Levels through the PPARγ Pathway. Int J Med Sci 2020; 17:720-727. [PMID: 32218693 PMCID: PMC7085263 DOI: 10.7150/ijms.42248] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
In clinical cohort studies, high expression of long-chain acyl-coenzyme A synthetases 1 (ACSL1 gene) in peripheral white blood cells of patients with acute myocardial infarction (AMI) has been utilized as molecular markers of myocardial infarction diagnosis. The plasma triglyceride level of AMI patients is significantly higher than that of healthy individuals. We hypothesized that the high expression of ACSL1 increases the level of triglyceride, which is one of the pathogenesis of AMI promoted by ACSL1. In this report, cell culture based methods were adopted to test the hypothesis and further investigate the effect and mechanism of ACSL1 on lipid metabolism. In this study, liver cells of healthy individuals were cultured, the overexpression and the knockdown vectors of ACSL1 were constructed and transfected into liver cells. The transfection was verified at the mRNA and protein level. Intracellular triglyceride content was quantitatively analyzed using ELISA. Changes of genes related to lipid metabolism were subsequently measured through PCR array. Overexpression of ACSL1 led to higher gene expression and protein levels compared to control and the triglyceride content was significantly increased in overexpressing cells. The expression level of fatty acid oxidation pathway PPARγ was significantly down-regulated compared with the control group, as were genes associated with fatty acid synthesis pathways: SREBP1, ACC, FAS, and SCD1. ACSL1 knockdown decreased the content of triglyceride whereas PPARγ was up-regulated and SREBP1, ACC, FAS, and SCD1 were down-regulated compared with the control group. In summary, high expression of ACSL1 reduced fatty acid β-oxidation through the PPARγ pathway, thereby increasing triglyceride levels.
Collapse
Affiliation(s)
| | | | | | | | - Fanbo Meng
- Department of Cardiology China-Japan Union Hospital of Jilin University, Changchun, China 130033
| |
Collapse
|
41
|
Wu WY, Ding XQ, Gu TT, Guo WJ, Jiao RQ, Song L, Sun Y, Pan Y, Kong LD. Pterostilbene Improves Hepatic Lipid Accumulation via the MiR-34a/Sirt1/SREBP-1 Pathway in Fructose-Fed Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1436-1446. [PMID: 31927917 DOI: 10.1021/acs.jafc.9b04259] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
High fructose intake promotes hepatic lipid accumulation. Pterostilbene, a natural analogue of resveratrol found in diet berries, exhibits a hepatoprotective property. Here, we studied the protection by pterostilbene against fructose-induced hepatic lipid accumulation and explored its possible mechanism. We observed a high expression of microRNA-34a (miR-34a, P < 0.05) and a low expression of its target, sirtuin1 (Sirt1, mRNA: P < 0.01; protein: P < 0.001), with the overactivation of downstream sterol regulatory element-binding protein-1 (SREBP-1) lipogenic pathway (nuclear SREBP-1 protein: P < 0.05; FAS and SCD1 mRNA: P < 0.01), in rat livers, as well as BRL-3A and HepG2 cells, stimulated by fructose. More interestingly, pterostilbene recovered the fructose-disturbed miR-34a expression (0.3-0.5-fold vs fructose control, P < 0.05), Sirt1 protein level (1.2- to 1.5-fold vs fructose control, P < 0.05), and SREBP-1 lipogenic pathway, resulting in significant amelioration of hepatocyte lipid accumulation in animal [hepatic triglyceride and total cholesterol (TG&TC) mg/g·wet tissue: 4.90 ± 0.19, 5.23 ± 0.16, 5.20 ± 0.29 vs fructose control 9.73 ± 1.06, P < 0.001; 3.18 ± 0.30, 3.31 ± 0.39, 3.37 ± 0.47 vs 5.67 ± 0.28, P < 0.001] and cell models (BRL-3A TG&TC mmol/g·protein: 0.123 ± 0.011 vs 0.177 ± 0.004, P < 0.001; 0.169 ± 0.011 vs 0.202 ± 0.008, P < 0.05; HepG2: 0.257 ± 0.005 vs 0.303 ± 0.016, P < 0.05; 0.143 ± 0.004 vs 0.201 ± 0.008, P < 0.001). These results provide the experimental evidence supporting the anti-lipogenic effect of pterostilbene against fructose-induced hepatic lipid accumulation via modulating the miR-34a/Sirt1/SREBP-1 pathway.
