1
|
Irawan A, Bionaz M. Liver Transcriptomic Profiles of Ruminant Species Fed Spent Hemp Biomass Containing Cannabinoids. Genes (Basel) 2024; 15:963. [PMID: 39062742 PMCID: PMC11275923 DOI: 10.3390/genes15070963] [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: 06/29/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The inclusion of spent hemp biomass (SHB), an extracted byproduct from industrial cannabidiol (CBD) production, in the diets of dairy cows and lambs appears to be safe with minor effects on the metabolism, including a decrease in circulating cholesterol and increase bilirubinemia, both associated with liver metabolism. Those effects could be consequence of the presence of cannabinoids, particularly Δ9-tetrahydrocannabinol (THC) and CBD in the SHB. This study aimed to study the transcriptional profile of the liver of dairy cows and lambs fed SHB. Dairy cows received SHB or alfalfa pellet for four weeks of intervention (IP) and four weeks of withdrawal periods (WP). Finishing lambs were fed a control diet (CON), 10% (LH2), or 20% (HH2) SHB for 2 months or 1 month followed by 1-month SHB withdrawal (LH1 and HH1, respectively). RNA sequencing was performed, and the mRNA was annotated using the latest reference genomes. The RNAseq data were filtered, normalized for library size and composition, and statistically analyzed by DESeq2. The bioinformatic analysis was performed by using DAVID, Gene Set Enrichment Analysis (GSEA), and the Dynamic Impact Approach. Using a 0.2 FDR cut-off, we identified only ≤24 differentially expressed genes (DEG) in the liver by feeding SHB in dairy cows and a larger number of DEGs in lambs (from 71 in HH1 vs. CON to 552 in LH1 vs. CON). The KEGG analysis demonstrated that feeding SHB in dairy cows and lambs had relatively minor to moderate metabolic alterations in dairy cows and lambs mainly associated with amino acids and lipid metabolism whereas cholesterol synthesis was overall activated in lambs. GSEA identified activation of the PPAR signaling pathway only in dairy cows. We found an opposite effect on activation of metabolism of drug and xenobiotics by cytochrome P450 enzymes in dairy cows and lambs receiving less SHB but an inhibition in HH2 lambs. Immune system-related pathways were inhibited by feeding SHB in lambs, but the impact was minor. Cumulatively, inclusion of SHB containing cannabinoids in dairy and lambs demonstrate very little effects on the alteration of transcriptomic profile of the liver.
Collapse
Affiliation(s)
- Agung Irawan
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA;
- Animal Science Study Program, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA;
| |
Collapse
|
2
|
Wang S, Ren H, Qin C, Su J, Song X, Li R, Cui K, Liu Y, Shi D, Liu Q, Li Z. A Characterization and Functional Analysis of Peroxisome Proliferator-Activated Receptor Gamma Splicing Variants in the Buffalo Mammary Gland. Genes (Basel) 2024; 15:779. [PMID: 38927715 PMCID: PMC11203352 DOI: 10.3390/genes15060779] [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: 04/10/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARG) has various splicing variants and plays essential roles in the regulation of adipocyte differentiation and lipogenesis. However, little is known about the expression pattern and effect of the PPARG on milk fat synthesis in the buffalo mammary gland. In this study, we found that only PPARG-X17 and PPARG-X21 of the splicing variant were expressed in the buffalo mammary gland. Amino acid sequence characterization showed that the proteins encoded by PPARG-X17 and PPARG-X21 are endonuclear non-secreted hydrophilic proteins. Protein domain prediction found that only the PPARG-X21-encoded protein had PPAR ligand-binding domains (NR_LBD_PPAR), which may lead to functional differences between the two splices. RNA interference (RNAi) and the overexpression of PPARG-X17 and PPARG-X21 in buffalo mammary epithelial cells (BMECs) were performed. Results showed that the expression of fatty acid synthesis-related genes (ACACA, CD36, ACSL1, GPAT, AGPAT6, DGAT1) was significantly modified (p < 0.05) by the RNAi and overexpression of PPARG-X17 and PPARG-X21. All kinds of FAs detected in this study were significantly decreased (p < 0.05) after RNAi of PPARG-X17 or PPARG-X21. Overexpression of PPARG-X17 or PPARG-X21 significantly decreased (p < 0.05) the SFA content, while significantly increased (p < 0.05) the UFA, especially the MUFA in the BMECs. In conclusion, there are two PPARG splicing variants expressed in the BMECs that can regulate FA synthesis by altering the expression of diverse fatty acid synthesis-related genes. This study revealed the expression characteristics and functions of the PPARG gene in buffalo mammary glands and provided a reference for further understanding of fat synthesis in buffalo milk.
Collapse
Affiliation(s)
- Shuwan Wang
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| | - Honghe Ren
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| | - Chaobin Qin
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| | - Jie Su
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| | - Xinhui Song
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| | - Ruijia Li
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| | - Kuiqing Cui
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China; (K.C.); (Q.L.)
| | - Yang Liu
- Guangxi Zhuang Autonomous Region Center for Analysis and Test Research, Nanning 530022, China
| | - Deshun Shi
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China; (K.C.); (Q.L.)
| | - Zhipeng Li
- Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (S.W.); (H.R.); (C.Q.); (J.S.); (X.S.); (R.L.); (D.S.)
| |
Collapse
|
3
|
Skibiel AL. Hepatic mitochondrial bioenergetics and metabolism across lactation and in response to heat stress in dairy cows. JDS COMMUNICATIONS 2024; 5:247-252. [PMID: 38646582 PMCID: PMC11026913 DOI: 10.3168/jdsc.2023-0432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/01/2023] [Indexed: 04/23/2024]
Abstract
Lactation is energetically demanding for the dairy cow. Numerous morphological and metabolic changes orchestrated across different tissues in the body partition nutrients for milk synthesis. The liver is a key organ coordinating modifications in metabolism that increase substrate availability for the mammary gland. Impaired capacity to make the needed physiological adjustments for lactation, such as occurs with heat stress, can result in metabolic disease and poor lactation performance. At the cellular level, increases in mitochondrial density and bioenergetic and biosynthetic capacity are critical adaptations for successful lactation, providing energy and substrates for milk synthesis. Mitochondria are also involved in coordinating adaptation to a variety of stressors by providing the metabolic foundation to enlist a stress response. Heat stress can damage mitochondrial structures and impair mitochondrial function, with implications for pathogenesis and production. This systematic review focuses on the hepatic mitochondrial adaptations to lactation and the mitochondrial responses to heat stress. Future research directions are also discussed that may lead to improvements in managing the metabolic needs of the lactating cow and diminishing the adverse production and health consequences from environmental stress.
Collapse
Affiliation(s)
- Amy L. Skibiel
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID 83844
| |
Collapse
|
4
|
Abebe BK, Wang H, Li A, Zan L. A review of the role of transcription factors in regulating adipogenesis and lipogenesis in beef cattle. J Anim Breed Genet 2024; 141:235-256. [PMID: 38146089 DOI: 10.1111/jbg.12841] [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: 09/30/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023]
Abstract
In the past few decades, genomic selection and other refined strategies have been used to increase the growth rate and lean meat production of beef cattle. Nevertheless, the fast growth rates of cattle breeds are often accompanied by a reduction in intramuscular fat (IMF) deposition, impairing meat quality. Transcription factors play vital roles in regulating adipogenesis and lipogenesis in beef cattle. Meanwhile, understanding the role of transcription factors in regulating adipogenesis and lipogenesis in beef cattle has gained significant attention to increase IMF deposition and meat quality. Therefore, the aim of this paper was to provide a comprehensive summary and valuable insight into the complex role of transcription factors in adipogenesis and lipogenesis in beef cattle. This review summarizes the contemporary studies in transcription factors in adipogenesis and lipogenesis, genome-wide analysis of transcription factors, epigenetic regulation of transcription factors, nutritional regulation of transcription factors, metabolic signalling pathways, functional genomics methods, transcriptomic profiling of adipose tissues, transcription factors and meat quality and comparative genomics with other livestock species. In conclusion, transcription factors play a crucial role in promoting adipocyte development and fatty acid biosynthesis in beef cattle. They control adipose tissue formation and metabolism, thereby improving meat quality and maintaining metabolic balance. Understanding the processes by which these transcription factors regulate adipose tissue deposition and lipid metabolism will simplify the development of marbling or IMF composition in beef cattle.
Collapse
Affiliation(s)
- Belete Kuraz Abebe
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
- Department of Animal Science, Werabe University, Werabe, Ethiopia
| | - Hongbao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Anning Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| |
Collapse
|
5
|
Castro-Navarro I, McGuire MA, Williams JE, Holdsworth EA, Meehan CL, McGuire MK. Maternal Cannabis Use during Lactation and Potential Effects on Human Milk Composition and Production: A Narrative Review. Adv Nutr 2024; 15:100196. [PMID: 38432590 PMCID: PMC10997876 DOI: 10.1016/j.advnut.2024.100196] [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: 12/18/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
Cannabis use has increased sharply in the last 20 y among adults, including reproductive-aged women. Its recent widespread legalization is associated with a decrease in risk perception of cannabis use during breastfeeding. However, the effect of cannabis use (if any) on milk production and milk composition is not known. This narrative review summarizes current knowledge related to maternal cannabis use during breastfeeding and provides an overview of possible pathways whereby cannabis might affect milk composition and production. Several studies have demonstrated that cannabinoids and their metabolites are detectable in human milk produced by mothers who use cannabis. Due to their physicochemical properties, cannabinoids are stored in adipose tissue, can easily reach the mammary gland, and can be secreted in milk. Moreover, cannabinoid receptors are present in adipocytes and mammary epithelial cells. The activation of these receptors directly modulates fatty acid metabolism, potentially causing changes in milk fatty acid profiles. Additionally, the endocannabinoid system is intimately connected to the endocrine system. As such, it is probable that interactions of exogenous cannabinoids with the endocannabinoid system might modify release of critical hormones (e.g., prolactin and dopamine) that regulate milk production and secretion. Nonetheless, few studies have investigated effects of cannabis use (including on milk production and composition) in lactating women. Additional research utilizing robust methodologies are needed to elucidate whether and how cannabis use affects human milk production and composition.
Collapse
Affiliation(s)
- Irma Castro-Navarro
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States.
| | - Mark A McGuire
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Janet E Williams
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | | | - Courtney L Meehan
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - Michelle K McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| |
Collapse
|
6
|
Guo Y, Wei Z, Zhang Y, Cao J. Research Progress on the Mechanism of Milk Fat Synthesis in Cows and the Effect of Conjugated Linoleic Acid on Milk Fat Metabolism and Its Underlying Mechanism: A Review. Animals (Basel) 2024; 14:204. [PMID: 38254373 PMCID: PMC10812695 DOI: 10.3390/ani14020204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Milk fat synthesis in cows mainly includes the synthesis of short- and medium-chain fatty acids, the uptake, transport, and activation of long-chain fatty acids (LCFAs), the synthesis of triglycerides, and the synthesis of the genes, transcription factors, and signaling pathways involved. Although the various stages of milk fat synthesis have been outlined in previous research, only partial processes have been revealed. CLA consists of an aggregation of positional and geometric isomers of linoleic fatty acid, and the accumulated evidence suggests that the two isomers of the active forms of CLA (cis-9, trans-11 conjugated linoleic acid and trans-10, cis-12 conjugated linoleic acid, abbreviated as c9, t11-CLA and t10, c12-CLA) can reduce the fat content in milk by regulating lipogenesis, fatty acid (FA) uptake, oxidation, and fat synthesis. However, the mechanism through which CLA inhibits milk fat synthesis is unique, with most studies focusing only on the effects of CLA on one of the genes, transcription factors, or signaling pathways involved. In this study, we summarized the structure and function of classic genes and pathways (mTOR, SREBP, AMPK, and PPARG) and new genes or pathways (THRSP, METTL3, ELOVL, and LPIN1) involved in each stage of milk fat synthesis and demonstrated the interactions between genes and pathways. We also examined the effects of other substances (melanin, nicotinic acid, SA, etc.). Furthermore, we evaluated the influence of β-sitosterol, sodium butyrate, Met arginine, and Camellia oleifera Abel on milk fat synthesis to improve the mechanism of milk fat synthesis in cows and provide a mechanistic reference for the use of CLA in inhibiting milk fat biosynthesis.
Collapse
Affiliation(s)
- Yuanyin Guo
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.G.); (Z.W.)
| | - Ziang Wei
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.G.); (Z.W.)
| | - Yi Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Jie Cao
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (Y.G.); (Z.W.)
| |
Collapse
|
7
|
Zeng L, Zhou J, Zhang Y, Wang X, Li Y, Song J, Shao J, Su P. Paternal cadmium exposure induces glucolipid metabolic reprogramming in offspring mice via PPAR signaling pathway. CHEMOSPHERE 2023; 339:139592. [PMID: 37482320 DOI: 10.1016/j.chemosphere.2023.139592] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
In industrialized societies, the prevalence of metabolic diseases has substantially increased over the past few decades, yet the underlying causes remain unclear. Cadmium (Cd) is a hazardous heavy metal and pervasive environmental endocrine disruptor. Here, we investigate the effects of paternal Cd exposure on offspring glucolipid metabolism. Paternal Cd exposure (1 mg kg-1 body weight) impaired glucose tolerance, increased random serum glucose and fasting serum insulin, elevated serum total cholesterol, and low-density lipoprotein in offspring mice. Untargeted metabolomics analysis of male offspring liver tissue revealed that paternal Cd exposure can affect offspring glucolipid metabolic reprogramming, which involved biosynthesis of phenylalanine, tyrosine and tryptophan, biosynthesis of unsaturated fatty acids, metabolism of linoleic acid, arachidonic acid and α-linolenic acid. Transcriptome sequencing of male offspring liver tissue showed that arachidonic acid metabolism, AMPK signaling pathway, PPAR signaling pathway and adipocytokine signaling pathway were significantly inhibited in the Cd-exposed group. The mRNA expression levels of PPAR signaling pathway related genes (Acsl1, Cyp4a14, Cyp4a10, Cd36, Ppard and Pck1) were significantly decreased. The protein expression levels of ACSL1, CD36, PPARD and PCK1 were also significantly reduced. Collectively, our findings suggest that paternal Cd exposure affect offspring glucolipid metabolic reprogramming via PPAR signaling pathway.
