1
|
Bai X, Li L, Wu Y, Jie B. Flavonoids of Euphorbia hirta inhibit inflammatory mechanisms via Nrf2 and NF-κB pathways. Cell Biochem Biophys 2025; 83:1167-1183. [PMID: 39505796 DOI: 10.1007/s12013-024-01551-y] [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] [Accepted: 09/11/2024] [Indexed: 11/08/2024]
Abstract
Euphorbia hirta has anti-inflammatory effects in traditional medicine, but its anti-inflammatory mechanism has not been explored at the cellular and molecular levels. To unravel these mechanisms, the main active components in the 65 and 95% ethanol extracts of Euphorbia hirta were first identified by UPLC-Q-TOF/MS. Subsequently, potential anti-inflammatory targets and signaling pathways were predicted using network pharmacology and experimentally validated using RT-PCR and flow cytometry in a lipopolysaccharide (LPS)-induced inflammation model of RAW264.7 cells. The results revealed flavonoids as the key active components. Network pharmacology uncovered 71 potential anti-inflammation targets, with a protein-protein interaction (PPI) network highlighting 8 cores targets, including IL-6, TNF, NFκB and Nrf2 et al. Furthermore, Euphorbia hirta exerts anti-inflammation effects through modulation of Nrf2 and NF-κB signaling pathways. Specifically, the 65% ethanol extract of Euphorbia hirta (EE65) and quercitrin (HPG) exerted anti-inflammatory activity by inhibiting the expression of inflammatory genes associated with the NF-κB signaling pathway, whereas baicalein (HCS) suppressed cellular inflammation by promoting Nrf2-mediated antioxidant gene expression and enhancing apoptosis of inflammatory cells. The results of the study suggest that Euphorbia hirta has potential for the development of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xiaolin Bai
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Lijun Li
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuning Wu
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Bai Jie
- College of Life Sciences, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Cai L, Chen Q, Hua C, Niu L, Kong Q, Wu L, Ni Y. Chronic Dexamethasone Disturbs the Circadian Rhythm of Melatonin and Clock Genes in Goats. Animals (Basel) 2025; 15:115. [PMID: 39795058 PMCID: PMC11718956 DOI: 10.3390/ani15010115] [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: 11/23/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Dex is a drug commonly used as an immunosuppressive and anti-inflammatory agent in humans and animals. GCs have a profound impact on melatonin expression and biological rhythm. However, the effect of chronic exposure to Dex on melatonin secretion and biological clock gene expression in ruminants is still unclear. Ten goats were randomly divided into two groups: the control group was injected with saline, and the Dex-treated group was intramuscularly injected daily for 21 d with 0.2 mg/kg Dex. The rhythm of melatonin secretion in the plasma was disturbed in the Dex group, and the plasma and colon levels of melatonin were lower in the Dex group compared to the control group (p < 0.05). Dex leads to a significant decrease in the expression of Arylalkylamine N-acetyltransferase (AANAT), a key melatonin synthase, in the pineal gland and colon. Detecting intestinal leakage-related indices showed that diamine oxidase (DAO) and lipopolysaccharide (LPS) content increased significantly in the Dex group (p < 0.05). We also detected genes associated with biological rhythms in the plasma. In the control group, the five tested genes showed circadian rhythms, but the circadian rhythms of Clock, Cry1, Cry2, and Per2 were abolished or blunted by the Dex (p < 0.05). Protein levels of CLOCK and BMAL1 in the colon changed significantly (p < 0.05). In conclusion, the above experimental results show that chronic exposure to Dex leads to the disorder of the circadian rhythms of melatonin secretion and clock genes.
