1
|
Fonseca PADS, Suarez-Vega A, Esteban-Blanco C, Marina H, Pelayo R, Gutiérrez-Gil B, Arranz JJ. Integration of epigenomic and genomic data to predict residual feed intake and the feed conversion ratio in dairy sheep via machine learning algorithms. BMC Genomics 2025; 26:313. [PMID: 40165084 PMCID: PMC11956460 DOI: 10.1186/s12864-025-11520-1] [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: 11/18/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Feed efficiency (FE) is an essential trait in livestock species because of the constant demand to increase the productivity and sustainability of livestock production systems. A better understanding of the biological mechanisms associated with FEs might help improve the estimation and selection of superior animals. In this work, differentially methylated regions (DMRs) were identified via genome-wide bisulfite sequencing (GWBS) by comparing the DNA methylation profiles of milk somatic cells from dairy ewes that were divergent in terms of residual feed intake. The DMRs were identified by comparing divergent groups for residual feed intake (RFI), the feed conversion ratio (FCR), and the consensus between both metrics (Cons). Additionally, the predictive performance of these DMRs and genetic variants mapped within these regions was evaluated via three machine learning (ML) models (xgboost, random forest (RF), and multilayer feedforward artificial neural network (deeplearning)). The average performance of each model was based on the root mean squared error (RMSE) and squared Spearman correlation (rho2). Finally, the best model for each scenario was selected on the basis of the highest ratio between rho2 and RMSE. RESULTS In total, 12,257, 9,328, and 6,723 genes were annotated for DMRs detected in the RFI, FCR, and Cons groups, respectively. These genes are associated with important pathways for regulating FE in dairy sheep, such as protein digestion and absorption, hormone synthesis and secretion, control of energy availability, cellular signaling, and feed behavior pathways. With respect to the ML predictions, the smallest mean RMSE (0.17) was obtained using RF, which was used to predict RFI. The highest mean rho2 (0.20) was obtained when the RFI was predicted via the mean methylation within the DMRs identified, the consensus groups were compared, and the genetic variants mapped within these DMRs were included. The best overall models were obtained for the prediction of RFI using the DMRs obtained in the comparison of RFI groups (RMSE = 0.10, rho2 = 0.86) using xgboost and the DMRs plus the genetic variants identified via the Cons groups (RMSE = 0.07, rho2 = 0.62) using RF. CONCLUSIONS The results provide new insights into the biological mechanisms associated with FE and the control of these processes through epigenetic mechanisms. Additionally, the potential use of epigenetic information as a biomarker for the prediction of FE can be suggested based on the obtained results.
Collapse
Affiliation(s)
| | - Aroa Suarez-Vega
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, Leon, 24007, Spain
| | - Cristina Esteban-Blanco
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, Leon, 24007, Spain
| | - Héctor Marina
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, Leon, 24007, Spain
| | - Rocío Pelayo
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, Leon, 24007, Spain
| | - Beatriz Gutiérrez-Gil
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, Leon, 24007, Spain
| | - Juan-José Arranz
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, Leon, 24007, Spain.