Collapse
Affiliation(s)
- Ling Zeng
- Medical Genetics Center, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, PR China; Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Jinzhao Zhou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Yanwei Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Xiaofei Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Yamin Li
- Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, PR China.
| | - Jieping Song
- Medical Genetics Center, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, PR China.
| | - JingFan Shao
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Ping Su
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
8
|
Torrecilhas JA, Pereira GL, Vito ES, Fiorentini G, Ramirez-Zamudio GD, Fonseca LS, Torres RDNS, Simioni TA, Duarte JM, Machado Neto OR, Curi RA, Chardulo LAL, Baldassini WA, Berchielli TT. Changes in the Lipid Metabolism of the Longissimus thoracis Muscle in Bulls When Using Different Feeding Strategies during the Growing and Finishing Phases. Metabolites 2023; 13:1042. [PMID: 37887367 PMCID: PMC10608670 DOI: 10.3390/metabo13101042] [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: 08/14/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023] Open
Abstract
The objective was to evaluate the supplementation strategy's effect on beef cattle during the growing phase and two systems during the finishing phase. One hundred and twenty young bulls were randomly divided in a 2 × 2 factorial design to receive either mineral (ad libitum) or protein + energy (3 g/kg body weight (BW)/day) during the growing phase and pasture plus concentrate supplementation (20 g/kg BW/day) or feedlot (25:75% corn silage:concentrate) during the finishing phase. Feedlot-fed bulls had meat (Longissimus thoracis-LT) with a higher content of lipids and saturated and monounsaturated fatty acids and a greater upregulation of stearoyl-CoA desaturase and sterol regulatory element-binding protein-1c than animals that fed on pasture (p < 0.05). On the other hand, pasture-fed bulls had meat with a higher content of α-linoleic acid, linolenic acid, and n6 and a greater n6:n3 ratio compared to the feedlot-fed group (p < 0.05). In addition, meat from pasture-fed bulls during the finishing phase had 17.6% more isocitrate dehydrogenase enzyme concentration than the feedlot group (p = 0.02). Mineral-fed and pasture-finished bulls showed down-regulation of peroxisome proliferator-activated receptor gamma (p < 0.05), while the bulls fed protein + energy and finished in the feedlot had higher carnitine palmitoyltransferase 2 expression (p ≤ 0.013). In conclusion, mineral or protein + energy supplementation in the growing does not affect the fatty acid composition of intramuscular fat of LT muscle. In the finishing phase, feeding bulls in the feedlot upregulates the lipogenic genes and consequently improves the intramuscular fat content in the meat.
Collapse
Affiliation(s)
- Juliana Akamine Torrecilhas
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Guilherme Luis Pereira
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Elias San Vito
- Confina Beef Cattle Consulting, Sinop 78555-603, MT, Brazil;
| | - Giovani Fiorentini
- Department of Animal Science, Federal University of Pelotas (UFPEL), Pelotas 96160-000, RS, Brazil;
| | - Germán Darío Ramirez-Zamudio
- College of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga 13635-900, SP, Brazil;
| | - Larissa Simielli Fonseca
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| | - Rodrigo de Nazaré Santos Torres
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Tiago Adriano Simioni
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| | - Juliana Messana Duarte
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| | - Otavio Rodrigues Machado Neto
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Rogério Abdallah Curi
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Luis Artur Loyola Chardulo
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Welder Angelo Baldassini
- School of Veterinary e Animal Science (FMVZ), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (J.A.T.); (R.d.N.S.T.); (O.R.M.N.); (R.A.C.); (L.A.L.C.); (W.A.B.)
| | - Telma Teresinha Berchielli
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (Unesp), Jaboticabal 14884-900, SP, Brazil; (L.S.F.); (T.A.S.); (J.M.D.); (T.T.B.)
| |
Collapse
|
9
|
Naomi R, Teoh SH, Halim S, Embong H, Hasain Z, Bahari H, Kumar J. Unraveling Obesity: Transgenerational Inheritance, Treatment Side Effects, Flavonoids, Mechanisms, Microbiota, Redox Balance, and Bioavailability-A Narrative Review. Antioxidants (Basel) 2023; 12:1549. [PMID: 37627544 PMCID: PMC10451614 DOI: 10.3390/antiox12081549] [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: 05/10/2023] [Revised: 07/02/2023] [Accepted: 07/06/2023] [Indexed: 08/27/2023] Open
Abstract
Obesity is known as a transgenerational vicious cycle and has become a global burden due to its unavoidable complications. Modern approaches to obesity management often involve the use of pharmaceutical drugs and surgeries that have been associated with negative side effects. In contrast, natural antioxidants, such as flavonoids, have emerged as a promising alternative due to their potential health benefits and minimal side effects. Thus, this narrative review explores the potential protective role of flavonoids as a natural antioxidant in managing obesity. To identify recent in vivo studies on the efficiency of flavonoids in managing obesity, a comprehensive search was conducted on Wiley Online Library, Scopus, Nature, and ScienceDirect. The search was limited to the past 10 years; from the search, we identified 31 articles to be further reviewed. Based on the reviewed articles, we concluded that flavonoids offer novel therapeutic strategies for preventing obesity and its associated co-morbidities. This is because the appropriate dosage of flavonoid compounds is able to reduce adipose tissue mass, the formation of intracellular free radicals, enhance endogenous antioxidant defences, modulate the redox balance, and reduce inflammatory signalling pathways. Thus, this review provides an insight into the domain of a natural product therapeutic approach for managing obesity and recapitulates the transgenerational inheritance of obesity, the current available treatments to manage obesity and its side effects, flavonoids and their sources, the molecular mechanism involved, the modulation of gut microbiota in obesity, redox balance, and the bioavailability of flavonoids. In toto, although flavonoids show promising positive outcome in managing obesity, a more comprehensive understanding of the molecular mechanisms responsible for the advantageous impacts of flavonoids-achieved through translation to clinical trials-would provide a novel approach to inculcating flavonoids in managing obesity in the future as this review is limited to animal studies.
Collapse
Affiliation(s)
- Ruth Naomi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Soo Huat Teoh
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Malaysia;
| | - Shariff Halim
- Faculty of Health Sciences, University Technology Mara (UiTM) Pulau Pinang, Bertam Campus, Kepala Batas 13200, Malaysia;
| | - Hashim Embong
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Zubaidah Hasain
- Unit of Physiology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur 57000, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
10
|
Bai H, Zhang M, Zhao Y, Wang R, Zhang G, Lambo MT, Zhang Y, Li Y, Wang L. Altering the ratio of palmitic, stearic, and oleic acids in dietary fat affects nutrient digestibility, plasma metabolites, growth performance, carcass, meat quality, and lipid metabolism gene expression of Angus bulls. Meat Sci 2023; 199:109138. [PMID: 36796287 DOI: 10.1016/j.meatsci.2023.109138] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
This study evaluated the effects of changing the ratio of palmitic, stearic, and oleic acids in dietary fat on nutritional metabolism, growth performance, and meat quality of finishing Angus bulls. Bulls received the following three treatments: (1) a control diet without fat supplement (CON), (2) CON + mixed fatty acid supplement (58% C16:0 + 28% cis-9 C18:1; MIX), (3) CON + saturated fatty acid supplement (87% C16:0 + 10% C18:0; SFA). In summary, both fat treatment diets simultaneously increased saturated fatty acids C16:0 (P = 0.025), C18:0 (P < 0.001) and total monounsaturated fatty acids (P = 0.008) in muscle, thus balancing the ratio of unsaturated to saturated fatty acids in muscle. MIX diet increased the digestibility of dry matter (P = 0.014), crude protein (P = 0.038), and ether extract (P = 0.036). SFA diet increased the daily gain (P = 0.032) and intramuscular fat content (P = 0.043). The high content of C16:0 and C18:0 in the SFA diet promoted weight gain and fat deposition of beef cattle by increasing feed intake, up-regulating the expression of lipid uptake genes and increasing deposition of total fatty acids, resulting in better growth performance and meat quality.
Collapse
Affiliation(s)
- Haixin Bai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Meimei Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yufan Zhao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Ruixue Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Guangning Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Modinat Tolani Lambo
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yonggen Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yang Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Liang Wang
- Research Institute of Applied Technologies, Honghe University, Mengzi 661199, China.
| |
Collapse
|
11
|
Zhang J, Gaowa N, Wang Y, Li H, Cao Z, Yang H, Zhang X, Li S. Complementary hepatic metabolomics and proteomics reveal the adaptive mechanisms of dairy cows to the transition period. J Dairy Sci 2023; 106:2071-2088. [PMID: 36567250 DOI: 10.3168/jds.2022-22224] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022]
Abstract
The transition period from late pregnancy to early lactation is a vital time of the lifecycle of dairy cows due to the marked metabolic challenges. Besides, the liver is the pivot point of metabolism in cattle. Nevertheless, the hepatic physiological molecular adaptation during the transition period has not been elucidated, especially from the metabolomics and proteomics view. Therefore, the present study aims to investigate the hepatic metabolic alterations in transition cows by using integrative metabolomics and proteomics methods. Gas chromatography quadrupole-time-of-flight mass spectrometry-based metabolomics and data-independent acquisition-based quantitative proteomics methods were used to analyze liver tissues collected from 8 healthy multiparous Holstein dairy cows 21 d before and after calving. In total, 44 metabolites and 250 proteins were identified as differentially expressed from 233 metabolites and 3,539 proteins detected from the liver biopsies during the transition period. Complementary functional analysis of different metabolites and proteins indicated the upregulated gluconeogenesis, tricarboxylic acid cycles, AA degradation, fatty acid oxidation, AMP-activated protein kinase signaling pathway, peroxisome proliferator-activated receptor signaling pathway, and ribosome proteins in postpartum dairy cows. In terms of the metabolites and proteins, glucose-6-phosphate, fructose-6-phosphate, carnitine palmitoyltransferase 1A, and phosphoenolpyruvate carboxykinase played a significant role in these pathways. The upregulated oxidative status may be accompanied by the pathways mentioned above. In addition, the upregulated glucagon and insulin signaling pathways also indicated the significant requirement for glucose in postpartum dairy cows. These outcomes, from the view of global metabolites and proteins, may present a better comprehension of the biology of the transition period, which can be helpful in further developing nutritional regulation strategies targeting the liver to help cows overcome this metabolically challenging time.
Collapse
Affiliation(s)
- Jun Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100 China; State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Naren Gaowa
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Yajing Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Huanxu Li
- Beijing Oriental Kingherd Biotechnology Company, Beijing 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Hongjian Yang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Xiaoming Zhang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China.
| |
Collapse
|
12
|
Ramírez-Zamudio GD, Ganga MJG, Pereira GL, Nociti RP, Chiaratti MR, Cooke RF, Chardulo LAL, Baldassini WA, Machado-Neto OR, Curi RA. Effect of Cow-Calf Supplementation on Gene Expression, Processes, and Pathways Related to Adipogenesis and Lipogenesis in Longissimus thoracis Muscle of F1 Angus × Nellore Cattle at Weaning. Metabolites 2023; 13:metabo13020160. [PMID: 36837780 PMCID: PMC9962728 DOI: 10.3390/metabo13020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
The aim of this study was to identify differentially expressed genes, biological processes, and metabolic pathways related to adipogenesis and lipogenesis in calves receiving different diets during the cow-calf phase. Forty-eight uncastrated F1 Angus × Nellore males were randomly assigned to two treatments from thirty days of age to weaning: no creep feeding (G1) or creep feeding (G2). The creep feed offered contained ground corn (44.8%), soybean meal (40.4%), and mineral core (14.8%), with 22% crude protein and 65% total digestible nutrients in dry matter. After weaning, the animals were feedlot finished for 180 days and fed a single diet containing 12.6% forage and 87.4% corn-based concentrate. Longissimus thoracis muscle samples were collected by biopsy at weaning for transcriptome analysis and at slaughter for the measurement of intramuscular fat content (IMF) and marbling score (MS). Animals of G2 had 17.2% and 14.0% higher IMF and MS, respectively (p < 0.05). We identified 947 differentially expressed genes (log2 fold change 0.5, FDR 5%); of these, 504 were upregulated and 443 were downregulated in G2. Part of the genes upregulated in G2 were related to PPAR signaling (PPARA, SLC27A1, FABP3, and DBI), unsaturated fatty acid synthesis (FADS1, FADS2, SCD, and SCD5), and fatty acid metabolism (FASN, FADS1, FADS2, SCD, and SCD5). Regarding biological processes, the genes upregulated in G2 were related to cholesterol biosynthesis (EBP, CYP51A1, DHCR24, and LSS), unsaturated fatty acid biosynthesis (FADS2, SCD, SCD5, and FADS1), and insulin sensitivity (INSIG1 and LPIN2). Cow-calf supplementation G2 positively affected energy metabolism and lipid biosynthesis, and thus favored the deposition of marbling fat during the postweaning period, which was shown here in an unprecedented way, by analyzing the transcriptome, genes, pathways, and enriched processes due to the use of creep feeding.