Collapse
Affiliation(s)
- Wen-Yuan Wu
- State Key Laboratory of Pharmaceutical Biotechnology , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Xiao-Qin Ding
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Ting-Ting Gu
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Wen-Jie Guo
- State Key Laboratory of Pharmaceutical Biotechnology , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Rui-Qing Jiao
- State Key Laboratory of Pharmaceutical Biotechnology , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Lin Song
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Ying Pan
- State Key Laboratory of Pharmaceutical Biotechnology , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| | - Ling-Dong Kong
- State Key Laboratory of Pharmaceutical Biotechnology , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
- School of Life Sciences , Nanjing University , Nanjing 210023 , Jiangsu Province , P. R. China
| |
Collapse
|
42
|
|
43
|
Shahzad K, Lopreiato V, Liang Y, Trevisi E, Osorio JS, Xu C, Loor JJ. Hepatic metabolomics and transcriptomics to study susceptibility to ketosis in response to prepartal nutritional management. J Anim Sci Biotechnol 2019; 10:96. [PMID: 31867104 PMCID: PMC6918647 DOI: 10.1186/s40104-019-0404-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
Background Ketosis in dairy cows is associated with body fat mobilization during the peripartal period. Sub-clinical and clinical ketosis arise more frequently in cows that are overfed energy during the entire dry (last 50 to 45 days prior to parturition) or close-up period (last ~ 28 days prepartum). Methods A retrospective analysis was performed on 12 cows from a larger cohort that were fed a higher-energy diet [1.54 Mcal/kg of dry matter (DM); 35.9% of DM corn silage and 13% of DM ground corn] during the close-up dry period, of which 6 did not develop clinical ketosis (OVE, 0.83 mmol/L plasma hydroxybutyrate; BHB) and 6 were diagnosed with clinical ketosis (KET, 1.4 mmol/L BHB) during the first week postpartum. A whole-transcriptome bovine microarray (Agilent Technologies) and metabolomics (GC-MS, LC-MS; Metabolon® Inc.) were used to perform transcript and metabolite profiling of liver tissue harvested at − 10 days relative to parturition which allowed to establish potential associations between prepartal transcriptome/metabolome profiles and susceptibility to clinical ketosis postpartum. Results Cows in KET had greater (P = 0.01) overall body weight between − 2 and 1 week around parturition, but similar body condition score than OVE. Although dry matter intake (DMI) did not differ prepartum, KET cows had lower (P < 0.01) DMI and similar milk yield as OVE cows during the first week postpartum. Transcriptome analysis revealed a total of 3065 differentially expressed genes (DEG; P ≤ 0.05) in KET. Metabolomics identified 15 out of 313 total biochemical compounds significantly affected (P ≤ 0.10) in KET. Among those, greater concentrations (P ≤ 0.06, + 2.3-fold) of glycochenodeoxycholate in KET cows also have been detected in humans developing non-alcoholic fatty liver disease. Bioinformatics analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database and the DEG revealed that, among the top 20 most-impacted metabolic pathway categories in KET, 65% were overall downregulated. Those included ‘Metabolism of cofactors and vitamins’, ‘Biosynthesis of other secondary metabolites’, ‘Lipid’, ‘Carbohydrate’, and ‘Glycan biosynthesis and metabolism’. The lower relative concentration of glucose-6-phosphate and marked downregulation of fructose-1,6-bisphosphatase 2 and pyruvate dehydrogenase kinase 4 support a strong impairment in gluconeogenesis in prepartal liver of cows developing KET postpartum. Among the top 20 most-impacted non-metabolic pathways, 85% were downregulated. Pathways such as ‘mTOR signalling’ and ‘Insulin signalling’ were among those. ‘Ribosome’, ‘Nucleotide excision repair’, and ‘Adherens junctions’ were the only upregulated pathways in cows with KET. Conclusions The combined data analyses revealed more extensive alterations of the prepartal liver transcriptome than metabolome in cows overfed energy and developing ketosis postpartum. The causative link between these tissue-level adaptations and onset of clinical ketosis needs to be studied further.