| |
Collapse
|
2
|
Abuduaini B, Jiyuan Z, Rehati A, Liang Z, Yunlin S. Regulation of Alternative Splicing of Lipid Metabolism Genes in Sepsis-Induced Liver Damage by RNA-Binding Proteins. Inflammation 2024; 47:1952-1968. [PMID: 38727856 PMCID: PMC11607010 DOI: 10.1007/s10753-024-02017-2] [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: 02/07/2024] [Revised: 03/23/2024] [Accepted: 03/31/2024] [Indexed: 11/30/2024]
Abstract
RNA binding proteins (RBPs) have the potential for transcriptional regulation in sepsis-induced liver injury, but precise functions remain unclear. Our aim is to conduct a genome-wide expression analysis of RBPs and illuminate changes in the regulation of alternative splicing in sepsis-induced liver injury. RNA-seq data on "sepsis and liver" from the publicly available NCBI data set was analyzed, and differentially expressed RBPs and alternative splicing events (ASEs) in the healthy and septic liver were identified. Co-expression analyses of sepsis-regulated RBPs and ASEs were performed. Models of sepsis were established to validate hepatic RBP gene expression patterns with different treatments. Pairwise analysis of gene expression profiles of sham, cecum ligation puncture (CLP), and CLP with dichloroacetate (CLPDCA) mice allowed 1208 differentially expressed genes (DEGs), of which 800 were up-regulated and 408 down-regulated, to be identified. DEGs were similar in both Sham and CLPDCA mice. The KEGG analysis showed that up-regulated genes as being involved in cytokine-cytokine receptor interaction and IL-17 signaling pathway and down-regulated genes in metabolic pathways. Differences in lipid metabolism-related alternative splicing events, including A3SS, were also found in CLP and CLPDCA compared with sham mice. Thirty-seven RBPs, including S100a11, Ads2, Fndc3b, Fn1, Ddx28, Car2, Cisd1, and Ptms, were differentially expressed in CLP mice and the regulated alternative splicing genes(RASG) with the RBP shown to be enriched in lipid metabolic and oxidation-reduction-related processes by GO functional analysis. In KEEG analysis the RASG mainly enriched in metabolic pathway. The models of sepsis were constructed with different treatment groups, and S100a11 expression in the CLP group found to be higher than in the sham group, a change that was reversed by DCA. The alternative splicing ratio of Srebf1 and Cers2 decreased compared with the sham group increased after DCA treatment. Abnormal profiles of gene expression and alternative splicing were associated with sepsis-induced liver injury. Unusual expression of RBPs, such as S100a11, may regulate alternative splicing of lipid metabolism-associated genes, such as Srebf1 and Cers2, in the septic liver. RBPs may constitute potential treatment targets for sepsis-induced liver injury.
Collapse
Affiliation(s)
- Buzukela Abuduaini
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University, 393 South Li Yu Shan Road, Ürümqi, 830054, Xinjiang, China
| | - Zhang Jiyuan
- First Clinical institute of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, China
| | - Aliya Rehati
- Department of Gastroenternology, The First Affiliated Hospital of Xinjiang Medical University, 393 South Li Yu Shan Road, Ürümqi, 830054, Xinjiang, China
| | - Zhao Liang
- Department of General Surgery, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, Xinjiang, China
| | - Song Yunlin
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University, 393 South Li Yu Shan Road, Ürümqi, 830054, Xinjiang, China.
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University,State Key Laboratory of Pathogenesis, Prevention and Treatment of high Incidence Diseases in Central Asia, 393 South Li Yu Shan Road, Ürümqi, 830054, Xinjiang, China.
| |
Collapse
|
3
|
Pereira AC, Serrano-Cuñarro L, Cruz MT, Cavadas C, Pereira CMF. The link between alterations in circadian rhythms and lipid metabolism in bipolar disorder: the hypothesis of lipid droplets. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2024; 46:e20243670. [PMID: 39102528 PMCID: PMC11744263 DOI: 10.47626/1516-4446-2024-3670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Bipolar disorder (BD) is a neuropsychiatric illness characterized by recurrent episodes of mania and depression, leading to significant cognitive and functional impairments, psychiatric and metabolic comorbidities, and substantial healthcare costs. The complex nature and lack of specific biomarkers for BD make it a daily challenge for clinicians. Therefore, advancing our understanding of BD pathophysiology is essential to identify novel diagnostic biomarkers and potential therapeutic targets. Although its neurobiology remains unclear, circadian disruption and lipid alterations have emerged as key hallmarks of BD. Lipids are essential components of the brain and play a critical role in regulating synaptic activity and neuronal development. Consequently, alterations in brain lipids may contribute to the neuroanatomical changes and reduced neuroplasticity observed in BD. Lipid droplets, which regulate the storage of neutral lipids, buffer the levels of toxic lipids within cells. These dynamic organelles adapt to cellular needs, and their dysregulated accumulation has been implicated in several pathological conditions. Notably, lipid droplets and different classes of lipids exhibit rhythmic oscillations throughout the 24-hour cycle, suggesting a link between lipid metabolism, circadian rhythms, and lipid droplets. In this review, we explore the impairment of circadian rhythms and lipid metabolism in BD and present evidence that circadian clocks regulate lipid droplet accumulation. Importantly, we propose the "hypothesis of lipid droplets for BD," which posits that impaired lipid metabolism in BD is closely linked to alterations in lipid droplet homeostasis driven by circadian clock disruption.