Collapse
Affiliation(s)
| | - Maria Júlia Generoso Ganga
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
| | - Guilherme Luis Pereira
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Ricardo Perecin Nociti
- College of Animal Science and Food Engineering, São Paulo University (USP), Pirassununga 13635-900, SP, Brazil
| | - Marcos Roberto Chiaratti
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCAR), São Carlos 13565-905, SP, Brazil
| | | | - Luis Artur Loyola Chardulo
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Welder Angelo Baldassini
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Otávio Rodrigues Machado-Neto
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Rogério Abdallah Curi
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
- Correspondence:
| |
Collapse
|
13
|
Khan MZ, Liu S, Ma Y, Ma M, Ullah Q, Khan IM, Wang J, Xiao J, Chen T, Khan A, Cao Z. Overview of the effect of rumen-protected limiting amino acids (methionine and lysine) and choline on the immunity, antioxidative, and inflammatory status of periparturient ruminants. Front Immunol 2023; 13:1042895. [PMID: 36713436 PMCID: PMC9878850 DOI: 10.3389/fimmu.2022.1042895] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Overproduction of reactive oxygen species (ROS) is a well-known phenomenon experienced by ruminants, especially during the transition from late gestation to successful lactation. This overproduction of ROS may lead to oxidative stress (OS), which compromises the immune and anti-inflammatory systems of animals, thus predisposing them to health issues. Besides, during the periparturient period, metabolic stress is developed due to a negative energy balance, which is followed by excessive fat mobilization and poor production performance. Excessive lipolysis causes immune suppression, abnormal regulation of inflammation, and enhanced oxidative stress. Indeed, OS plays a key role in regulating the metabolic activity of various organs and the productivity of farm animals. For example, rapid fetal growth and the production of large amounts of colostrum and milk, as well as an increase in both maternal and fetal metabolism, result in increased ROS production and an increased need for micronutrients, including antioxidants, during the last trimester of pregnancy and at the start of lactation. Oxidative stress is generally neutralized by the natural antioxidant system in the body. However, in some special phases, such as the periparturient period, the animal's natural antioxidant system is unable to cope with the situation. The effect of rumen-protected limiting amino acids and choline on the regulation of immunity, antioxidative, and anti-inflammatory status and milk production performance, has been widely studied in ruminants. Thus, in the current review, we gathered and interpreted the data on this topic, especially during the perinatal and lactational stages.
Collapse
Affiliation(s)
- Muhammad Zahoor Khan
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China,Faculty of Veterinary and Animal Sciences, the University of Agriculture, Dera Ismail Khan, Pakistan
| | - Shuai Liu
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yulin Ma
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mei Ma
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qudrat Ullah
- Faculty of Veterinary and Animal Sciences, the University of Agriculture, Dera Ismail Khan, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Jingjun Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jianxin Xiao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tianyu Chen
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Adnan Khan
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China,*Correspondence: Zhijun Cao,
| |
Collapse
|
14
|
Roque-Jiménez JA, Oviedo-Ojeda MF, Whalin M, Lee-Rangel HA, Relling AE. Ewe early gestation supplementation with eicosapentaenoic and docosahexaenoic acids affects the liver, muscle, and adipose tissue fatty acid profile and liver mRNA expression in the offspring. J Anim Sci 2023; 101:skad144. [PMID: 37158288 PMCID: PMC10263116 DOI: 10.1093/jas/skad144] [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: 12/08/2022] [Accepted: 05/05/2023] [Indexed: 05/10/2023] Open
Abstract
Our objectives were to assess the effects of eicosapentaenoic (EPA) and docosahexaenoic acids (DHA) supplementation to pregnant ewes during the first third of gestation on their offspring's liver, adipose, and muscle tissues fatty acid (FA) profile and liver mRNA expression after a finishing period receiving diets with different FA profiles. Twenty-four post-weaning lambs, blocked by sex and body weight, were used in a 2 × 2 factorial arrangement of treatments. The first factor was dam supplementation (DS) in the first third of gestation with 1.61% of Ca salts of palm fatty acid distillate (PFAD) or Ca salts enriched with EPA-DHA. Ewes were exposed to rams with marking paint harnesses during the breeding. Ewes started DS at the day of mating, considered day 1 of conception. Twenty-eight days after mating, ultrasonography was used to confirm pregnancy, and nonpregnant ewes were removed from the groups. After weaning, the offspring lambs were supplemented (LS, second main factor) with two different FA sources (1.48% of PFAD or 1.48% of EPA-DHA) during the growing and fattening phase. Lambs were fed the LS diet for 56 d and sent to slaughter, where the liver, muscle, and adipose tissue samples were collected for FA analysis. Liver samples were collected for relative mRNA expression for genes associated with FA transport and metabolism. The data were analyzed as a mixed model in SAS (9.4). In the liver, the amount of C20:5 and C22:6 (P < 0.01) increased in lambs with LS-EPA-DHA, while some C18:1 cis FA isomers were greater in the lambs from DS-PFAD. In muscle, amounts of C22:1, C20:5, and C22:5 increased (P < 0.05) in lambs born from DS-EPA-DHA. The adipose tissue amounts of C20:5, C22:5, and C22:6 were greater (P < 0.01) in lambs from LS-EPA-DHA. Interactions (DS × LS; P < 0.05) were observed for DNMT3β, FABP-1, FABP-5, SCD, and SREBP-1; having greater mRNA expression in liver tissue of LS-EPA-DHA, DS-PFAD and LS-PFAD, DS-EPA-DHA lambs compared with the lambs in the other two treatments. Liver ELOVL2 mRNA relative expression (P < 0.03) was greater in the offspring of DS-PFAD. Relative mRNA expression (P < 0.05) of GLUT1, IGF-1, LPL, and PPARγ increased in the liver from LS-EPA-DHA lambs. Dam supplementation during early gestation using with different FA sources changed the lipid FA profile in MT, LT, and SAT during the finishing period depending on the tissue and type of FA source administered during the growing phase.
Collapse
Affiliation(s)
- José A Roque-Jiménez
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA
- Universidad Autónoma de San Luis Potosí, Facultad de Agronomía y Veterinaria, San Luis Potosí 78175, México
| | - Mario F Oviedo-Ojeda
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA
- Universidad Autónoma de San Luis Potosí, Facultad de Agronomía y Veterinaria, San Luis Potosí 78175, México
| | - Megan Whalin
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA
| | - Héctor A Lee-Rangel
- Universidad Autónoma de San Luis Potosí, Facultad de Agronomía y Veterinaria, San Luis Potosí 78175, México
| | - Alejandro E Relling
- Department of Animal Sciences, The Ohio State University, Ohio Agricultural Research and Development Center (OARDC), Wooster, OH 44691, USA
| |
Collapse
|
15
|
Padilha CG, Ribeiro CVDM, Oliveira DE. Milk fatty acids as covariates in multiple regression analysis is a robust approach to model the decrease in milk fat concentration and yield in small ruminants. J Anim Physiol Anim Nutr (Berl) 2022. [DOI: 10.1111/jpn.13792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 10/09/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Charline G. Padilha
- Department of Animal Production Santa Catarina State University Lages Santa Catarina Brazil
| | | | - Dimas E. Oliveira
- Department of Animal Production Santa Catarina State University Lages Santa Catarina Brazil
| |
Collapse
|
16
|
Cheng J, Xu D, Chen L, Guo W, Hu G, Liu J, Fu S. CIDEA Regulates De Novo Fatty Acid Synthesis in Bovine Mammary Epithelial Cells by Targeting the AMPK/PPARγ Axis and Regulating SREBP1. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11324-11335. [PMID: 36040348 DOI: 10.1021/acs.jafc.2c05226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cell-death-inducing DNA fragmentation factor-α-like effector A (CIDEA) is a lipid-droplet-associated protein that helps to promote lipid metabolism in adipocytes of mice and humans. However, studies on the regulatory mechanism of CIDEA on lipid metabolism in the mammary glands of dairy cows are rare. Therefore, the role of CIDEA in bovine mammary epithelial cells (bMECs) was investigated in this study. The CIDEA expression levels in the mammary glands of high-fat-milk-producing cows were significantly higher compared to those in low-fat-milk-producing cows. Results of in vitro studies in bMECs showed that the inhibition of CIDEA inhibited the expression of fatty acid synthesis-related genes and triglyceride (TAG) synthesis-related genes. Conversely, the overexpression of CIDEA leads to an increase in the content of TAG and fatty acid. The results of mechanistic studies indicated that the overexpression of CIDEA inhibits AMP-activated protein kinase (AMPK) activity, which enhances the expression of peroxisome proliferator-activated receptor-γ (PPARγ) and consequently increases the TAG content. Furthermore, the overexpression of CIDEA promoted the nuclear translocation of sterol regulatory element-binding protein 1 (SREBP1). Therefore, a theoretical framework is provided by this study for the regulation of lipid metabolism in dairy cows by means of nutrition and the hormone targeting of CIDEA.
Collapse
Affiliation(s)
- Ji Cheng
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin130062, China
| | - Dianwen Xu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin130062, China
| | - Lisha Chen
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin130062, China
| | - Wenjin Guo
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin130062, China
| | - Guiqiu Hu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin130062, China
| | - Juxiong Liu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin130062, China
| | - Shoupeng Fu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin130062, China
| |
Collapse
|
17
|
Jin X, Zhen Z, Wang Z, Gao X, Li M. GPRC6A is a key mediator of palmitic acid regulation of lipid synthesis in bovine mammary epithelial cells. Cell Biol Int 2022; 46:1747-1758. [PMID: 35979663 DOI: 10.1002/cbin.11886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/29/2021] [Accepted: 03/07/2022] [Indexed: 11/07/2022]
Abstract
Fatty acids (FAs) can promote lipid synthesis in the mammary gland via stimulating lipogenic gene expression, but the underlying molecular mechanism is still not fully understood. Here, we showed the dose-dependent effects of palmitic acid (PA) on lipid synthesis in primary bovine mammary epithelial cells (BMECs) and explored the corresponding molecular mechanism. BMECs were treated with PA (0, 50, 100, 150, and 200 μM), and the 100 μM treatment had the best stimulatory effect on lipid synthesis and expression and maturation of sterol regulatory element-binding protein 1c (SREBP-1c) in cells. Inhibition of phosphatidylinositol 3-kinase (PI3K) almost totally blocked the stimulation of PA on SREBP-1c expression, whereas protein kinase Cα (PKCα) knockdown only partially decreased the stimulation of PA on SREBP-1c expression but abolished the stimulation of PA on its maturation. Knockdown of GPR120 did not change the stimulation of PA on the SREBP-1c signaling. G protein-coupled receptor family C group 6 member A (GPRC6A) knockdown almost totally blocked the stimulation of FA on PI3K and PKCα phosphorylation as well as SREBP-1c expression and maturation. Furthermore, PA dose-dependently promoted GPRC6A expression and plasma membrane localization. Together, these above results reveal that GPRC6A is a key mediator of PA signaling to lipid synthesis in BMECs via the PI3K/PKCα-SREBP-1c pathways.
Collapse
Affiliation(s)
- Xin Jin
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhen Zhen
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhaoxiong Wang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Meng Li
- College of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
18
|
Impacts of Circadian Gene Period2 Knockout on Intestinal Metabolism and Hepatic Antioxidant and Inflammation State in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7896371. [PMID: 35910841 PMCID: PMC9325607 DOI: 10.1155/2022/7896371] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/19/2022] [Accepted: 06/17/2022] [Indexed: 11/26/2022]
Abstract
The period circadian regulator 2 (Per2) gene is important for the modulations of rhythmic homeostasis in the gut and liver; disruption will cause metabolic diseases, such as obesity, diabetes, and fatty liver. Herein, we investigated the alterations in intestinal metabolic and hepatic functions in Per2 knockout (Per2−/−, KO) and wild-type (Per2+/+, WT) mice. Growth indices, intestinal metabolomics, hepatic circadian rhythms, lipid metabolism, inflammation-related genes, antioxidant capacity, and transcriptome sequencing were performed after euthanasia. Data indicated that KO decreased the intestinal concentrations of amino acids such as γ-aminobutyric acid, aspartic acid, glycine, L-allothreonine, methionine, proline, serine, and valine while it increased the concentrations of carbohydrates such as cellobiose, D-talose, fucose, lyxose, and xylose compared with WT. Moreover, the imbalance of intestinal metabolism further seemed to induce liver dysfunction. Data indicated that Per2 knockout altered the expression of hepatic circadian rhythm genes, such as Clock, Bmal1, Per1, Per3, Cry1, and Cry2. KO also induced hepatic lipid metabolism, because of the increase of liver index and serum concentrations of low-density lipoprotein, and the upregulated expression of Pparα, Cyp7a1, and Cpt1. In addition, KO improved hepatic antioxidant capacity due to the increase activities of SOD and GSH-Px and the decrease in concentrations of MDA. Lastly, KO increased the relative expression levels of hepatic inflammation-related genes, such as Il-1β, Il-6, Tnf-α, Myd88, and Nf-κB p65, which may potentially lead to hepatic inflammation. Overall, Per2 knockout induces gut metabolic dysregulation and may potentially trigger alterations in hepatic antioxidant and inflammation responses.