Collapse
Affiliation(s)
- Khuram Shahzad
- 1COMSATS Institute of Information Technology, ChakShahzad, Islamabad, 44000 Pakistan.,2Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Vincenzo Lopreiato
- 3Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Yusheng Liang
- 2Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Erminio Trevisi
- 3Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Johan S Osorio
- 4Department of Dairy Science, South Dakota State University, Brookings, SD 57006 USA
| | - Chuang Xu
- 5College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Xinyang Rd. 5, Daqing, 163319 China
| | - Juan J Loor
- 2Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| |
Collapse
|
44
|
Lopreiato V, Vailati-Riboni M, Bellingeri A, Khan I, Farina G, Parys C, Loor J. Inflammation and oxidative stress transcription profiles due to in vitro supply of methionine with or without choline in unstimulated blood polymorphonuclear leukocytes from lactating Holstein cows. J Dairy Sci 2019; 102:10395-10410. [DOI: 10.3168/jds.2019-16413] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/28/2019] [Indexed: 01/06/2023]
|
45
|
Li S, Baiyun R, Lv Z, Li J, Han D, Zhao W, Yu L, Deng N, Liu Z, Zhang Z. Exploring the kidney hazard of exposure to mercuric chloride in mice:Disorder of mitochondrial dynamics induces oxidative stress and results in apoptosis. CHEMOSPHERE 2019; 234:822-829. [PMID: 31247492 DOI: 10.1016/j.chemosphere.2019.06.096] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/25/2019] [Accepted: 06/12/2019] [Indexed: 06/09/2023]
Abstract
Mercury is one of the 10 toxic chemicals with major public health concerns. Continuous exposure to low levels of heavy metals including mercury is related to renal injury, especially in children. This study investigated the possible molecular mechanism of inorganic mercury-induced kidney injury. Twenty eight Kunming mice were divided into four groups (n = 7), and treated with 0, 20, 40, 80 mg/L mercuric chloride (HgCl2) in drinking water for 16 weeks respectively. All the HgCl2 exposure mice displayed different degrees of renal injury, which was diagnosed by hematoxylin and eosin stain, biochemical analysis, and ultrastructure examination. The treatment of HgCl2 inhibited the silent information regulator two ortholog 1 (Sirt1)/peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) signaling pathway and resulted the disorder of mitochondrial dynamics, as evidenced by the increasing expression of dynamin-related protein 1 and decreasing expression of mitofusin 2. Meanwhile, HgCl2 inhibited the nuclear factor erythroid 2-related factor 2 (Nrf2) axis. The abnormality of mitochondrial dynamics and the suppression of Nrf2 axis exacerbated oxidative stress, and then induced cell apoptosis. These findings demonstrated that the disorder of mitochondrial dynamics induced by HgCl2 activated oxidative stress, and further resulted in renal apoptosis through inhibiting the Sirt1/PGC-1α signaling pathway and the Nrf2 axis.
Collapse
Affiliation(s)
- Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Ruiqi Baiyun
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Dongxu Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Wenyue Zhao
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Lanjie Yu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Ning Deng
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhiyan Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin 150030, China.