Collapse
Affiliation(s)
- Liuping Cai
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.C.); (Q.K.)
| | - Qu Chen
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Q.C.); (C.H.); (L.N.); (L.W.)
| | - Canfeng Hua
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Q.C.); (C.H.); (L.N.); (L.W.)
| | - Liqiong Niu
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Q.C.); (C.H.); (L.N.); (L.W.)
| | - Qijun Kong
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.C.); (Q.K.)
| | - Lei Wu
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Q.C.); (C.H.); (L.N.); (L.W.)
| | - Yingdong Ni
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Q.C.); (C.H.); (L.N.); (L.W.)
| |
Collapse
|
3
|
Mazhar MU, Naz S, Khan JZ, Azam S, Ghazanfar S, Tipu MK. Protective potential of Bacillus subtilis (NMCC-path-14) against extraarticular manifestations during acute and sub-acute phase of arthritis using mice model. Biochem Biophys Res Commun 2024; 733:150708. [PMID: 39298918 DOI: 10.1016/j.bbrc.2024.150708] [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: 07/16/2024] [Revised: 08/29/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Extra-articular manifestations (EAM), which are associated with rheumatoid arthritis (RA), affect the quality of life of patients and are one of the critical causes of early mortality. This study was aimed at investigating whether Bacillus subtilis NMCC-path-14 (1 × 108 CFU/animal/day) could serve as a valuable therapeutic agent in managing EAM using complete Freund's adjuvant (CFA) induced arthritis during acute and sub-acute phases. Arthritis was induced using intra-dermal administration of CFA in the right hind paw of mice on day 1. Dexamethasone (Dexa) (5 mg/kg/day/animal) was used as a standard treatment. Animals in Dexa and Bacillus subtilis concurrent treatment (BS-CT) received treatments on day 1. The Bacillus subtilis pre-treatment (BS-PT) group received a probiotic dose 7 days before arthritis induction. Parameters like body weight, relative organ weight, colon length, hematology, serum biochemistry, antioxidant capacity, and histopathology of liver, kidney, spleen, colon, stress-related behavioral changes, and cortisol levels were evaluated on days 7 (acute) and 14 (sub-acute). Dexa failed to manage the EAM in arthritic mice and instead exacerbated them. On the other hand, B. subtilis NMCC-path-14 significantly declined EAM with no notable side effects, highlighting its safety and effectiveness. The current data show that B. subtilis NMCC-path-14 may be an alternative option for arthritis treatment that can reduce systemic symptoms associated with arthritis. More studies are required to comprehend the underlying mechanisms of mitigating the EAM by B. subtilis NMCC-path-14.
Collapse
Affiliation(s)
- Muhammad Usama Mazhar
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Sadaf Naz
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Shahzad Azam
- Department of Pathology, Fazaia Medical College, Air University, Islamabad, Pakistan.
| | - Shakira Ghazanfar
- National Institute for Genomics and Advanced Biotechnology (NIGAB), National Agricultural Research Centre (NARC), Islamabad, Pakistan.
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
4
|
Tan J, Yang L, Ye M, Geng Y, Guo Y, Zou H, Hou L. Effects of cortisone in zebrafish (Danio rerio): Insights into gut microbiota interactions and molecular mechanisms underlying DNA damage and apoptosis. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135576. [PMID: 39173371 DOI: 10.1016/j.jhazmat.2024.135576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
Cortisone can enter aquatic ecosystems and pose a risk to organisms therein. However, few studies have explored the effects of cortisone on the gut microbiota of aquatic organisms. Here, we exposed zebrafish (Danio rerio) to cortisone at environmentally relevant concentrations (5.0, 50.0, or 500.0 ng L-1) for 60 days to explore its toxicological effects and their association with gut microbiota changes. The terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling assay revealed that exposure to 50 ng L-1 cortisone significantly increased the intestinal cell apoptosis rate, 8-hydroxydeoxyguanosine contents, and caspase-3 and caspase-8 activities. Moreover, the transcriptome analysis results demonstrated a notable downregulation in the expression of most differentially expressed genes associated with apoptosis pathways, as well as changes in DNA replication, oxidative stress, and drug metabolism pathways; these results indicated the occurrence of cortisone-induced stress response in zebrafish. Molecular docking analysis revealed that cortisone can bind to caspase-3 through hydrogen bonds and hydrophobic interactions but that no such interactions occur between cortisone and caspase-8. Thus, cortisone may induce oxidative DNA damage and apoptosis by activating caspase-3. Finally, the 16S rRNA sequencing results demonstrated that cortisone significantly affected microbial community structures and functions in the intestinal ecosystem. These changes may indicate gut microbiota response to cortisone-induced intestinal damage and inflammation. In conclusion, the current results clarify the mechanisms underlying intestinal response to cortisone exposure and provide a basis for evaluating the health risks of cortisone in animals.