Collapse
|
19
|
Busato S, Ford HR, Abdelatty AM, Estill CT, Bionaz M. Peroxisome Proliferator-Activated Receptor Activation in Precision-Cut Bovine Liver Slices Reveals Novel Putative PPAR Targets in Periparturient Dairy Cows. Front Vet Sci 2022; 9:931264. [PMID: 35903133 PMCID: PMC9315222 DOI: 10.3389/fvets.2022.931264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolic challenges experienced by dairy cows during the transition between pregnancy and lactation (also known as peripartum), are of considerable interest from a nutrigenomic perspective. The mobilization of large amounts of non-esterified fatty acids (NEFA) leads to an increase in NEFA uptake in the liver, the excess of which can cause hepatic accumulation of lipids and ultimately fatty liver. Interestingly, peripartum NEFA activate the Peroxisome Proliferator-activated Receptor (PPAR), a transcriptional regulator with known nutrigenomic properties. The study of PPAR activation in the liver of periparturient dairy cows is thus crucial; however, current in vitro models of the bovine liver are inadequate, and the isolation of primary hepatocytes is time consuming, resource intensive, and prone to errors, with the resulting cells losing characteristic phenotypical traits within hours. The objective of the current study was to evaluate the use of precision-cut liver slices (PCLS) from liver biopsies as a model for PPAR activation in periparturient dairy cows. Three primiparous Jersey cows were enrolled in the experiment, and PCLS from each were prepared prepartum (−8.0 ± 3.6 DIM) and postpartum (+7.7± 1.2 DIM) and treated independently with a variety of PPAR agonists and antagonists: the PPARα agonist WY-14643 and antagonist GW-6471; the PPARδ agonist GW-50156 and antagonist GSK-3787; and the PPARγ agonist rosiglitazone and antagonist GW-9662. Gene expression was assayed through RT-qPCR and RNAseq, and intracellular triacylglycerol (TAG) concentration was measured. PCLS obtained from postpartum cows and treated with a PPARγ agonist displayed upregulation of ACADVL and LIPC while those treated with PPARδ agonist had increased expression of LIPC, PPARD, and PDK4. In PCLS from prepartum cows, transcription of LIPC was increased by all PPAR agonists and NEFA. TAG concentration tended to be larger in tissue slices treated with PPARδ agonist compared to CTR. Use of PPAR isotype-specific antagonists in PCLS cultivated in autologous blood serum failed to decrease expression of PPAR targets, except for PDK4, which was confirmed to be a PPARδ target. Transcriptome sequencing revealed considerable differences in response to PPAR agonists at a false discovery rate-adjusted p-value of 0.2, with the most notable effects exerted by the PPARδ and PPARγ agonists. Differentially expressed genes were mainly related to pathways involved with lipid metabolism and the immune response. Among differentially expressed genes, a subset of 91 genes were identified as novel putative PPAR targets in the bovine liver, by cross-referencing our results with a publicly available dataset of predicted PPAR target genes, and supplementing our findings with prior literature. Our results provide important insights on the use of PCLS as a model for assaying PPAR activation in the periparturient dairy cow.
Collapse
Affiliation(s)
- Sebastiano Busato
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Hunter R. Ford
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Alzahraa M. Abdelatty
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Charles T. Estill
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
- *Correspondence: Massimo Bionaz
| |
Collapse
|
20
|
Hassan FU, Nadeem A, Javed M, Saif-ur-Rehman M, Shahzad MA, Azhar J, Shokrollahi B. Nutrigenomic Interventions to Address Metabolic Stress and Related Disorders in Transition Cows. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2295017. [PMID: 35726316 PMCID: PMC9206560 DOI: 10.1155/2022/2295017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/23/2022] [Indexed: 12/21/2022]
Abstract
For dairy cattle, the period involving a shift from late pregnancy to early lactation termed transition or periparturient is an excruciating phase. Health-related disorders are likely to happen in this time frame. Timely postpartum and metabolic adjustments to this new physical state demands correct management strategies to fulfill the cow's needs for a successful transition to this phase. Among the management strategies, one of the most researched methods for managing transition-related stress is nutritional supplementation. Dietary components directly or indirectly affect the expression of various genes that are believed to be involved in various stress-related responses during this phase. Nutrigenomics, an interdisciplinary approach that combines nutritional science with omics technologies, opens new avenues for studying the genome's complicated interactions with food. This revolutionary technique emphasizes the importance of food-gene interactions on various physiological and metabolic mechanisms. In animal sciences, nutrigenomics aims to promote the welfare of livestock animals and enhance their commercially important qualities through nutritional interventions. To this end, an increasing volume of research shows that nutritional supplementation can be effectively used to manage the metabolic stress dairy cows undergo during the transition period. These nutritional supplements, including polyunsaturated fatty acids, vitamins, dietary amino acids, and phytochemicals, have been shown to modulate energy homeostasis through different pathways, leading to addressing metabolic issues in transition cows.
Collapse
Affiliation(s)
- Faiz-ul Hassan
- Institute of Animal and Dairy Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Asif Nadeem
- Department of Biotechnology, Virtual University of Pakistan, Lahore, Pakistan
| | - Maryam Javed
- Institute of Biochemistry & Biotechnology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | | | | | - Jahanzaib Azhar
- Department of Biotechnology, Virtual University of Pakistan, Lahore, Pakistan
| | - Borhan Shokrollahi
- Department of Animal Science, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| |
Collapse
|
21
|
Sánchez Viafara JA, de Vasconcelos GL, Maculan R, Alves NG, Ferreira MBD, Sudano MJ, Mingoti GZ, Nunes GB, de Lima RR, Drumond RM, Dos Santos RN, Eberlin MN, Negrão F, Donato MAM, Peixoto CA, Camisão de Souza J. Peroxisome proliferator-activated receptor delta-PPARδ agonist (L-165041) enhances bovine embryo survival and post vitrification viability. Reprod Fertil Dev 2022; 34:658-668. [PMID: 35468312 DOI: 10.1071/rd21245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 02/09/2022] [Indexed: 11/23/2022] Open
Abstract
The effect of L-165041 (PPARδ-agonist) on decreasing apoptosis and intracellular lipid content was assessed in fresh and vitrified-warmed in vitro -produced bovine embryos. It was hypothesised that the addition of L-165041 to the culture medium enhances development and cryopreservation. Oocytes were allocated to one of two treatments: control-standard culture medium, or L-165041 added to the medium on day1 with no media change. Ultrastructure, cleavage, and blastocyst rates were evaluated in fresh, and in post-vitrification cultured embryos by optical and electronic microscopy. A subset of fresh embryos were fixed for TUNEL assay and for Sudan-Black-B histochemical staining. Vitrified-warmed embryos were assessed using MALDI-MS technique. Cleavage and blastocyst rates (control 49.4±5.2, L-165041 51.8±4.3) were not influenced by L-165041. The proportion of inner cell mass cells (ICM) was higher in fresh embryos, and the rate of total and ICM apoptosis was lower in L-165041. In warmed-embryos, total and ICM apoptosis was lower in L-165041. The overall hatching rate was higher in L-165041 (66.62±2.83% vs 53.19±2.90%). There was less lipid accumulation in fresh L-165041-embryos. In conclusion, the use of L-165041 is recommended to improve the viability of in vitro -derived bovine embryos.
Collapse
Affiliation(s)
- Jesús Alfonso Sánchez Viafara
- Departamento de Medicina Veterinária, Universidade Federal de Lavras, Lavras, Minas Gerais, Brasil; and Universidad de Santander, Facultad de Ciencias Agrícolas y Veterinarias, Valledupar, Colombia
| | | | - Renata Maculan
- Instituto Federal do Sul de Minas, Machado, Minas Gerais, Brasil
| | - Nadja Gomes Alves
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, Brasil
| | | | | | - Gisele Zoccal Mingoti
- Escola de Medicina Veterinária, Laboratório de Fisiologia da Reprodução, Universidade Estadual Paulista, Campus Araçatuba, São Paulo, Brasil
| | - Giovana Barros Nunes
- Escola de Medicina Veterinária, Laboratório de Fisiologia da Reprodução, Universidade Estadual Paulista, Campus Araçatuba, São Paulo, Brasil
| | - Renato Ribeiro de Lima
- Departamento de Estatística, Universidade Federal de Lavras, Lavras, Minas Gerais, Brasil
| | | | | | - Marcos Nogueira Eberlin
- Universidade Estadual de Campinas, Laboratório ThoMSon de Espectrometria de Massas, Campinas, São Paulo, Brasil
| | - Fernanda Negrão
- Universidade Estadual de Campinas, Laboratório ThoMSon de Espectrometria de Massas, Campinas, São Paulo, Brasil
| | -
- NUMPEX-Bio, Universidade Federal do Rio de Janeiro, Campus Duque de Caxias, Rio de Janeiro, Brasil
| | | | | | - José Camisão de Souza
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, Brasil
| |
Collapse
|
22
|
Abdelrahman M, Wang W, Shaukat A, Kulyar MFEA, Lv H, Abulaiti A, Yao Z, Ahmad MJ, Liang A, Yang L. Nutritional Modulation, Gut, and Omics Crosstalk in Ruminants. Animals (Basel) 2022; 12:ani12080997. [PMID: 35454245 PMCID: PMC9029867 DOI: 10.3390/ani12080997] [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: 01/10/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Over the last decade, animal nutrition science has been significantly developed, supported by the great advancements in molecular technologies. For scientists, the present "feedomics and nutrigenomics" era continues to evolve and shape how research is designed, performed, and understood. The new omics interpretations have established a new point of view for the nutrition–gene interaction, integrating more comprehensive findings from animal physiology, molecular genetics, and biochemistry. In the ruminant model, this modern approach addresses rumen microbes as a critical intermediate that can deepen the studies of diet–gut interaction with host genomics. The present review discusses nutrigenomics’ and feedomics’ potential contribution to diminishing the knowledge gap about the DNA cellular activities of different nutrients. It also presents how nutritional management can influence the epigenetic pathway, considering the production type, life stage, and species for more sustainable ruminant nutrition strategies. Abstract Ruminant nutrition has significantly revolutionized a new and prodigious molecular approach in livestock sciences over the last decade. Wide-spectrum advances in DNA and RNA technologies and analysis have produced a wealth of data that have shifted the research threshold scheme to a more affluent level. Recently, the published literature has pointed out the nutrient roles in different cellular genomic alterations among different ruminant species, besides the interactions with other factors, such as age, type, and breed. Additionally, it has addressed rumen microbes within the gut health and productivity context, which has made interpreting homogenous evidence more complicated. As a more systematic approach, nutrigenomics can identify how genomics interacts with nutrition and other variables linked to animal performance. Such findings should contribute to crystallizing powerful interpretations correlating feeding management with ruminant production and health through genomics. This review will present a road-mapping discussion of promising trends in ruminant nutrigenomics as a reference for phenotype expression through multi-level omics changes.
Collapse
Affiliation(s)
- Mohamed Abdelrahman
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
- Animal Production Department, Faculty of Agriculture, Assuit University, Asyut 71515, Egypt
| | - Wei Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
| | - Aftab Shaukat
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
| | | | - Haimiao Lv
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
| | - Adili Abulaiti
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
| | - Zhiqiu Yao
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
| | - Muhammad Jamil Ahmad
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
| | - Aixin Liang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, China
| | - Liguo Yang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agriculture University, Wuhan 430070, China; (M.A.); (W.W.); (A.S.); (H.L.); (A.A.); (Z.Y.); (M.J.A.); (A.L.)
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: ; Tel.: +86-138-7105-6592
| |
Collapse
|
23
|
Song Y, Zhang Q, Shi J, Fu L, Cheng S. Screening of Genes Related to Growth, Development and Meat Quality of Sahan Crossbred F1 Sheep Based on RNA-Seq Technology. Front Vet Sci 2022; 9:831519. [PMID: 35464379 PMCID: PMC9021821 DOI: 10.3389/fvets.2022.831519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/28/2022] [Indexed: 12/22/2022] Open
Abstract
This study aimed to identify genes related to sheep growth, development and meat quality. Small-tailed Han sheep (STH), and small-tailed Han sheep and Suffolk crossbred F1 (STH×SFK), were selected to determine the growth performance, slaughter performance, and meat quality. The longissimus dorsi muscle was selected for transcriptome sequencing, and the target gene was screened based on bioinformatics analysis; real-time fluorescent quantitative PCR (RT-PCR) and western blotting (WB) were conducted to verify the target gene. Locations of genes in tissues were confirmed via immunofluorescence. The results showed that the pre-slaughter live weight, bust circumference, slaughter performance, and marbling score of the STH×SFK population were significantly higher than those of the STH population (P < 0.01). Sequencing results showed that 560 differentially expressed genes (DEGs) were identified in the STH×SFK population, of which 377 exhibited up-regulated and 183 exhibited down-regulated expression levels. GO annotation revealed that DEGs could be classified into 13 cell components, 10 molecular functions, and 22 biological processes. The KEGG enrichment analysis showed that DEGs were mainly enriched in the Rap1 signaling pathway, Ras signaling pathway, and other pathways related to growth and meat quality. Based on the GO and KEGG analyses, four candidate genes related to sheep growth and meat quality, namely myostain (MSTN), interferon-related developmental regulator 1 (IFRD1), peroxisome proliferator activator receptor delta (PPARD), and myosin light chain 2 (MLC2 or MYL2), were screened. The expression levels of genes and proteins were verified via RT-PCR and WB, and the results were consistent with the trend of transcriptome sequencing. Immunofluorescence results showed that IFRD1 was expressed in the cytoplasm and nucleus, and MYL2 was expressed in the cytoplasm. This study revealed the mechanism of gene regulation of sheep growth and development at the molecular level and provided a theoretical basis for studying sheep genetics and breeding.