| |
Collapse
|
46
|
Alharthi AS, Coleman DN, Liang Y, Batistel F, Elolimy AA, Yambao RC, Abdel-Hamied E, Pan YX, Parys C, Alhidary IA, Abdelrahman MM, Loor JJ. Hepatic 1-carbon metabolism enzyme activity, intermediate metabolites, and growth in neonatal Holstein dairy calves are altered by maternal supply of methionine during late pregnancy. J Dairy Sci 2019; 102:10291-10303. [PMID: 31477291 DOI: 10.3168/jds.2019-16562] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 07/10/2019] [Indexed: 01/24/2023]
Abstract
Maternal supply of methyl donors such as methionine (Met) during late pregnancy can affect offspring growth and development. The objective was to investigate the effect of postruminal Met supply during late pregnancy on 1-carbon, Met cycle, and transsulfuration pathways in the calf liver. During the last 28 d of pregnancy, cows were individually fed a control diet or the control diet plus rumen-protected dl-Met (MET; 0.09% dry matter intake). Liver samples obtained from calves (n = 14/group) at 4, 14, 28, and 50 d of age were used for metabolomics, real-time PCR, and enzyme activity analyses. Genes associated with 1-carbon metabolism, DNA methylation, and the cytidine 5'-diphosphocholine-choline pathway were analyzed via real-time PCR. Activity of betaine homocysteine methyltransferase, cystathionine β-synthase, and 5-methyltetrahydrofolate homocysteine methyltransferase (MTR) was analyzed using 14C isotopes. Data were analyzed using a mixed model that included the fixed effects of maternal treatment, day, and their interaction, and the random effect was calf within maternal diet. Calves born to dams offered MET tended to have greater birth body weight and had overall greater body weight during the first 9 wk of life. However, no differences were detected for daily feed intake and average daily gain between groups. Concentrations of betaine and choline, reflecting Met cycle activity, at d 14 through 28 were greater in MET calves. Transsulfuration pathway intermediates also were altered in MET calves, with concentrations of cysteine sulfinic acid and hypotaurine (d 4 and 14) and taurine being greater (d 4, 14, 28, and 50). Despite the lack of differences in daily feed intake, the greater concentrations of the tricarboxylic acid cycle intermediates fumarate and glutamate along with NAD/NADH in MET calves indicated enhanced rates of energy metabolism. Although activity of betaine homocysteine methyltransferase was greater in MET calves at d 14, cystathionine β-synthase was lower and increased at d 14 and 28, where it was greater compared with the control diet. Activity of MTR was lower at d 4 and 50 in MET calves. Among gene targets measured, MET calves had greater overall expression of MTR, phosphatidylethanolamine N-methyltransferase, and choline kinase α and β. An interaction of maternal diet by time was detected for mRNA abundance of DNA methyltransferase 3α (involved in de novo methylation) due to greater values at d 4 and 14 in MET calves. Overall, the data indicate that enhanced postruminal supply of Met to cows during late pregnancy may program hepatic metabolism of the calf in the context of maintaining Met homeostasis, phosphatidylcholine and taurine synthesis, DNA methylation, and energy metabolism. These alterations potentially result in better efficiency of nutrient use, hence conferring the calf a physiologic advantage during a period of rapid growth and development. The precise biologic mechanisms remain to be established.
Collapse
Affiliation(s)
- A S Alharthi
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801; Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - D N Coleman
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Y Liang
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - F Batistel
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan 84322-4815
| | - A A Elolimy
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - R C Yambao
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - E Abdel-Hamied
- Animal Medicine Department, Beni-Suef University, Beni-Suef, Egypt 62511
| | - Y-X Pan
- Food Science and Human Nutrition, Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - C Parys
- Evonik Nutrition and Care GmbH, Hanau-Wolfgang, Germany 63457
| | - I A Alhidary
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - M M Abdelrahman
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
47
|
Alharthi AS, Lopreiato V, Dai H, Bucktrout R, Abdelmegeid M, Batistel F, Parys C, Shen X, Ballou MA, Trevisi E, Alhidary IA, Abdelrahman MM, Loor JJ. Short communication: Supply of methionine during late pregnancy enhances whole-blood innate immune response of Holstein calves partly through changes in mRNA abundance in polymorphonuclear leukocytes. J Dairy Sci 2019; 102:10599-10605. [PMID: 31447163 DOI: 10.3168/jds.2018-15676] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 07/02/2019] [Indexed: 12/18/2022]
Abstract