Collapse
Affiliation(s)
- Jiefeng Tan
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China; School of Marine Sciences, Sun Yat-Sen University, Zhuhai 519082, China
| | - Lihua Yang
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai 519082, China
| | - Meixin Ye
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Yuxin Geng
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Yanfang Guo
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Hong Zou
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Liping Hou
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China.
| |
Collapse
|
5
|
Zhidkova EM, Tilova LR, Fetisov TI, Kirsanov KI, Kulikov EP, Enikeev AD, Budunova IV, Badun GA, Chernysheva MG, Shirinian VZ, Yakubovskaya MG, Lesovaya EA. Synthesis and Anti-Cancer Activity of the Novel Selective Glucocorticoid Receptor Agonists of the Phenylethanolamine Series. Int J Mol Sci 2024; 25:8904. [PMID: 39201590 PMCID: PMC11354514 DOI: 10.3390/ijms25168904] [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: 06/27/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Glucocorticoids (GCs) are widely used for treating hematological malignancies despite their multiple adverse effects. The biological response to GCs relies on glucocorticoid receptor (GR) transrepression (TR) that mediates the anticancer effects and transactivation (TA) associated with the side effects. Selective GR agonists (SEGRAs) preferentially activating GR TR could offer greater benefits in cancer treatment. One of the well-characterized SEGRAs, 2-(4-acetoxyphenyl)-2-chloro-N-methylethylammonium-chloride (CpdA), exhibited anticancer activity; however, its translational potential is limited due to chemical instability. To overcome this limitation, we obtained CpdA derivatives, CpdA-01-CpdA-08, employing two synthetic strategies and studied their anti-tumor activity: 4-(1-hydroxy-2-(piperidin-1-yl)ethyl)phenol or CpdA-03 demonstrated superior GR affinity and stability compared to CpdA. In lymphoma Granta and leukemia CEM cell lines, CpdA-03 ligand exhibited typical SEGRA properties, inducing GR TR without triggering GR TA. CpdA-03 effects on cell viability, growth, and apoptosis were similar to the reference GR ligand, dexamethasone (Dex), and the source compound CpdA. In vivo testing of CpdA-03 activity against lymphoma on the transplantable P388 murine lymphoma model showed that CpdA-03 reduced tumor volume threefold, outperforming Dex and CpdA. In conclusion, in this work, we introduce a novel SEGRA CpdA-03 as a promising agent for lymphoma treatment with fewer side effects.
Collapse
Affiliation(s)
- Ekaterina M. Zhidkova
- Department of Chemical Carcinogenesis, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center for Oncology, Kashirskoe Shosse 24-15, Moscow 115478, Russia; (E.M.Z.); (T.I.F.); (K.I.K.); (M.G.Y.)
| | - Leyla R. Tilova
- Faculty of Normal and Pathological Anatomy, H.M. Berbekov Kabardino-Balkarian State University, Chernyshevsky Str 173, Nalchik 3620004, Russia;
| | - Timur I. Fetisov
- Department of Chemical Carcinogenesis, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center for Oncology, Kashirskoe Shosse 24-15, Moscow 115478, Russia; (E.M.Z.); (T.I.F.); (K.I.K.); (M.G.Y.)
| | - Kirill I. Kirsanov
- Department of Chemical Carcinogenesis, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center for Oncology, Kashirskoe Shosse 24-15, Moscow 115478, Russia; (E.M.Z.); (T.I.F.); (K.I.K.); (M.G.Y.)