Collapse
Affiliation(s)
- Yali Song
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Quanwei Zhang
- College of Life Science and Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Jinping Shi
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Lingjuan Fu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shuru Cheng
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
24
|
Erol SA, Anuk AT, Tanaçan A, Semiz H, Keskin HL, Neşelioğlu S, Erel Ö, Moraloğlu Tekin Ö, Şahin D. An evaluation of maternal serum dynamic thiol-disulfide homeostasis and ischemia modified albumin changes in pregnant women with COVID-19. Turk J Obstet Gynecol 2022; 19:21-27. [PMID: 35343216 PMCID: PMC8966320 DOI: 10.4274/tjod.galenos.2022.72929] [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] [Indexed: 12/01/2022] Open
Abstract
Objective: It is thought that oxidative stress, free radicals, reactive oxygen species and reactive nitrogen species affect the pathophysiology of coronavirus disease-2019 (COVID-19). This study aimed to evaluate the oxidative status in pregnant patients with COVID-19 infection according to the changes seen in the levels of maternal serum thiol-disulfide and ischemia-modified albumin (IMA). Materials and Methods: A study group was formed of 40 pregnant women with confirmed COVID-19 infection (study group) and a control group of 40 healthy pregnant women with no risk factors determined. In this prospective, case-controlled study, analyses were made of the maternal serum native thiol, total thiol, disulfide, IMA, and disulfide/native thiol concentrations. Results: The maternal serum native thiol and total thiol concentrations in the study group were determined to be statistically significantly lower (p=0.007 and p=0.006, respectively), and the disulfide/native thiol ratio was higher but not to a level of statistical significance (p=0.473). There was no difference between the two groups regarding IMA levels (p=0.731). Conclusion: The thiol-disulfide balance was seen to shift in the oxidant direction in pregnancies with COVID-19, which might support the view that ischemic processes play a role in the etiopathogenesis of this novel disease.
Collapse
|
25
|
Conte G, Palombo V, Serra A, Correddu F, D’Andrea M, Macciotta NPP, Mele M. Study of the Fatty Acid Profile of Milk in Different Sheep Breeds: Evaluation by Multivariate Factorial Analysis. Animals (Basel) 2022; 12:ani12060722. [PMID: 35327119 PMCID: PMC8944521 DOI: 10.3390/ani12060722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary The quality of milk is strongly influenced by its lipid profile. The increase in fats with nutraceutical properties at the expense of those negative for human health, has always been a goal to improve the functional properties of milk. To achieve this goal, it is essential to know the metabolism of the mammary gland and the relationship between the various lipid components. Much is known about bovine milk, while the aspect relating to the sheep species has not been developed. The present work aims to investigate the relationships between the various fatty acids in sheep’s milk through a multivariate approach, which can highlight the mammary role of lipid synthesis. Abstract A multivariate analysis was used to investigate the fatty acid (FA) profile in three different Italian sheep breeds: Comisana, Massese, and Sarda. A sample of 852 animals was considered: 118 Massese, 303 Comisana, 431 Sarda. Sarda sheep were divided into two groups, based on their breeding origin (298 and 133 reared in Sardinia and Tuscany, respectively). Sarda sheep, bred both in Sardinia and in Tuscany, were considered in different groups, both because in these two regions most of the sheep of this breed are reared, and because they differ in geographical characteristics and in the farming system. The individual milk FA composition of dairy ewes was analyzed with multivariate factor analysis. The extracted factors were representative of the following eight groups of fatty acids or functions: factor 1 (odd branched fatty acids and long-chain fatty acids), factor 2 (sn3_position), factor 3 (alternative biohydrogenation), factor 4 (SCD_1), factor 5 (SCD_2), factor 6 (SCD_3), factor 7 (fat secretion) and factor 8 (omega-3). A factor analysis suggested the presence of different metabolic pathways for de novo short- and medium-chain fatty acids and Δ9-desaturase products. The ANOVA of factor scores highlighted the significant effects of the breed. The results of the present study showed that breed is an important factor in defining the fatty acid profile of milk, combined with the effect of the diet. Breeds reared in the same farming system (Comisana, Massese and Sarda reared in Tuscany) showed significant differences for all the factors extracted. At the same time, we found differences between the Sarda sheep reared in Sardinia and Tuscany, two different regions of Italy.
Collapse
Affiliation(s)
- Giuseppe Conte
- Department of Agriculture, Food and Environment, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy; (A.S.); (M.M.)
- Research Center of Nutraceutical and Food for Health, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy
- Correspondence:
| | - Valentino Palombo
- Dipartimento Agricoltura, Ambiente e Alimenti, Università degli Studi del Molise, Via De Sanctis snc, 86100 Campobasso, Italy; (V.P.); (M.D.)
| | - Andrea Serra
- Department of Agriculture, Food and Environment, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy; (A.S.); (M.M.)
- Research Center of Nutraceutical and Food for Health, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy
| | - Fabio Correddu
- Department of Agriculture, University of Sassari, Via de Nicola 9, 07100 Sassari, Italy; (F.C.); (N.P.P.M.)
| | - Mariasilvia D’Andrea
- Dipartimento Agricoltura, Ambiente e Alimenti, Università degli Studi del Molise, Via De Sanctis snc, 86100 Campobasso, Italy; (V.P.); (M.D.)
| | | | - Marcello Mele
- Department of Agriculture, Food and Environment, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy; (A.S.); (M.M.)
- Research Center of Nutraceutical and Food for Health, University of Pisa, Via del Borghetto 80, 56124 Pisa, Italy
| |
Collapse
|
26
|
Xuan R, Chao T, Zhao X, Wang A, Chu Y, Li Q, Zhao Y, Ji Z, Wang J. Transcriptome profiling of the nonlactating mammary glands of dairy goats reveals the molecular genetic mechanism of mammary cell remodeling. J Dairy Sci 2022; 105:5238-5260. [DOI: 10.3168/jds.2021-21039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/12/2022] [Indexed: 11/19/2022]
|
27
|
Veshkini A, M Hammon H, Sauerwein H, Tröscher A, Viala D, Delosière M, Ceciliani F, Déjean S, Bonnet M. Longitudinal liver proteome profiling in dairy cows during the transition from gestation to lactation: Investigating metabolic adaptations and their interactions with fatty acids supplementation via repeated measurements ANOVA-simultaneous component analysis. J Proteomics 2022; 252:104435. [PMID: 34823037 DOI: 10.1016/j.jprot.2021.104435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/11/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022]
Abstract
Repeated measurements analysis of variance - simultaneous component analysis (ASCA) has been developed to handle complex longitudinal omics datasets and combine novel information with existing data. Herein, we aimed at applying ASCA to 64 liver proteomes collected at 4-time points (day -21, +1, +28, and + 63 relative to parturition) from 16 Holstein cows treated from 9 wk. antepartum to 9 wk. postpartum (PP) with coconut oil (CTRL) or a mixture of essential fatty acids (EFA) and conjugated linoleic acid (CLA) (EFA + CLA). The ASCA modeled 116, 43, and 97 differentially abundant proteins (DAP) during the transition to lactation, between CTRL and EFA + CLA, and their interaction, respectively. Time-dependent DAP were annotated to pathways related to the metabolism of carbohydrates, FA, and amino acid in the PP period. The DAP between FA and the interaction effect were annotated to the metabolism of xenobiotics by cytochrome P450, drug metabolism - cytochrome P450, retinol metabolism, and steroid hormone biosynthesis. Collectively, ASCA provided novel information on molecular markers of metabolic adaptations and their interactions with EFA + CLA supplementation. Bioinformatics analysis suggested that supplemental EFA + CLA amplified hepatic FA oxidation; cytochrome P450 was enriched to maintain metabolic homeostasis by oxidation/detoxification of endogenous compounds and xenobiotics. SIGNIFICANCE: This report is among the first ones applying repeated measurement analysis of variance-simultaneous component analysis (ASCA) to deal with longitudinal proteomics results. ASCA separately identified differentially abundant proteins (DAP) in 'transition time', 'between fatty acid treatments', and 'their interaction'. We first identified the molecular signature of hepatic metabolic adaptations during postpartum negative energy balance; the enriched pathways were well-known pathways related to mobilizing fatty acids (FA) and amino acids to support continuous energy production through fatty acid oxidation, TCA cycle, and gluconeogenesis. Some of the DAP were not previously reported in transition dairy cows. Secondly, we provide novel information on the mechanisms by which supplemented essential FA and conjugated linoleic acids interact with hepatic metabolism. In this regard, FA amplified hepatic detoxifying and oxidation capacity through ligand activation of nuclear receptors. Finally, we briefly compared the strengths and weaknesses of the ASCA model with PLS-DA and outlined why these methods are complementary.
Collapse
Affiliation(s)
- Arash Veshkini
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany; Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France; Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| | - Harald M Hammon
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany
| | | | - Didier Viala
- INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| | - Mylène Delosière
- INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| | - Fabrizio Ceciliani
- Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| | - Sébastien Déjean
- Institut de Mathématiques de Toulouse, UMR5219, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Muriel Bonnet
- INRAE, Université Clermont Auvergne, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France.
| |
Collapse
|
28
|
Major Nutritional Metabolic Alterations Influencing the Reproductive System of Postpartum Dairy Cows. Metabolites 2022; 12:metabo12010060. [PMID: 35050182 PMCID: PMC8781654 DOI: 10.3390/metabo12010060] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 12/25/2022] Open
Abstract
Early successful conception of postpartum dairy cows is crucial in determining the optimum reproductive efficiency and profitability in modern dairy farming. Due to the inherent high production potential of modern dairy cows, the extra stress burden of peri-parturient events, and associated endocrine and metabolic changes causes negative energy balance (NEBAL) in postpartum cows. The occurrence of NEBAL is associated with excessive fat mobilization in the form of non-esterified fatty acids (NEFAs). The phenomenon of NEFA mobilization furthers with occurrence of ketosis and fatty liver in postpartum dairy cows. High NEFAs and ketones are negatively associated with health and reproductive processes. An additional burden of hypocalcemia, ruminal acidosis, and high protein metabolism in postpartum cows presents further consequences for health and reproductive performance of postpartum dairy cows. This review intends to comprehend these major nutritional metabolic alterations, their mechanisms of influence on the reproduction process, and relevant mitigation strategies.
Collapse
|
29
|
Loor JJ. Nutrigenomics in livestock: potential role in physiological regulation and practical applications. ANIMAL PRODUCTION SCIENCE 2022. [DOI: 10.1071/an21512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Mu T, Hu H, Ma Y, Feng X, Zhang J, Gu Y. Regulation of Key Genes for Milk Fat Synthesis in Ruminants. Front Nutr 2021; 8:765147. [PMID: 34901115 PMCID: PMC8659261 DOI: 10.3389/fnut.2021.765147] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/27/2021] [Indexed: 12/26/2022] Open
Abstract
Milk fat is the most important and energy-rich substance in milk and plays an important role in the metabolism of nutrients during human growth and development. It is mainly used in the production of butter and yogurt. Milk fat not only affects the flavor and nutritional value of milk, but also is the main target trait of ruminant breeding. There are many key genes involve in ruminant milk fat synthesis, including ACSS2, FASN, ACACA, CD36, ACSL, SLC27A, FABP3, SCD, GPAM, AGPAT, LPIN, DGAT1, PLIN2, XDH, and BTN1A1. Taking the de novo synthesis of fatty acids (FA) and intaking of long-chain fatty acids (LCFA) in blood to the end of lipid droplet secretion as the mainline, this manuscript elucidates the complex regulation model of key genes in mammary epithelial cells (MECs) in ruminant milk fat synthesis, and constructs the whole regulatory network of milk fat synthesis, to provide valuable theoretical basis and research ideas for the study of milk fat regulation mechanism of ruminants.
Collapse
Affiliation(s)
| | | | | | | | | | - Yaling Gu
- School of Agriculture, Ningxia University, Yinchuan, China
| |
Collapse
|
31
|
Jing Y, Chen Y, Wang S, Ouyang J, Hu L, Yang Q, Wang M, Zhang B, Loor JJ. Circadian Gene PER2 Silencing Downregulates PPARG and SREBF1 and Suppresses Lipid Synthesis in Bovine Mammary Epithelial Cells. BIOLOGY 2021; 10:biology10121226. [PMID: 34943141 PMCID: PMC8698707 DOI: 10.3390/biology10121226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 01/01/2023]
Abstract
Simple Summary The present study was constructed to determine the effects of the core circadian clock gene, Period 2 (PER2), on lipid synthesis in bovine mammary epithelial cells (BMECs). Data revealed that PER2-regulated genes were involved in fatty acid de novo synthesis, desaturation, TAG accumulation, and lipid droplet secretion in primary BMECs, partly by inhibiting PPARG and SREBF1. Our overall data suggests that PER2 in bovine mammary cells plays a role in regulating milk fat synthesis directly, or via the activation of the transcription regulators PPARG and SREBF1. This study provides molecular evidence underscoring a link between the circadian clock and lipid metabolism in bovines. Abstract PER2, a circadian clock gene, is associated with mammary gland development and lipid synthesis in rodents, partly via regulating peroxisome proliferator-activated receptor gamma (PPARG). Whether such a type of molecular link existed in bovines was unclear. We hypothesized that PER2 was associated with lipid metabolism and regulated cell cycles and apoptosis in bovine mammary epithelial cells (BMECs). To test this hypothesis, BMECs isolated from three mid-lactation (average 110 d postpartum) cows were used. The transient transfection of small interfering RNA (siRNA) was used to inhibit PER2 transcription in primary BMECs. The silencing of PER2 led to lower concentrations of cellular lipid droplets and triacylglycerol along with the downregulation of lipogenic-related genes such as ACACA, FASN, LPIN1, and SCD, suggesting an overall inhibition of lipogenesis and desaturation. The downregulation of PPARG and SREBF1 in response to PER2 silencing underscored the importance of circadian clock signaling and the transcriptional regulation of lipogenesis. Although the proliferation of BMECs was not influenced by PER2 silencing, the number of cells in the G2/GM phase was upregulated. PER2 silencing did not affect cell apoptosis. Overall, the data provided evidence that PER2 participated in the coordination of mammary lipid metabolism and was potentially a component of the control of lipid droplets and TAG synthesis in ruminant mammary cells. The present data suggested that such an effect could occur through direct effects on transcriptional regulators.
Collapse
Affiliation(s)
- Yujia Jing
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi 832000, China; (Y.J.); (Q.Y.)