Collapse
Affiliation(s)
- Ana Catarina Pereira
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra (UC), Coimbra, Portugal
- Centro de Inovação em Biotecnologia e Biomedicina (CIBB), UC, Coimbra, Portugal
- Faculdade de Medicina, UC, Coimbra, Portugal
- Centro Académico Clínico de Coimbra, Coimbra, Portugal
| | - Laura Serrano-Cuñarro
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra (UC), Coimbra, Portugal
- Centro de Inovação em Biotecnologia e Biomedicina (CIBB), UC, Coimbra, Portugal
| | - Maria Teresa Cruz
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra (UC), Coimbra, Portugal
- Centro de Inovação em Biotecnologia e Biomedicina (CIBB), UC, Coimbra, Portugal
- Centro Académico Clínico de Coimbra, Coimbra, Portugal
- Faculdade de Farmácia, UC, Coimbra, Portugal
| | - Cláudia Cavadas
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra (UC), Coimbra, Portugal
- Centro de Inovação em Biotecnologia e Biomedicina (CIBB), UC, Coimbra, Portugal
- Faculdade de Farmácia, UC, Coimbra, Portugal
| | - Cláudia Maria Fragão Pereira
- Centro de Neurociências e Biologia Celular, Universidade de Coimbra (UC), Coimbra, Portugal
- Centro de Inovação em Biotecnologia e Biomedicina (CIBB), UC, Coimbra, Portugal
- Faculdade de Medicina, UC, Coimbra, Portugal
- Centro Académico Clínico de Coimbra, Coimbra, Portugal
| |
Collapse
|
4
|
Li X, Wu Y, Yang X, Gao R, Lu Q, Lv X, Chen Z. The cooperative regulatory effect of the miRNA-130 family on milk fat metabolism in dairy cows. Anim Biosci 2024; 37:1289-1302. [PMID: 38665085 PMCID: PMC11222843 DOI: 10.5713/ab.23.0485] [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: 11/15/2023] [Revised: 01/02/2024] [Accepted: 03/02/2024] [Indexed: 07/05/2024] Open
Abstract
OBJECTIVE There is a strong relationship between the content of beneficial fatty acids in milk and milk fat metabolic activity in the mammary gland. To improve milk quality, it is therefore necessary to study fatty acid metabolism in bovine mammary gland tissue. In adipose tissue, peroxisome proliferator-activated receptor gamma (PPARG), the core transcription factor, regulates the fatty acid metabolism gene network and determines fatty acid deposition. However, its regulatory effects on mammary gland fatty acid metabolism during lactation have rarely been reported. METHODS Transcriptome sequencing was performed during the prelactation period and the peak lactation period to examine mRNA expression. The significant upregulation of PPARG drew our attention and led us to conduct further research. RESULTS According to bioinformatics prediction, dual-luciferase reporter system detection, real-time quantitative reverse transcription polymerase chain reaction and Western blotting, miR-130a and miR-130b could directly target PPARG and inhibit its expression. Furthermore, triglyceride and oil red O staining proved that miR-130a and miR-130b inhibited milk fat metabolism in bovine mammary epithelial cells (BMECs), while PPARG promoted this metabolism. In addition, we also found that the coexpression of miR-130a and miR-130b significantly enhanced their ability to regulate milk fat metabolism. CONCLUSION In conclusion, our findings indicated that miR-130a and miR-130b could target and repress PPARG and that they also have a functional superposition effect. miR-130a and miR-130b seem to synergistically regulate lipid catabolism via the control of PPARG in BMECs. In the long-term, these findings might be helpful in developing practical means to improve high-quality milk.
