1
|
Pérez-Gómez A, González-Brusi L, Flores-Borobia I, Martínez De Los Reyes N, Toledano-Díaz A, López-Sebastián A, Santiago Moreno J, Ramos-Ibeas P, Bermejo-Álvarez P. PPARG is dispensable for bovine embryo development up to tubular stages†. Biol Reprod 2024; 111:557-566. [PMID: 38832705 PMCID: PMC11402522 DOI: 10.1093/biolre/ioae083] [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: 03/08/2024] [Revised: 04/25/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024] Open
Abstract
Following blastocyst hatching, ungulate embryos undergo a prolonged preimplantation period termed conceptus elongation. Conceptus elongation constitutes a highly susceptible period for embryonic loss, and the embryonic requirements during this process are largely unknown, but multiple lipid compounds have been identified in the fluid nourishing the elongating conceptuses. Peroxisome proliferator-activated receptors mediate the signaling actions of prostaglandins and other lipids, and, between them, PPARG has been pointed out to play a relevant role in conceptus elongation by a functional study that depleted PPARG in both uterus and conceptus. The objective of this study has been to determine if embryonic PPARG is required for bovine embryo development. To that aim, we have generated bovine PPARG knock-out embryos in vitro using two independent gene ablation strategies and assessed their developmental ability. In vitro development to Day 8 blastocyst was unaffected by PPARG ablation, as total, inner cell mass, and trophectoderm cell numbers were similar between wild-type and knock-out D8 embryos. In vitro post-hatching development to D12 was also comparable between different genotypes, as embryo diameter, epiblast cell number, embryonic disk formation, and hypoblast migration rates were unaffected by the ablation. The development of tubular stages equivalent to E14 was assessed in vivo, following a heterologous embryo transfer experiment, observing that the development of extra-embryonic membranes and of the embryonic disk was not altered by PPARG ablation. In conclusion, PPARG ablation did not impaired bovine embryo development up to tubular stages.
Collapse
Affiliation(s)
- Alba Pérez-Gómez
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Leopoldo González-Brusi
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Inés Flores-Borobia
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Nuria Martínez De Los Reyes
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Adolfo Toledano-Díaz
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Antonio López-Sebastián
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Julián Santiago Moreno
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Priscila Ramos-Ibeas
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Pablo Bermejo-Álvarez
- Animal Reproduction Department, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
2
|
Wang HL, Liu Y, Zhou T, Gao L, Li J, Wu X, Yin YL. Uridine affects amino acid metabolism in sow-piglets model and increases viability of pTr2 cells. Front Nutr 2022; 9:1018349. [DOI: 10.3389/fnut.2022.1018349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAs an important nucleoside precursor in salvage synthesis pathway of uridine monophosphate, uridine (UR) is the most abundant nucleotide in sow milk. This study aimed to investigate the effects of maternal UR supplementation during second trimester of gestation on reproductive performance and amino acid metabolism of Sows.ResultsResults showed that compared to CON group, the average number of stillborn piglets per litter was significantly reduced (P < 0.05) with higher average piglet weight at birth in UR group (P = 0.083). Besides, dietary UR supplementation significantly increased TP in sow serum, BUN content in cord serum, and TP and ALB in newborn piglet serum (P < 0.05); but decreased AST level in sow serum and BUN level in piglet serum (P < 0.05). Importantly, free amino acids profile in sow serum newborn piglet serum and colostrum was changed by maternal UR supplementation during day 60 of pregnancy, as well as the expression of amino acids transporter (P < 0.05). In addition, from 100 to 2,000 μM UR can increased the viability of pTr2 cells. The UR exhibited higher distribution of G1/M phase of cell cycle at 400 μM compared with 0 μM, and reduced S-phases of cell cycle compared with 0 and 100μM (P < 0.05).ConclusionSupplementation of uridine during day 60 of pregnancy can improve reproductive performance, regulate amino acid metabolism of sows and their offspring, and increase the viability of pTr2 cells.
Collapse
|
3
|
Guo Y, Song W, Yang Y. Inhibition of ALKBH5-mediated m 6 A modification of PPARG mRNA alleviates H/R-induced oxidative stress and apoptosis in placenta trophoblast. ENVIRONMENTAL TOXICOLOGY 2022; 37:910-924. [PMID: 34995009 DOI: 10.1002/tox.23454] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
The alpha-ketoglutarate-dependent (ALKB) homolog 5 (ALKBH5), an m6 A demethylase, has been reported to be involved in the pathogenesis of preeclampsia (PE), but the exact mechanism requires further investigation. RT-qPCR or Western blotting were used to determine ALKBH5 and peroxisome proliferator-activated receptor gamma (PPARG) expression in placentas from PE patients and normal volunteers, as well as in HTR-8/SVneo cells treated with hypoxia/reoxygenation (H/R). Our results showed that the expression of ALKBH5 was significantly upregulated and PPARG was downregulated in preeclamptic placentas and H/R-treated cells. ALKBH5 interference reduced m6 A levels of PPARG mRNA, and increased PPARG mRNA stability and promoted PPARG translation level. In addition, ALKBH5 silencing increased the cell proliferation, migration, and vimentin protein level, and inhibited cell apoptosis, oxidative stress, and protein levels of endoglin (ENG) and E-cadherin in H/R-treated cells, whereas PPARG interference reversed these effects. Furthermore, PPARG repressed the H3K9me2 levels at activated leukocyte cell adhesion molecule (ALCAM) promoter region by increasing the expression and activity of lysine demethylase 3B (KDM3B). ALCAM inhibition reversed the effects of PPARG overexpression on H/R-treated cell functions. PKF115-584 suppressed the effects of ALKBH5 interference on the behaviors of H/R-treated cells. Finally, inhibition of ALKBH5 alleviates PE-like features in pregnant mice. Inhibition of ALKBH5 promotes KDM3B-mediated ALCAM demethylation by facilitating PPARG mRNA m6 A modification, and further activates the Wnt/β-catenin pathway, and in turn alleviates PE progression.