The supply of methionine (Met) in late pregnancy can alter mRNA abundance of genes associated with metabolism and immune response in liver and polymorphonuclear leukocytes (PMN) of the neonatal calf. Whether prenatal supply of Met elicits postnatal effects on systemic inflammation and innate immune response of the calf is not well known. We investigated whether enhancing the maternal supply of Met via feeding ethyl-cellulose rumen-protected Met (RPM) was associated with differences in calf innate immune response mRNA abundance in PMN and systemic indicators of inflammation during the first 50 d of life. Calves (n = 14 per maternal diet) born to cows fed RPM at 0.09% of diet dry matter per day (MET) for the last 28 ± 2 d before calving or fed a control diet with no added Met (CON) were used. Blood for biomarker analysis and isolation of PMN for innate immune function assays and mRNA abundance was harvested at birth (before colostrum feeding) and at 7, 21 and 50 d of age. Whole blood was challenged with enteropathogenic bacteria (Escherichia coli 0118:H8) and phagocytosis and oxidative burst of neutrophils and monocytes were quantified via flow cytometry. Although concentration of haptoglobin and activity of myeloperoxidase among calves from both maternal groups increased markedly between 0 and 7 d of age followed by a decrease to baseline at d 21 the responses were lower in MET compared with CON calves. Nitric oxide concentration decreased markedly between 0 and 7 d regardless of maternal group but MET calves tended to have lower overall concentrations during the study. In vitro phagocytosis in stimulated neutrophils increased markedly over time in both CON and MET calves but responses were overall greater in MET calves. Oxidative burst in both neutrophils and monocytes increased over time regardless of maternal treatment. The mRNA abundance of lactate dehydrogenase (LDHA) signal transducer and activator of transcription 3 (STAT3) and S100 calcium binding protein A8 (S100A8) in PMN was overall greater in MET calves. Overall data suggest that increasing the maternal supply of Met during late pregnancy could affect the neonatal calf inflammatory status and innate immune response. Although changes in mRNA abundance could play a role in coordinating the immune response the exact mechanisms merit further study.
Collapse
Affiliation(s)
- A S Alharthi
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801; Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - V Lopreiato
- Department of Animal Sciences, Food and Nutrition, Università Cattolica del Sacro Cuore, Piacenza 29122, Italy
| | - H Dai
- Department of Clinical Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - R Bucktrout
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - M Abdelmegeid
- Department of Animal Medicine, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt
| | - F Batistel
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan 84322
| | - C Parys
- Evonik Nutrition and Care GmbH, 63457 Hanau-Wolfgang, Germany
| | - X Shen
- Department of Clinical Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - M A Ballou
- Department of Animal Sciences, Texas Tech University, Lubbock 79409
| | - E Trevisi
- Department of Animal Sciences, Food and Nutrition, Università Cattolica del Sacro Cuore, Piacenza 29122, Italy
| | - I A Alhidary
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - M M Abdelrahman
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - J J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
48
|
Han B, Li S, Lv Y, Yang D, Li J, Yang Q, Wu P, Lv Z, Zhang Z. Dietary melatonin attenuates chromium-induced lung injury via activating the Sirt1/Pgc-1α/Nrf2 pathway. Food Funct 2019; 10:5555-5565. [PMID: 31429458 DOI: 10.1039/c9fo01152h] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Exposure to chromium (Cr) causes a number of respiratory diseases, including lung cancer and pulmonary fibrosis. However, there is currently no safe treatment for Cr-induced lung damage. Here, we used in vivo and in vitro approaches to examine the protective effects of melatonin (MEL) on Cr-induced lung injury and to identify the underlying molecular mechanisms. We found that treatment of rats or a mouse lung epithelial cell MLE-12 with MEL attenuated K2Cr2O7-induced lung injury by reducing the production of oxidative stress and inflammatory mediators and inhibiting cell apoptosis. MEL treatment upregulated the expression of silent information regulator 1 (Sirt1), which deacetylated the transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator-1α (Pgc-1α). In turn, this increased the expression of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and key anti-oxidant target genes. These results suggest that melatonin attenuates chromium-induced lung injury via activating the Sirt1/Pgc-1α/Nrf2 pathway. Dietary MEL supplement may be a potential new strategy for the treatment of Cr poisoning.