Collapse
Affiliation(s)
- Xiaofen Li
- School of Animal Science and Technology, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300,
China
| | - Yanni Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009,
China
| | - Xiaozhi Yang
- School of Animal Science and Technology, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300,
China
| | - Rui Gao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009,
China
| | - Qinyue Lu
- Laboratory of Animal Developmental Biology, Department of Animal Science, Chungbuk National University, Cheongju 28644,
Korea
| | - Xiaoyang Lv
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou 225009,
China
- International Joint Research Laboratory, Universities of Jiangsu Province of China, Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou 225009,
China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009,
China
| |
Collapse
|
5
|
Erickson ML, Dobias D, Keleher MR, Dabelea D, Bergman BC, Broussard JL, Boyle KE. In Vitro Circadian Clock Gene Expression Assessments in Mesenchymal Stem Cells from Human Infants: A Pilot Study. Nutrients 2023; 16:52. [PMID: 38201882 PMCID: PMC10780581 DOI: 10.3390/nu16010052] [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: 11/16/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Exposure to intrauterine obesity can disrupt clock gene rhythmicity in animal models. The aim of this pilot study was to determine if maternal obesity alters rhythmic expression of core clock in mesenchymal stem cells (MSCs) from umbilical cords of human infants born to mothers with obesity (Ob-MSC) vs. normal weight (NW-MSC). METHODS We compared in vitro rhythmic expression patterns of core clock (BMAL1, CLOCK, PER2) and clock-output (NR1D1), components in undifferentiated Ob-MSCs (n = 3) vs. NW-MSCs (n = 3). MSCs were harvested every 2 h, following a dexamethasone shock, for 30 h. Adipogenesis or myogenesis was induced in vitro and markers of adipogenesis and fat storage were assessed, respectively. RESULTS We detected significant rhythmicity in expression patterns of BMAL1, PER2, and NR1D1 at the group level in Ob- and NW-MSCs (p < 0.05). PER2 oscillatory amplitude was 3-fold higher in Ob-MSCs vs. NW-MSCs (p < 0.006). During adipogenesis, Ob-MSCs had higher PPARγ protein content (p = 0.04) vs. NW-MSC. During myogenesis, Ob-MSCs had higher saturated triacylglycerols (p = 0.04) vs. NW-MSC. CONCLUSION Rhythmic expressions of BMAL1, PER2, and NR1D1 are detectable in undifferentiated MSCs. Higher PER2 oscillatory amplitude was paralleled by higher markers of fat storage during differentiation in Ob-MSCs vs. NW-MSCs, and supports that the core clock and cellular metabolism may be linked in infant MSCs.
Collapse
Affiliation(s)
| | - Devin Dobias
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.D.)
| | - Madeline Rose Keleher
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.D.)
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO 80045, USA;
| | - Bryan C. Bergman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (B.C.B.); (J.L.B.)
| | - Josiane L. Broussard
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (B.C.B.); (J.L.B.)
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO 80011, USA
| | - Kristen E. Boyle
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.D.)
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO 80045, USA;
| |
Collapse
|
6
|
Fonseca PAS, Suárez-Vega A, Esteban-Blanco C, Pelayo R, Marina H, Gutiérrez-Gil B, Arranz JJ. Epigenetic regulation of functional candidate genes for milk production traits in dairy sheep subjected to protein restriction in the prepubertal stage. BMC Genomics 2023; 24:511. [PMID: 37658326 PMCID: PMC10472666 DOI: 10.1186/s12864-023-09611-y] [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: 03/06/2023] [Accepted: 08/21/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND As the prepubertal stage is a crucial point for the proper development of the mammary gland and milk production, this study aims to evaluate how protein restriction at this stage can affect methylation marks in milk somatic cells. Here, 28 Assaf ewes were subjected to 42.3% nutritional protein restriction (14 animals, NPR) or fed standard diets (14 animals, C) during the prepubertal stage. During the second lactation, the milk somatic cells of these ewes were sampled, and the extracted DNA was subjected to whole-genome bisulfite sequencing. RESULTS A total of 1154 differentially methylated regions (DMRs) were identified between the NPR and C groups. Indeed, the results of functional enrichment analyses of the genes harboring these DMRs suggested their relevant effects on the development of the mammary gland and lipid metabolism in sheep. The additional analysis of the correlations of the mean methylation levels within these DMRs with fat, protein, and dry extract percentages in the milk and milk somatic cell counts suggested associations between several DMRs and milk production traits. However, there were no phenotypic differences in these traits between the NPR and C groups. CONCLUSION In light of the above, the results obtained in the current study might suggest potential candidate genes for the regulation of milk production traits in the sheep mammary gland. Further studies focusing on elucidating the genetic mechanisms affected by the identified DMRs may help to better understand the biological mechanisms modified in the mammary gland of dairy sheep as a response to nutritional challenges and their potential effects on milk production.