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.C.); (S.W.); (J.O.); (L.H.)
| | - Yifei Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.C.); (S.W.); (J.O.); (L.H.)
| | - Shan Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.C.); (S.W.); (J.O.); (L.H.)
| | - Jialiang Ouyang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.C.); (S.W.); (J.O.); (L.H.)
| | - Liangyu Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.C.); (S.W.); (J.O.); (L.H.)
| | - Qingyong Yang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi 832000, China; (Y.J.); (Q.Y.)
| | - Mengzhi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.C.); (S.W.); (J.O.); (L.H.)
- Correspondence: (M.W.); (B.Z.); (J.J.L.)
| | - Bin Zhang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi 832000, China; (Y.J.); (Q.Y.)
- Correspondence: (M.W.); (B.Z.); (J.J.L.)
| | - Juan J. Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
- Correspondence: (M.W.); (B.Z.); (J.J.L.)
| |
Collapse
|
32
|
Role of Peroxisome Proliferator-Activated Receptors (PPARs) in Energy Homeostasis of Dairy Animals: Exploiting Their Modulation through Nutrigenomic Interventions. Int J Mol Sci 2021; 22:ijms222212463. [PMID: 34830341 PMCID: PMC8619600 DOI: 10.3390/ijms222212463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/31/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are the nuclear receptors that could mediate the nutrient-dependent transcriptional activation and regulate metabolic networks through energy homeostasis. However, these receptors cannot work properly under metabolic stress. PPARs and their subtypes can be modulated by nutrigenomic interventions, particularly under stress conditions to restore cellular homeostasis. Many nutrients such as polyunsaturated fatty acids, vitamins, dietary amino acids and phytochemicals have shown their ability for potential activation or inhibition of PPARs. Thus, through different mechanisms, all these nutrients can modulate PPARs and are ultimately helpful to prevent various metabolic disorders, particularly in transition dairy cows. This review aims to provide insights into the crucial role of PPARs in energy metabolism and their potential modulation through nutrigenomic interventions to improve energy homeostasis in dairy animals.
Collapse
|
33
|
Xu T, Liu R, Lu X, Wu X, Heneberg P, Mao Y, Jiang Q, Loor J, Yang Z. Lycium barbarum polysaccharides alleviate LPS-induced inflammatory responses through PPARγ/MAPK/NF-κB pathway in bovine mammary epithelial cells. J Anim Sci 2021; 100:6429718. [PMID: 34791267 DOI: 10.1093/jas/skab345] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
As the main component of the gram-negative bacterial cell wall, lipopolysaccharide (LPS) is well-documented as an inducer of inflammation in bovine mammary cells. Lycium barbarum (goji) polysaccharides (LBP) have been used in non-ruminants as prebiotics to improve growth performance, immune ability and antioxidant capacity. We aimed to investigate the underlying effects of LBPs on pro-inflammatory responses in LPS-stimulated primary bovine mammary epithelial cells (bMECs). Cells were isolated from mammary tissue of 3 lactating Holstein cows without clinical disease (30.26 ± 3.1 kg/d of milk yield; 175 ± 6 DIM). For the pre-experimental treatment, bMECs were precultured with serum-free medium for 12 h. Treatments were as follows: pretreatment with culture medium devoid of LPS or LBP for 30 h (CON); CON for 24 h followed by challenge with 2 μg/mL LPS for 6 h (LPS); pretreatment with 100 μg/mL or 300 μg/mL LBP for 24 h followed by LPS challenge (2 μg/mL) for 6 h (LBP(100)+LPS; LBP(300)+LPS). To further determine if the effect of LBP on immune-regulation is PPARγ activation-dependent, an inhibitor of PPARγ, GW9662, at a concentration of 1 μM was used. Cells treated with LBP at 100, 300 and 500 μg/mL had upregulated protein abundance of PPARγ, while PGC1α had a higher expression only at 300 μg/mL of LBP treatment. Compared with CON, cells pretreated with LBP at 100 and 300 μg/mL had greater protein abundance of SCD1 and SREBP1. EdU staining and cell wound healing assays showed that the negative effect of LPS alone on cell proliferation was reversed by pretreatment with LBP at both 100 and 300 μg/mL. Upregulation of gene and protein abundance of proinflammatory factors and cytokines (COX-2, NLRP3, TNF-α, IL-1β and IL-6) induced by LPS stimulation were alleviated by LBP pretreatment at 300 μg/mL (more than 2-fold decrease). Compared with LPS challenge alone, phosphorylation of proteins involved in NF-κB (IκBα and p65) and MAPK (p38, JNK and ERK) pathways was downregulated following LBP treatment. Additionally, inhibition of PPARγ by GW9662 weakened the protective effect of LBP on LPS-induced protein abundance of phosphorylated p65, COX-2, IL-1β and TNF-α. These results indicated that the protective effect of LBP on LPS-induced bMECs inflammatory responses is PPARγ activation-dependent. As such, this knowledge might help design strategies for intervening against the detrimental effects of bovine mastitis.
Collapse
Affiliation(s)
- Tianle Xu
- Joint International Research Laboratory of Agriculture and Agri-product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, P. R. China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Run Liu
- Joint International Research Laboratory of Agriculture and Agri-product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, P. R. China
| | - Xubin Lu
- Joint International Research Laboratory of Agriculture and Agri-product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, P. R. China
| | - Xinyue Wu
- Joint International Research Laboratory of Agriculture and Agri-product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, P. R. China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Petr Heneberg
- Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Yongjiang Mao
- Joint International Research Laboratory of Agriculture and Agri-product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, P. R. China
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, USA
| | - Juan Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, USA
| | - Zhangping Yang
- Joint International Research Laboratory of Agriculture and Agri-product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, P. R. China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| |
Collapse
|
34
|
Zhou F, Ouyang Y, Miao Y. Peroxisome proliferator-activated receptor gamma regulates genes involved in milk fat synthesis in mammary epithelial cells of water buffalo. Anim Sci J 2021; 92:e13537. [PMID: 33682250 DOI: 10.1111/asj.13537] [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: 03/28/2020] [Revised: 01/17/2021] [Accepted: 01/25/2021] [Indexed: 01/17/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a critical transcription factor regulating lipid and glucose metabolism. However, the regulatory effect of PPARγ on milk fat synthesis in buffalo mammary gland is not clear. In order to explore the role of buffalo PPARG gene in milk fat synthesis, lentivirus-mediated interference was used to knock it down and then the recombinant fusion expression vector was transfected into buffalo mammary epithelial cell (BMEC) to overexpress it. PPARG gene knockdown significantly decreased the expression of CD36, FABP3, FABP4, ACSS2, ELOVL6, DGAT2, BTN1A1, AGPAT6, LPIN1, ABCG2, PPARGC1A, INSIG1, FASN, and SREBF2 genes and significantly upregulated the expression of INSIG2 gene but had no significant effect on the expression of ACSL1, GPAM, and SREBF1 genes. PPARG overexpression significantly increased the relative mRNA abundance of CD36, FABP3, FABP4, ACSS2, ELOVL6, DGAT2, BTN1A1, AGPAT6, LPIN1, PPARGC1A, INSIG1, and SREBF2 genes and significantly downregulated the expression of INSIG2 gene but had no significant effect on the expression of ACSL1, GPAM, ABCG2, FASN, and SREBF1 genes. In addition, knockdown/overexpression of PPARG gene significantly decreased/increased triacylglycerol (TAG) content in BMECs. This study revealed that buffalo PPARG gene is a key gene regulating buffalo milk fat synthesis.
Collapse
Affiliation(s)
- Fangting Zhou
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China.,College of Chemistry, Biology and Environment, Yuxi Normal University, Yuxi, China
| | - Yina Ouyang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China.,Herbivorous Livestock Research Institute, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Yongwang Miao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
35
|
Busato S, Bionaz M. When Two plus Two Is More than Four: Evidence for a Synergistic Effect of Fatty Acids on Peroxisome Proliferator-Activated Receptor Activity in a Bovine Hepatic Model. Genes (Basel) 2021; 12:genes12081283. [PMID: 34440457 PMCID: PMC8393910 DOI: 10.3390/genes12081283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
The inclusion of fat in livestock diets represents a valuable and cost-effective way to increase the animal’s caloric intake. Beyond their caloric value, fatty acids can be understood in terms of their bioactivity, via the modulation of the ligand-dependent nuclear peroxisome proliferator-activated receptors (PPAR). Isotypes of PPAR regulate important metabolic processes in both monogastric and ruminant animals, including the metabolism of fatty acids (FA), the production of milk fat, and the immune response; however, information on the modulation of bovine PPAR by fatty acids is limited. The objective of this study was to expand our understanding on modulation of bovine PPAR by FA, both when used individually and in combination, in an immortalized cell culture model of bovine liver. Of the 10 FA included in the study, the greatest activation of the PPAR reporter was detected with saturated FA C12:0, C16:0, and C18:0, as well as phytanic acid, and the unsaturated FA C16:1 and C18:1. When supplemented in mixtures of 2 FA, the most effective combination was C12:0 + C16:0, while in mixtures of 3 FA, the greatest activation was caused by combinations of C12:0 with C16:0 and either C18:0, C16:1, or C18:1. Some mixtures display a synergistic effect that leads to PPAR activation greater than the sum of their parts, which may be explained by structural dynamics within the PPAR ligand-binding pocket. Our results provide fundamental information for the development of tailored dietary plans that focus on the use of FA mixtures for nutrigenomic purposes.
Collapse
|
36
|
Genome-Wide SNP Analysis for Milk Performance Traits in Indigenous Sheep: A Case Study in the Egyptian Barki Sheep. Animals (Basel) 2021; 11:ani11061671. [PMID: 34205212 PMCID: PMC8228706 DOI: 10.3390/ani11061671] [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: 04/27/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary The Barki sheep is one of the three main breeds in Egypt, which is spread mainly throughout the northwestern coastal zone, which has harsh conditions. Considering the harsh, semi-arid habitat of this breed, milk performance traits such as milk yield and milk composition have a very important role in the feeding of newborn lambs and affect their growth during the early stage of life. In this study, rare milk performance data and genomic information of Barki sheep were used to uncover diversified genomic regions that could explain the variability of milk yield and milk quality traits in the studied population of Barki ewes. Genome-wide analysis identified genomic regions harboring interesting candidate genes such as SLC5A8, NUB1, TBC1D1, KLF3 and ABHD5 for milk yield and PPARA and FBLN1 genes for milk quality traits. The findings offer valuable information for obtaining a better understanding of the genetics of milk performance traits and contribute to the genetic improvement of these traits in Barki sheep. Abstract Sheep milk yield and milk composition traits play an important role in supplying newborn lambs with essential components such as amino acids, energy, vitamins and immune antibodies and are also of interest in terms of the nutritional value of the milk for human consumption. The aim of this study was to identify genomic regions and candidate genes for milk yield and milk composition traits through genome-wide SNP analyses between high and low performing ewes of the Egyptian Barki sheep breed, which is well adapted to the harsh conditions of North-East Africa. Therefore, out of a herd of 111 ewes of the Egyptian Barki sheep breed (IBD = 0.08), ewes representing extremes in milk yield and milk quality traits (n = 25 for each group of animals) were genotyped using the Illumina OvineSNP50 V2 BeadChip. The fixation index (FST) for each SNP was calculated between the diversified groups. FST values were Z-transformed and used to identify putative SNPs for further analysis (Z(FST) > 10). Genome-wide SNP analysis revealed genomic regions covering promising candidate genes related to milk performance traits such as SLC5A8, NUB1, TBC1D1, KLF3 and ABHD5 for milk yield and PPARA and FBLN1 genes for milk quality trait. The results of this study may contribute to the genetic improvement of milk performance traits in Barki sheep breed and to the general understanding of the genetic contribution to variability in milk yield and quality traits.
Collapse
|
37
|
D'Oliveira MC, Vedovatto M, Cortada Neto IM, Coelho RN, Morais MDG, Gomes MDNB, Negrão JA, Franco GL. Effect of walking exercise and nutritional plan on goat performance. Livest Sci 2021. [DOI: 10.1016/j.livsci.2021.104450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
38
|
Coleman DN, Alharthi AS, Liang Y, Lopes MG, Lopreiato V, Vailati-Riboni M, Loor JJ. Multifaceted role of one-carbon metabolism on immunometabolic control and growth during pregnancy, lactation and the neonatal period in dairy cattle. J Anim Sci Biotechnol 2021; 12:27. [PMID: 33536062 PMCID: PMC7860211 DOI: 10.1186/s40104-021-00547-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Dairy cattle undergo dramatic metabolic, endocrine, physiologic and immune changes during the peripartal period largely due to combined increases in energy requirements for fetal growth and development, milk production, and decreased dry matter intake. The negative nutrient balance that develops results in body fat mobilization, subsequently leading to triacylglycerol (TAG) accumulation in the liver along with reductions in liver function, immune dysfunction and a state of inflammation and oxidative stress. Mobilization of muscle and gluconeogenesis are also enhanced, while intake of vitamins and minerals is decreased, contributing to metabolic and immune dysfunction and oxidative stress. Enhancing post-ruminal supply of methyl donors is one approach that may improve immunometabolism and production synergistically in peripartal cows. At the cellular level, methyl donors (e.g. methionine, choline, betaine and folic acid) interact through one-carbon metabolism to modulate metabolism, immune responses and epigenetic events. By modulating those pathways, methyl donors may help increase the export of very low-density lipoproteins to reduce liver TAG and contribute to antioxidant synthesis to alleviate oxidative stress. Thus, altering one-carbon metabolism through methyl donor supplementation is a viable option to modulate immunometabolism during the peripartal period. This review explores available data on the regulation of one-carbon metabolism pathways in dairy cows in the context of enzyme regulation, cellular sensors and signaling mechanisms that might respond to increased dietary supply of specific methyl donors. Effects of methyl donors beyond the one-carbon metabolism pathways, including production performance, immune cell function, mechanistic target or rapamycin signaling, and fatty acid oxidation will also be highlighted. Furthermore, the effects of body condition and feeding system (total mixed ration vs. pasture) on one-carbon metabolism pathways are explored. Potential effects of methyl donor supply during the pepartum period on dairy calf growth and development also are discussed. Lastly, practical nutritional recommendations related to methyl donor metabolism during the peripartal period are presented. Nutritional management during the peripartal period is a fertile area of research, hence, underscoring the importance for developing a systems understanding of the potential immunometabolic role that dietary methyl donors play during this period to promote health and performance.