- Institute of Medicine, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, Moscow 117198, Russia
| | - Evgeny P. Kulikov
- Faculty of Oncology, I.P. Pavlov Ryazan State Medical University, Vysokovol’tnaya Str 9, Ryazan 390026, Russia;
| | - Adel D. Enikeev
- Oncogene Regulation Department, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center for Oncology, Kashirskoe Shosse 24-15, Moscow 115478, Russia;
| | - Irina V. Budunova
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, East Chicago Avenue 303, Chicago, IL 60611, USA;
| | - Gennadii A. Badun
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, Leninskiye Gory 1, Moscow 119991, Russia; (G.A.B.); (M.G.C.)
| | - Maria G. Chernysheva
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, Leninskiye Gory 1, Moscow 119991, Russia; (G.A.B.); (M.G.C.)
| | - Valerii Z. Shirinian
- Laboratory of Heterocyclic Compounds, N.D. Zelinsky Institute of Organic Chemistry, 47, Leninsky Prospect, Moscow 119991, Russia;
| | - Marianna G. Yakubovskaya
- Department of Chemical Carcinogenesis, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center for Oncology, Kashirskoe Shosse 24-15, Moscow 115478, Russia; (E.M.Z.); (T.I.F.); (K.I.K.); (M.G.Y.)
- Institute of Medicine, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, Moscow 117198, Russia
| | - Ekaterina A. Lesovaya
- Department of Chemical Carcinogenesis, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center for Oncology, Kashirskoe Shosse 24-15, Moscow 115478, Russia; (E.M.Z.); (T.I.F.); (K.I.K.); (M.G.Y.)
- Institute of Medicine, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, Moscow 117198, Russia
- Faculty of Oncology, I.P. Pavlov Ryazan State Medical University, Vysokovol’tnaya Str 9, Ryazan 390026, Russia;
| |
Collapse
|
6
|
Kutpruek S, Suksri K, Maneethorn P, Semprasert N, Yenchitsomanus PT, Kooptiwut S. Imatinib prevents dexamethasone-induced pancreatic β-cell apoptosis via decreased TRAIL and DR5. J Cell Biochem 2023; 124:1309-1323. [PMID: 37555250 DOI: 10.1002/jcb.30450] [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: 02/22/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023]
Abstract
Prolonged administration of dexamethasone, a potent anti-inflammatory drug, can lead to steroid-induced diabetes. Imatinib, a medication commonly prescribed for chronic myeloid leukemia (CML), has been shown to improve diabetes in CML patients. Our recent study demonstrated that dexamethasone induces pancreatic β-cell apoptosis by upregulating the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptor, death receptor 5 (DR5). We hypothesized that imatinib may protect against dexamethasone-induced pancreatic β-cell apoptosis by reducing the expression of TRAIL and DR5, thereby favorably modulating downstream effectors in apoptotic pathways. We test this hypothesis by assessing the effects of imatinib on dexamethasone-induced apoptosis in rat insulinoma cell line cells. As anticipated, dexamethasone treatment led to increased TRAIL and DR5 expression, as well as an elevation in superoxide production. Conversely, expression of the TRAIL decoy receptor (DcR1) was decreased. Moreover, key effectors in the extrinsic and intrinsic apoptosis pathways, such as B-cell lymphoma 2 (BCL-2) associated X (BAX), nuclear factor kappa B (NF-κb), P73, caspase 8, and caspase 9, were upregulated, while the antiapoptotic protein BCL-2 was downregulated. Interestingly and importantly, imatinib at a concentration of 10 µM reversed the effect of dexamethasone on TRAIL, DR5, DcR1, superoxide production, BAX, BCL-2, NF-κB, P73, caspase 3, caspase 8, and caspase 9. Similar effects of imatinib on dexamethasone-induced TRAIL and DR5 expression were also observed in isolated mouse islets. Taken together, our findings suggest that imatinib protects against dexamethasone-induced pancreatic β-cell apoptosis by reducing TRAIL and DR5 expression and modulating downstream effectors in the extrinsic and intrinsic apoptosis pathways.