Collapse
Affiliation(s)
- P. A. S. Fonseca
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071 León, Spain
| | - A. Suárez-Vega
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071 León, Spain
| | - C. Esteban-Blanco
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071 León, Spain
| | - R. Pelayo
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071 León, Spain
| | - H. Marina
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071 León, Spain
| | - B. Gutiérrez-Gil
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071 León, Spain
| | - J. J. Arranz
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071 León, Spain
| |
Collapse
|
7
|
Wendrich KS, Azimi H, Ripperger JA, Ravussin Y, Rainer G, Albrecht U. Deletion of the Circadian Clock Gene Per2 in the Whole Body, but Not in Neurons or Astroglia, Affects Sleep in Response to Sleep Deprivation. Clocks Sleep 2023; 5:204-225. [PMID: 37092429 PMCID: PMC10123656 DOI: 10.3390/clockssleep5020017] [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: 01/25/2023] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023] Open
Abstract
The sleep-wake cycle is a highly regulated behavior in which a circadian clock times sleep and waking, whereas a homeostatic process controls sleep need. Both the clock and the sleep homeostat interact, but to what extent they influence each other is not understood. There is evidence that clock genes, in particular Period2 (Per2), might be implicated in the sleep homeostatic process. Sleep regulation depends also on the proper functioning of neurons and astroglial cells, two cell-types in the brain that are metabolically dependent on each other. In order to investigate clock-driven contributions to sleep regulation we non-invasively measured sleep of mice that lack the Per2 gene either in astroglia, neurons, or all body cells. We observed that mice lacking Per2 in all body cells (Per2Brdm and TPer2 animals) display earlier onset of sleep after sleep deprivation (SD), whereas neuronal and astroglial Per2 knock-out animals (NPer2 and GPer2, respectively) were normal in that respect. It appears that systemic (whole body) Per2 expression is important for physiological sleep architecture expressed by number and length of sleep bouts, whereas neuronal and astroglial Per2 weakly impacts night-time sleep amount. Our results suggest that Per2 contributes to the timing of the regulatory homeostatic sleep response by delaying sleep onset after SD and attenuating the early night rebound response.
Collapse
Affiliation(s)
- Katrin S Wendrich
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Hamid Azimi
- Section of Medicine, Department of Neuroscience, University of Fribourg, 1700 Fribourg, Switzerland
| | - Jürgen A Ripperger
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Yann Ravussin
- Section of Medicine, Department of Endocrinology, Metabolism and Cardiovascular System, University of Fribourg, 1700 Fribourg, Switzerland
| | - Gregor Rainer
- Section of Medicine, Department of Neuroscience, University of Fribourg, 1700 Fribourg, Switzerland
| | - Urs Albrecht
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
8
|
An Integrated Analysis of Lactation-Related miRNA and mRNA Expression Profiles in Donkey Mammary Glands. Genes (Basel) 2022; 13:genes13091637. [PMID: 36140805 PMCID: PMC9498803 DOI: 10.3390/genes13091637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Donkey milk is consumed by humans for its nutritional and therapeutic properties. MicroRNAs (miRNAs) and messenger RNAs (mRNAs) have been implicated in the regulation of milk component synthesis and mammary gland development. However, the regulatory profile of the miRNAs and mRNAs involved in lactation in donkeys is unclear. We performed mRNA-seq and miRNA-seq and constructed coexpression regulatory networks for the mammary glands during the lactating and nonlactating period of jennies. We identified 3144 differentially expressed (DE) mRNAs (987 upregulated mRNAs and 2157 downregulated mRNAs) and 293 DE miRNAs (231 upregulated miRNAs and 62 downregulated miRNAs) in the lactating group compared to the nonlactating group. The DE miRNA target mRNA were significantly associated with pathways related to RNA polymerase, glycosphingolipid biosynthesis, mRNA surveillance, ribosome biogenesis in eukaryotes, glycerophospholipid metabolism, Ras signaling, and the fly hippo signaling pathway. The mRNA–miRNA coregulation analysis showed that novel-m0032-3p, miR-195, miR-26-5p, miR-23-3p, miR-674-3p, and miR-874-3p are key miRNAs that target mRNAs involved in immunity and milk lipid, protein, and vitamin metabolism in the jenny mammary gland. Our results improve the current knowledge of the molecular mechanisms regulating bioactive milk component metabolism in the mammary glands and could be used to improve milk production in donkeys.
Collapse
|
9
|
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: 3.0] [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
|