Collapse
Affiliation(s)
- Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Yueying Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Daqian Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China. and Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China. and Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, 150030, China
| |
Collapse
|
49
|
Wang C, Zhang Y, Deng M, Wang X, Tu W, Fu Z, Jin Y. Bioaccumulation in the gut and liver causes gut barrier dysfunction and hepatic metabolism disorder in mice after exposure to low doses of OBS. ENVIRONMENT INTERNATIONAL 2019; 129:279-290. [PMID: 31146162 DOI: 10.1016/j.envint.2019.05.056] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 05/23/2023]
Abstract
The compound sodium ρ-perfluorous nonenoxybenzene sulfonate (OBS), a new kind of perfluoroalkyl and polyfluoroalkyl compound, is a surfactant for increasing oil production, and it has been widely detected in various organisms. Because of its wide use, OBS is detectable in the environment. However, knowledge about the biological toxicity of OBS to animals is very limited. Here, male mice were exposed to 0, 0.1, 1 or 10 μg/L of OBS for 6 weeks via drinking water. It was demonstrated that OBS was highly bioaccumulated both in the liver and gut in the mice after low doses of OBS exposure. Curiously, a low dose of OBS exposure also caused gut barrier dysfunction by decreasing mucus secretion and altering Ionic transport in the gut via the CFTR pathway. In addition, liver function was influenced by OBS at both the histopathological and physiological levels. Hepatic transcriptomics and metabolomics analysis showed a total of 1157 genes, and multiple metabolites changed significantly in the livers of mice exposed to low-dose OBS for 6 weeks. The functions of these changed genes and metabolites are tightly related to glycolysis, fatty acid synthesis, fatty acid transport, and β-oxidation. All these results indicate that the liver and gut are important target tissues for OBS exposure. Importantly, it is possible that high levels of bioaccumulation of OBS in the gut and liver might directly cause gut barrier dysfunction and hepatic metabolism disorder in mice.
Collapse
Affiliation(s)
- Caiyun Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yi Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Mi Deng
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China
| | - Xia Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China.
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
50
|
Yang X, Zhang X, Zhang J, Ji Q, Huang W, Zhang X, Li Y. Spermatogenesis disorder caused by T-2 toxin is associated with germ cell apoptosis mediated by oxidative stress. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 251:372-379. [PMID: 31091501 DOI: 10.1016/j.envpol.2019.05.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/22/2019] [Accepted: 05/05/2019] [Indexed: 06/09/2023]
Abstract
T-2 toxin is an unavoidable contaminant in human food, animal feeds, and agricultural products. T-2 toxin has been found to impair male reproductive function. But, few data is available that reveals the reproductive toxicity mechanism. In the study, male Kunming mice were orally administrated with T-2 toxin at the doses of 0, 0.5, 1 or 2 mg/kg body weight for 28 days. The body and reproductive organs weight, the concentration, malformation rate and ultrastructure of sperm in cauda epididymis were detected. Oxidative stress biomarkers and apoptosis were also measured in testes. Histological change of testes was performed by H&E and TUNEL staining. T-2 toxin down-regulated body and reproductive organs (testis, epididymis and seminal vesicle) weight, sperm concentration, increased sperm malformation rate and damaged the ultrastructure of sperm and structure of testes. T-2 toxin treatment increased the reactive oxygen species (ROS) and malondialdehyde content, while, decreased the total anti-oxidation capacity (T-AOC) and the superoxide dismutase activity in testes. T-2 toxin exposure increased the TUNEL-positive germ cells, the activities and mRNA expressions of caspase-3, caspase-8 and caspase-9, the mRNA expression of Bax, and inhibited the Bcl-2 mRNA expression. Furthermore, the expressions of caspase-3, caspase-8 caspase-9 and Bax were positively correlated with ROS level, but negatively correlated with T-AOC in testis. In summary, T-2 toxin caused spermatogenesis disorder associated with the germ cell apoptosis medicated by oxidative stress, impairing the male reproductive function.
Collapse
Affiliation(s)
- Xu Yang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Xuliang Zhang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Jian Zhang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Qiang Ji
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Wanyue Huang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China
| | - Xueyan Zhang
- Northeast Agricultural University Hospital, Northeast Agricultural University, Harbin, 150030, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|