Collapse
Affiliation(s)
- Danielle N. Coleman
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Abdulrahman S. Alharthi
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451 Saudi Arabia
| | - Yusheng Liang
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Matheus Gomes Lopes
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Vincenzo Lopreiato
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Mario Vailati-Riboni
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| | - Juan J. Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801 USA
| |
Collapse
|
39
|
Boesche K, Donkin S. Bovine pyruvate carboxylase gene proximal promoter activity is regulated by saturated and unsaturated fatty acids in Madin-Darby bovine kidney cells. J Dairy Sci 2021; 104:2308-2317. [DOI: 10.3168/jds.2020-18803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/17/2020] [Indexed: 01/10/2023]
|
40
|
Díaz Galván C, Méndez Olvera ET, Martínez Gómez D, Gloria Trujillo A, Hernández García PA, Espinosa Ayala E, Palacios Martínez M, Lara Bueno A, Mendoza Martínez GD, Velázquez Cruz LA. Influence of a Polyherbal Mixture in Dairy Calves: Growth Performance and Gene Expression. Front Vet Sci 2021; 7:623710. [PMID: 33575280 PMCID: PMC7870704 DOI: 10.3389/fvets.2020.623710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/21/2020] [Indexed: 11/23/2022] Open
Abstract
A polyherbal feed mixture containing (Achyrantes aspera, Trachyspermum ammi, Citrullus colocynthis, Andrographis paniculata, and Azadirachta indica) was evaluated in growing calves through blood chemistry, blood biometry, and gene expression during the pre-ruminant to weaning period. Forty Holstein calves (initial BW 45.6 ± 3.2 kg; 22.8 ± 0.9 days post birth) from a dairy farm were randomly assigned to the following treatments: 0, 3, 4, and 5 g/d of a polyherbal mixture, dosed in colloid gels with gelatin. Calves were housed in individual outdoor boxes with ad libitum access to a 21.5% CP calf starter and water and fed individually with a mixture of milk and a non-medicated milk replacer (22% CP). Blood samples were collected on day 59 for blood chemistry, blood biometry, and gene expression analysis in leukocyte through microarray assays. Immunoglobulins were quantified by enzyme-linked immunosorbent assay. The animals treated with the polyherbal mixture showed a quadratic effect on final body weight, daily weight gain, final hip height, and final thoracic girth. The best performance results were obtained with a treatment dose of 4 g/d. The serum IgG increased linearly with the treatment doses. Gene set enrichment analysis of upregulated genes revealed that the three biological processes with higher fold change were tight junction, mucin type O-Glycan biosynthesis, and intestinal immune network for IgA production. Also, these upregulated genes influenced arachidonic acid metabolism, and pantothenate and CoA biosynthesis. Gene ontology enrichment analysis indicated that the pathways enriched were PELP1 estrogen receptor interacting protein pathways, nuclear receptors in lipid metabolism and toxicity, tight junction, ECM-receptor interaction, thyroid hormone signaling pathways, vascular smooth muscle contraction, ribosome function, glutamatergic synapse pathway, focal adhesion, Hippo, calcium, and MAPK signaling pathways.
Collapse
Affiliation(s)
- Cesar Díaz Galván
- Doctorado en Ciencias Agropecuarias, Universidad Autónoma Metropolitana Xochimilco, Ciudad de México, Mexico
| | - Estela Teresita Méndez Olvera
- Laboratorio de Biología Molecular, Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana Xochimilco, Ciudad de México, Mexico
| | - Daniel Martínez Gómez
- Laboratorio de Microbiología Agropecuaria, Universidad Autónoma Metropolitana Xochimilco, Ciudad de México, Mexico
| | - Adrián Gloria Trujillo
- Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana Xochimilco, Ciudad de México, Mexico
| | | | - Enrique Espinosa Ayala
- Centro Universitario UAEM Amecameca, Universidad Autónoma del Estado de México, Amecameca, Mexico
| | - Monika Palacios Martínez
- Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana Xochimilco, Ciudad de México, Mexico
| | | | - Germán David Mendoza Martínez
- Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana Xochimilco, Ciudad de México, Mexico
| | | |
Collapse
|
41
|
Bionaz M, Vargas-Bello-Pérez E, Busato S. Advances in fatty acids nutrition in dairy cows: from gut to cells and effects on performance. J Anim Sci Biotechnol 2020; 11:110. [PMID: 33292523 PMCID: PMC7667790 DOI: 10.1186/s40104-020-00512-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
High producing dairy cows generally receive in the diet up to 5-6% of fat. This is a relatively low amount of fat in the diet compared to diets in monogastrics; however, dietary fat is important for dairy cows as demonstrated by the benefits of supplementing cows with various fatty acids (FA). Several FA are highly bioactive, especially by affecting the transcriptome; thus, they have nutrigenomic effects. In the present review, we provide an up-to-date understanding of the utilization of FA by dairy cows including the main processes affecting FA in the rumen, molecular aspects of the absorption of FA by the gut, synthesis, secretion, and utilization of chylomicrons; uptake and metabolism of FA by peripheral tissues, with a main emphasis on the liver, and main transcription factors regulated by FA. Most of the advances in FA utilization by rumen microorganisms and intestinal absorption of FA in dairy cows were made before the end of the last century with little information generated afterwards. However, large advances on the molecular aspects of intestinal absorption and cellular uptake of FA were made on monogastric species in the last 20 years. We provide a model of FA utilization in dairy cows by using information generated in monogastrics and enriching it with data produced in dairy cows. We also reviewed the latest studies on the effects of dietary FA on milk yield, milk fatty acid composition, reproduction, and health in dairy cows. The reviewed data revealed a complex picture with the FA being active in each step of the way, starting from influencing rumen microbiota, regulating intestinal absorption, and affecting cellular uptake and utilization by peripheral tissues, making prediction on in vivo nutrigenomic effects of FA challenging.
Collapse
Affiliation(s)
- Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, 97331, USA.
| | - Einar Vargas-Bello-Pérez
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 3, DK-1870, Frederiksberg C, Denmark
| | - Sebastiano Busato
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
42
|
Gao ST, Girma DD, Bionaz M, Ma L, Bu DP. Hepatic transcriptomic adaptation from prepartum to postpartum in dairy cows. J Dairy Sci 2020; 104:1053-1072. [PMID: 33189277 DOI: 10.3168/jds.2020-19101] [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: 06/17/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022]
Abstract
The transition from pregnancy to lactation is the most challenging period for high-producing dairy cows. The liver plays a key role in biological adaptation during the peripartum. Prior works have demonstrated that hepatic glucose synthesis, cholesterol metabolism, lipogenesis, and inflammatory response are increased or activated during the peripartum in dairy cows; however, those works were limited by a low number of animals used or by the use of microarray technology, or both. To overcome such limitations, an RNA sequencing analysis was performed on liver biopsies from 20 Holstein cows at 7 ± 5d before (Pre-P) and 16 ± 2d after calving (Post-P). We found 1,475 upregulated and 1,199 downregulated differently expressed genes (DEG) with a false discovery rate adjusted P-value < 0.01 between Pre-P and Post-P. Bioinformatic analysis revealed an activation of the metabolism, especially lipid, glucose, and amino acid metabolism, with increased importance of the mitochondria and a key role of several signaling pathways, chiefly peroxisome proliferators-activated receptor (PPAR) and adipocytokines signaling. Fatty acid oxidation and gluconeogenesis, with a likely increase in amino acid utilization to produce glucose, were among the most important functions revealed by the transcriptomic adaptation to lactation in the liver. Although gluconeogenesis was induced, data indicated decrease in expression of glucose transporters. The analysis also revealed high activation of cell proliferation but inhibition of xenobiotic metabolism, likely due to the liver response to inflammatory-like conditions. Co-expression network analysis disclosed a tight connection and coordination among genes driving biological processes associated with protein synthesis, energy and lipid metabolism, and cell proliferation. Our data confirmed the importance of metabolic adaptation to lipid and glucose metabolism in the liver of early Post-P cows, with a pivotal role of PPAR and adipocytokines.
Collapse
Affiliation(s)
- S T Gao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - D D Girma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - M Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97331
| | - L Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - D P Bu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
43
|
Ferreira L, Soares MAM, Rodrigues MT, de Araujo JLS, de Melo ALP, Gasparino E, Garcia OSR. UCP2 and PPARG gene polymorphisms and their association with milk yield and composition traits in goats. Small Rumin Res 2020. [DOI: 10.1016/j.smallrumres.2020.106210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
44
|
Zhou F, Teng X, Wang P, Zhang Y, Miao Y. Isolation, identification, expression and subcellular localization of PPARG gene in buffalo mammary gland. Gene 2020; 759:144981. [PMID: 32707300 DOI: 10.1016/j.gene.2020.144981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 11/15/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARG), as a member of the nuclear receptor superfamily, plays an important role in adipocyte differentiation and regulation of lipid and glucose metabolism. In this study, the transcripts of PPARG gene were isolated and identified in buffalo mammary gland. The results showed that two types of transcripts (PPARG1 and PPARG2) of PPARG gene produced by alternative 5' end use were expressed in buffalo mammary gland, and each of them had four different alternative splicing variants. The PPARG1 includes PPARG1a, PPARG1b, PPARG1c and PPARG1d, while the PPARG2 contains PPARG2a, PPARG2b, PPARG2c and PPARG2d. Among them, only PPARG1a, PPARG2a and PPARG2d can encode complete functional proteins with three complete functional domains, and the rest encode truncated proteins with incomplete functional domains. All the eight variants of PPARG protein do not contain transmembrane regions and signal peptides, but their conserved domain, secondary and tertiary structure and subcellular localization were different. Subcellular localization confirmed that the main transcripts PPARG1a and PPARG2a played a functional role in the nucleus, which was consistent with the results by in silico prediction. RT-qPCR analysis of buffalo mammary tissue showed that the mRNA expression levels of PPARG1 and PPARG2 in lactation were higher than those in non-lactation, and the expression levels of transcripts PPARG2d and PPARG1b + PPARG2b in lactating stage were also higher than those in non-lactating stage, but the mRNA abundance of transcripts PPARG1c, PPARG1d and PPARG2c in non-lactating period was higher than that in lactating period. The results of this study suggest that PPARG1 and PPARG2 may play important role in buffalo milk fat synthesis, and the eight alternative splicing variants found here are likely to be related to the post-transcriptional regulation of lactation.
Collapse
Affiliation(s)
- Fangting Zhou
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Xiaohong Teng
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Pei Wang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Yongyun Zhang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China; Teaching Demonstration Center of the Basic Experiments of Agricultural Majors, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Yongwang Miao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China.
| |
Collapse
|
45
|
Caprarulo V, Erb SJ, Chandler TL, Zenobi MG, Barton BA, Staples CR, White HM. The effects of prepartum energy intake and peripartum rumen-protected choline supplementation on hepatic genes involved in glucose and lipid metabolism. J Dairy Sci 2020; 103:11439-11448. [PMID: 33222856 DOI: 10.3168/jds.2020-18840] [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: 05/04/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Nutritional interventions, either by controlling dietary energy (DE) or supplementing rumen-protected choline (RPC) or both, may mitigate negative postpartum metabolic health outcomes. A companion paper previously reported the effects of DE density and RPC supplementation on production and health outcomes. The objective of this study was to examine the effects of DE and RPC supplementation on the expression of hepatic oxidative, gluconeogenic, and lipid transport genes during the periparturient period. At 47 ± 6 d relative to calving (DRTC), 93 multiparous Holstein cows were randomly assigned in groups to dietary treatments in a 2 × 2 factorial of (1) excess energy (EXE) without RPC supplementation (1.63 Mcal of NEL/kg of dry matter; EXE-RPC); (2) maintenance energy (MNE) without RPC supplementation (1.40 Mcal of NEL/kg dry matter; MNE-RPC); (3) EXE with RPC supplementation (EXE+RPC); and (4) MNE with RPC supplementation (MNE+RPC). To achieve the objective of this research, liver biopsy samples were collected at -14, +7, +14, and +21 DRTC and analyzed for mRNA expression (n = 16/treatment). The interaction of DE × RPC decreased glucose-6-phosphatase and increased peroxisome proliferator-activated receptor α in MNE+RPC cows. Expression of cytosolic phosphoenolpyruvate carboxykinase was altered by the interaction of dietary treatments with reduced expression in EXE+RPC cows. A dietary treatment interaction was detected for expression of pyruvate carboxylase although means were not separated. Dietary treatment interactions did not alter expression of carnitine palmitoyltransferase 1A or microsomal triglyceride transfer protein. The 3-way interaction of DE × RPC × DRTC affected expression of carnitine palmitoyltransferase 1A, glucose-6-phosphatase, and peroxisome proliferator-activated receptor α and tended to affect cytosolic phosphoenolpyruvate carboxykinase. Despite previously reported independent effects of DE and RPC on production variables, treatments interacted to influence hepatic metabolism through altered gene expression.