Collapse
Affiliation(s)
- Suchanoot Kutpruek
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanchana Suksri
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Petcharee Maneethorn
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Namoiy Semprasert
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Research Department, Division of Molecular Medicine, Mahidol University, Bangkok, Thailand
| | - Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
7
|
Tena-Garitaonaindia M, Arredondo-Amador M, Mascaraque C, Asensio M, Marin JJG, Martínez-Augustin O, Sánchez de Medina F. MODULATION OF INTESTINAL BARRIER FUNCTION BY GLUCOCORTICOIDS: LESSONS FROM PRECLINICAL MODELS. Pharmacol Res 2022; 177:106056. [PMID: 34995794 DOI: 10.1016/j.phrs.2022.106056] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/22/2021] [Accepted: 01/01/2022] [Indexed: 12/15/2022]
Abstract
Glucocorticoids (GCs) are widely used drugs for their anti-inflammatory and immunosuppressant effects, but they are associated with multiple adverse effects. Despite their frequent oral administration, relatively little attention has been paid to the effects of GCs on intestinal barrier function. In this review, we present a summary of the published studies on this matter carried out in animal models and cultured cells. In cultured intestinal epithelial cells, GCs have variable effects in basal conditions and generally enhance barrier function in the presence of inflammatory cytokines such as tumor necrosis factor (TNF). In turn, in rodents and other animals, GCs have been shown to weaken barrier function, with increased permeability and lower production of IgA, which may account for some features observed in stress models. When given to animals with experimental colitis, barrier function may be debilitated or strengthened, despite a positive anti-inflammatory activity. In sepsis models, GCs have a barrier-enhancing effect. These effects are probably related to the inhibition of epithelial cell proliferation and wound healing, modulation of the microbiota and mucus production, and interference with the mucosal immune system. The available information on underlying mechanisms is described and discussed.
Collapse
Affiliation(s)
- Mireia Tena-Garitaonaindia
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María Arredondo-Amador
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Mascaraque
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Maitane Asensio
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Wang M, Ibeagha-Awemu EM. Impacts of Epigenetic Processes on the Health and Productivity of Livestock. Front Genet 2021; 11:613636. [PMID: 33708235 PMCID: PMC7942785 DOI: 10.3389/fgene.2020.613636] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/21/2020] [Indexed: 12/23/2022] Open
Abstract
The dynamic changes in the epigenome resulting from the intricate interactions of genetic and environmental factors play crucial roles in individual growth and development. Numerous studies in plants, rodents, and humans have provided evidence of the regulatory roles of epigenetic processes in health and disease. There is increasing pressure to increase livestock production in light of increasing food needs of an expanding human population and environment challenges, but there is limited related epigenetic data on livestock to complement genomic information and support advances in improvement breeding and health management. This review examines the recent discoveries on epigenetic processes due to DNA methylation, histone modification, and chromatin remodeling and their impacts on health and production traits in farm animals, including bovine, swine, sheep, goat, and poultry species. Most of the reports focused on epigenome profiling at the genome-wide or specific genic regions in response to developmental processes, environmental stressors, nutrition, and disease pathogens. The bulk of available data mainly characterized the epigenetic markers in tissues/organs or in relation to traits and detection of epigenetic regulatory mechanisms underlying livestock phenotype diversity. However, available data is inadequate to support gainful exploitation of epigenetic processes for improved animal health and productivity management. Increased research effort, which is vital to elucidate how epigenetic mechanisms affect the health and productivity of livestock, is currently limited due to several factors including lack of adequate analytical tools. In this review, we (1) summarize available evidence of the impacts of epigenetic processes on livestock production and health traits, (2) discuss the application of epigenetics data in livestock production, and (3) present gaps in livestock epigenetics research. Knowledge of the epigenetic factors influencing livestock health and productivity is vital for the management and improvement of livestock productivity.
Collapse
Affiliation(s)
- Mengqi Wang
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC, Canada
- Department of Animal Science, Laval University, Quebec, QC, Canada
| | - Eveline M. Ibeagha-Awemu
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC, Canada
| |
Collapse
|