Collapse
Affiliation(s)
- V Caprarulo
- Department of Dairy Science, University of Wisconsin-Madison, Madison 53706; Department of Health, Animal Science and Food Safety, University of Milan, Milan 20134, Italy
| | - S J Erb
- Department of Dairy Science, University of Wisconsin-Madison, Madison 53706
| | - T L Chandler
- Department of Dairy Science, University of Wisconsin-Madison, Madison 53706
| | - M G Zenobi
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | | | - C R Staples
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | - H M White
- Department of Dairy Science, University of Wisconsin-Madison, Madison 53706.
| |
Collapse
|
46
|
Sevrin T, Boquien CY, Gandon A, Grit I, de Coppet P, Darmaun D, Alexandre-Gouabau MC. Fenugreek Stimulates the Expression of Genes Involved in Milk Synthesis and Milk Flow through Modulation of Insulin/GH/IGF-1 Axis and Oxytocin Secretion. Genes (Basel) 2020; 11:E1208. [PMID: 33081164 PMCID: PMC7602737 DOI: 10.3390/genes11101208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/06/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
We previously demonstrated galactagogue effect of fenugreek in a rat model of lactation challenge, foreshadowing its use in women's breastfeeding management. To assess longitudinal molecular mechanisms involved in milk synthesis/secretion in dams submitted to fenugreek supplementation, inguinal mammary, pituitary glands and plasma were isolated in forty-three rats nursing large 12 pups-litters and assigned to either a control (CTL) or a fenugreek-supplemented (FEN) diet during lactation. RT-PCR were performed at days 12 and 18 of lactation (L12 and L18) and the first day of involution (Inv1) to measure the relative expression of genes related to both milk synthesis and its regulation in the mammary gland and lactogenic hormones in the pituitary gland. Plasma hormone concentrations were measured by ELISA. FEN diet induced 2- to 3-times higher fold change in relative expression of several genes related to macronutrient synthesis (Fasn, Acaca, Fabp3, B4galt1, Lalba and Csn2) and energy metabolism (Cpt1a, Acads) and in IGF-1 receptor in mammary gland, mainly at L12. Pituitary oxytocin expression and plasma insulin concentration (+77.1%) were also significantly increased. Altogether, these findings suggest fenugreek might extend duration of peak milk synthesis through modulation of the insulin/GH/IGF-1 axis and increase milk ejection by activation of oxytocin secretion.
Collapse
Affiliation(s)
- Thomas Sevrin
- FRANCE Bébé Nutrition (FBN) Laboratory, 53000 Laval, France;
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Clair-Yves Boquien
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Alexis Gandon
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Isabelle Grit
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Pierre de Coppet
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| | - Dominique Darmaun
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
- Nantes University Hospital (CHU) Nantes, F-44000 Nantes, France
| | - Marie-Cécile Alexandre-Gouabau
- Mixed Research Unit 1280 Pathophysiology of Nutritional adaptations (UMR 1280 PhAN) Nantes University, Research Center in Human Nutrition-West (CRNH-O), Institute of Digestive Tract Diseases (IMAD), French National Research Institute for Agriculture, Food and Environment (INRAE), F-44000 Nantes, France; (C.-Y.B.); (A.G.); (I.G.); (P.d.C.); (D.D.)
| |
Collapse
|
47
|
Peroxisome proliferator-activated receptor γ isoforms differentially regulate preadipocyte proliferation, apoptosis, and differentiation in chickens. Poult Sci 2020; 99:6410-6421. [PMID: 33248556 PMCID: PMC7705046 DOI: 10.1016/j.psj.2020.09.086] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/24/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) has 2 protein isoforms (PPARγ1 and PPARγ2) generated by alternative promoter usage and alternative splicing. However, their functional uniqueness and similarity remain unclear. In the study, we investigated the effects of lentivirus-mediated overexpression of PPARγ1 and PPARγ2 on proliferation, apoptosis, and differentiation of the immortalized chicken preadipocytes. Cell Counting Kit–8 assay showed PPARγ1 and PPARγ2 overexpression markedly suppressed cell proliferation, and fluorescence activated cell sorting analysis showed that PPARγ1 and PPARγ2 overexpression caused cell cycle arrest at G0/G1 phase. Cell death detection ELISA analysis showed both PPARγ1 and PPARγ2 overexpression induced cell apoptosis. Oil red O staining and gene expression analysis showed both PPARγ1 and PPARγ2 overexpression promoted preadipocyte differentiation. In the presence of PPARγ ligand, rosiglitazone, PPARγ2 overexpression was more potent in inducing apoptosis, promoting adipogenesis, and suppressing cell proliferation than PPARγ1 overexpression. We further explored the molecular basis for their functional differences. Reporter gene assay showed that under ligand conditions, PPARγ2 overexpression resulted in 1.68-fold increase in transcription activity compared with PPARγ1. Electrophoretic mobility shift assay showed both PPARγ1 and PPARγ2 could bind to PPAR response element (PPRE) as heterodimer with retinoid X receptor alpha, and by comparison, PPARγ2 had a higher affinity for PPRE than PPARγ1. Reporter gene assay showed expression PPARγ1 and PPARγ2 similarly induced fatty acid synthase and adipocyte fatty acid–binding protein promoter activity but differentially induced lipoprotein lipase and perilipin 1 promoter activities. Coimmunoprecipitation analysis showed that PPARγ1 and PPARγ2 interacted similarly with the coactivators, Tat-interacting protein 60. Taken together, our results demonstrate that PPARγ1 and PPARγ2 differentially regulate preadipocyte proliferation, apoptosis, and differentiation as a result of their distinct and overlapping molecular functions.
Collapse
|
48
|
Wang H, Niu W, Wu F, Qiu X, Yu Z, He Y, Li H, Su H, Cao B. Effects of dietary energy on antioxidant capacity, glucose-lipid metabolism and meat fatty acid profile of Holstein bulls at different ages. J Anim Physiol Anim Nutr (Berl) 2020; 105:199-209. [PMID: 33006191 DOI: 10.1111/jpn.13457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/15/2020] [Accepted: 09/03/2020] [Indexed: 01/01/2023]
Abstract
This experiment was conducted to investigate the effects of dietary energy on antioxidant capacity, liver glucose-lipid-related gene expressions and meat fatty acid of Holstein bulls. Thirty-six Holstein bulls (age, 17.0 ± 0.49 months; body weight, 493.3 ± 39.7 kg) were randomly allocated to three dietary treatments. The metabolizable energy of diets was 10.12, 10.90 and 11.68 MJ/kg. Bulls in each dietary treatment were sampled at the age of 20, 23 or 26 months. Results showed that serum glutathione peroxidase and superoxide dismutase decreased with the increasing age. Dietary energy and age had interaction effects on the expressions of fatty acid synthase, peroxisome proliferator-activated receptor alpha, acyl coenzyme A oxidase 1 and carnitine palmitoyl-transferase 1 alpha. Besides, the increase of age and dietary energy increased the expression of liver phosphoenolpyruvate carboxykinase 1. The expressions of liver glucose-6-phosphatase, tumour necrosis factor alpha and sterol regulatory element binding protein 1 increased with the increasing age. The increase of age and dietary energy increased the proportions of C18:1cis-9, C18:2n-6trans and monounsaturated fatty acid. In summary, the increase of age and dietary energy enhanced the intensity of metabolic changes and inflammatory responses. Dietary energy and age affected the expressions of liver lipid metabolism-related genes, further affected meat fatty acid composition of Holstein bulls.
Collapse
Affiliation(s)
- Haibo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization (Southwest Minzu University), Ministry of Education, Chengdu, China
| | - Wenjing Niu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fei Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xinjun Qiu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhantao Yu
- Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| | - Yang He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hang Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Huawei Su
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Binghai Cao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
49
|
Liang Y, Alharthi AS, Elolimy AA, Bucktrout R, Lopreiato V, Martinez-Cortés I, Xu C, Fernandez C, Trevisi E, Loor JJ. Molecular networks of insulin signaling and amino acid metabolism in subcutaneous adipose tissue are altered by body condition in periparturient Holstein cows. J Dairy Sci 2020; 103:10459-10476. [PMID: 32921465 DOI: 10.3168/jds.2020-18612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/05/2020] [Indexed: 12/28/2022]
Abstract
Peripartal cows mobilize not only body fat but also body protein to satisfy their energy requirements. The objective of this study was to determine the effect of prepartum BCS on blood biomarkers related to energy and nitrogen metabolism, and mRNA and protein abundance associated with AA metabolism and insulin signaling in subcutaneous adipose tissue (SAT) in peripartal cows. Twenty-two multiparous Holstein cows were retrospectively classified into a high BCS (HBCS; n = 11, BCS ≥ 3.5) or normal BCS (NBCS; n = 11, BCS ≤ 3.17) group at d 28 before expected parturition. Cows were fed the same diet as a total mixed ration before parturition and were fed the same lactation diet postpartum. Blood samples collected at -10, 7, 15, and 30 d relative to parturition were used for analyses of biomarkers associated with energy and nitrogen metabolism. Biopsies of SAT harvested at -15, 7, and 30 d relative to parturition were used for mRNA (real time-PCR) and protein abundance (Western blotting) assays. Data were subjected to ANOVA using the MIXED procedure of SAS (v. 9.4; SAS Institute Inc., Cary, NC), with P ≤ 0.05 being the threshold for significance. Cows in HBCS had greater overall plasma nonesterified fatty acid concentrations, due to marked increases at 7 and 15 d postpartum. This response was similar (BCS × Day effect) to protein abundance of phosphorylated (p) protein kinase B (p-AKT), the insulin-induced glucose transporter (SLC2A4), and the sodium-coupled neutral AA transporter (SLC38A1). Abundance of these proteins was lower at -15 d compared with NBCS cows, and either increased (SLC2A4, SLC38A1) or did not change (p-AKT) at 7 d postpartum in HBCS. Unlike protein abundance, however, overall mRNA abundances of the high-affinity cationic (SLC7A1), proton-coupled (SLC36A1), and sodium-coupled amino acid transporters (SLC38A2) were greater in HBCS than NBCS cows, due to upregulation in the postpartum phase. Those responses were similar to protein abundance of p-mTOR, which increased (BCS × Day effect) at 7 d in HBCS compared with NBCS cows. mRNA abundance of argininosuccinate lyase (ASL) and arginase 1 (ARG1) also was greater overall in HBCS cows. Together, these responses suggested impaired insulin signaling, coupled with greater postpartum AA transport rate and urea cycle activity in SAT of HBCS cows. An in vitro study using adipocyte and macrophage cocultures stimulated with various concentrations of fatty acids could provide some insights into the role of immune cells in modulating adipose tissue immunometabolic status, including insulin resistance and AA metabolism.
Collapse
Affiliation(s)
- Y Liang
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - A S Alharthi
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - A A Elolimy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock 72205; Arkansas Children's Nutrition Center, Little Rock 72205; Department of Animal Production, National Research Centre, Giza, 12611, Egypt
| | - R Bucktrout
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - V Lopreiato
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - I Martinez-Cortés
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801; Agricultural and Animal Production Department, UAM-Xochimilco, Mexico City 04960, Mexico
| | - C Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - C Fernandez
- Animal Science Department, Universitàt Politècnica de Valencia, 46022 Valencia, Spain
| | - E Trevisi
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
50
|
Zhang J, Xue L, Nie A, Yang Q, Peng X, Chen Z, Yang L, Xie Y, Yuan A, Xu J. Spatiotemporal heterogeneity of PPARγ expression in porcine uteroplacenta for regulating of placental angiogenesis through VEGF-mediated signalling. Reprod Domest Anim 2020; 55:1479-1489. [PMID: 32762098 DOI: 10.1111/rda.13797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/24/2020] [Accepted: 08/01/2020] [Indexed: 11/26/2022]
Abstract
Non-infectious prenatal mortality severely affects the porcine industry, with pathological placentation as a likely key reason. Previous studies have demonstrated that peroxisome proliferator-activated receptor gamma (PPARγ) deficiency causes defects in the uteroplacental vasculature and induces embryonic losses in mice. However, its role in porcine placental angiogenesis remains unclear. In the present study, PPARγ expression was investigated in porcine uteroplacental tissues at gestational day (GD) 25, GD40 and GD70 via quantitative polymerase chain reaction (qPCR), Western blot and immunohistochemistry (IHC). Moreover, the roles of PPARγ in porcine placental angiogenesis were investigated using a cell model of porcine umbilical vein endothelial cells (PUVECs) to conduct proliferation, migration and tube formation assays in vitro and a mouse xenograft model to assess capillary formation in vivo. The results showed that PPARγ was mainly located in the glandular epithelium, trophoblast, amniotic chorion epithelium and vascular endothelium, as indicated by the higher expression levels at GD25 and GD40 than at GD70 in endometrium and by higher expression levels at GD40 and GD70 than at GD25 in placenta. Moreover, PPARγ expression was significantly downregulated in placenta with dead foetus. In PUVECs, knocking out PPARγ significantly inhibited proliferation, migration and tube formation in vitro and inhibited capillary formation in mouse xenografts in vivo by blocking S-phase, promoting apoptosis and downregulating the angiogenic factors of VEGF and its receptors. Overall, the spatiotemporal heterogeneity of PPARγ expression in porcine uteroplacental tissue suggests its vital role in endometrial remodelling and placental angiogenesis, and PPARγ regulates placental angiogenesis through VEGF-mediated signalling.
Collapse
Affiliation(s)
- Juzuo Zhang
- College of Biological and Food Engineering, 'Double First-Class' Applied Characteristic Discipline of Bioengineering in Hunan High Educational Institution, Huaihua University, Huaihua, China.,Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Liqun Xue
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Ang Nie
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.,Huaihua Municipal Center of Animal Husbandry and Aquatic Products Bureau, Huaihua, China
| | - Qing Yang
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Xuan Peng
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Zhilong Chen
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Lisha Yang
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Yang Xie
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Anwen Yuan
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Junfei Xu
- College of Biological and Food Engineering, 'Double First-Class' Applied Characteristic Discipline of Bioengineering in Hunan High Educational Institution, Huaihua University, Huaihua, China
| |
Collapse
|