Collapse
Affiliation(s)
- Yongping Guo
- Department of Obstetrics, Baoji Maternal and Child Health Care Hospital, Baoji, China
| | - Wenxia Song
- Department of Obstetrics, Baoji Maternal and Child Health Care Hospital, Baoji, China
| | - Yali Yang
- Department of Obstetrics, Baoji Maternal and Child Health Care Hospital, Baoji, China
| |
Collapse
|
4
|
Meister S, Hahn L, Beyer S, Paul C, Mitter S, Kuhn C, von Schönfeldt V, Corradini S, Sudan K, Schulz C, Kolben TM, Mahner S, Jeschke U, Kolben T. Regulation of Epigenetic Modifications in the Placenta during Preeclampsia: PPARγ Influences H3K4me3 and H3K9ac in Extravillous Trophoblast Cells. Int J Mol Sci 2021; 22:ijms222212469. [PMID: 34830351 PMCID: PMC8622744 DOI: 10.3390/ijms222212469] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to analyze the expression of peroxisome proliferator-activated receptor γ (PPARγ) and retinoid X receptor α (RxRα), a binding heterodimer playing a pivotal role in the successful trophoblast invasion, in the placental tissue of preeclamptic patients. Furthermore, we aimed to characterize a possible interaction between PPARγ and H3K4me3 (trimethylated lysine 4 of the histone H3), respectively H3K9ac (acetylated lysine 9 of the histone H3), to illuminate the role of histone modifications in a defective trophoblast invasion in preeclampsia (PE). Therefore, the expression of PPARγ and RxRα was analyzed in 26 PE and 25 control placentas by immunohistochemical peroxidase staining, as well as the co-expression with H3K4me3 and H3K9ac by double immunofluorescence staining. Further, the effect of a specific PPARγ-agonist (Ciglitazone) and PPARγ-antagonist (T0070907) on the histone modifications H3K9ac and H3K4me3 was analyzed in vitro. In PE placentas, we found a reduced expression of PPARγ and RxRα and a reduced co-expression with H3K4me3 and H3K9ac in the extravillous trophoblast (EVT). Furthermore, with the PPARγ-antagonist treated human villous trophoblast (HVT) cells and primary isolated EVT cells showed higher levels of the histone modification proteins whereas treatment with the PPARγ-agonist reduced respective histone modifications. Our results show that the stimulation of PPARγ-activity leads to a reduction of H3K4me3 and H3K9ac in trophoblast cells, but paradoxically decreases the nuclear PPARγ expression. As the importance of PPARγ, being involved in a successful trophoblast invasion has already been investigated, our results reveal a pathophysiologic connection between PPARγ and the epigenetic modulation via H3K4me3 and H3K9ac in PE.
Collapse
Affiliation(s)
- Sarah Meister
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
- Correspondence: (S.M.); (U.J.); Tel.: +49-89-4400-54266 (S.M.); Fax: +49-89-4400-54916 (S.M.)
| | - Laura Hahn
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Susanne Beyer
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Corinna Paul
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Sophie Mitter
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Christina Kuhn
- Department of Gynecology and Obstetrics, University Hospital Augsburg, 86156 Augsburg, Germany;
| | - Viktoria von Schönfeldt
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Stefanie Corradini
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany;
| | - Kritika Sudan
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (K.S.); (C.S.)
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (K.S.); (C.S.)
| | - Theresa Maria Kolben
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Sven Mahner
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Udo Jeschke
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
- Department of Gynecology and Obstetrics, University Hospital Augsburg, 86156 Augsburg, Germany;
- Correspondence: (S.M.); (U.J.); Tel.: +49-89-4400-54266 (S.M.); Fax: +49-89-4400-54916 (S.M.)
| | - Thomas Kolben
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| |
Collapse
|
5
|
Apigenin Alleviates Liver Fibrosis by Inhibiting Hepatic Stellate Cell Activation and Autophagy via TGF- β1/Smad3 and p38/PPAR α Pathways. PPAR Res 2021; 2021:6651839. [PMID: 33574836 PMCID: PMC7861947 DOI: 10.1155/2021/6651839] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/10/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
Objective The aim of this study is to confirm the hepatocellular protective functions of apigenin and the molecular mechanism on liver fibrosis in mice. Methods Carbon tetrachloride (CCl4) and bile duct ligature (BDL) mouse fibrosis models were used to investigate the effects of apigenin on liver fibrosis. Sixty-six male C57 mice were randomly divided into eight groups, including the vehicle group, CCl4 group, CCl4+L-apigenin (20 mg/kg) group, CCl4+H-apigenin (40 mg/kg) group, sham group, BDL group, BDL+L-apigenin(20 mg/kg) group, and BDL+H-apigenin(40 mg/kg) group. Serum liver enzymes (ALT and AST), proteins associated with autophagy, and indicators linked with the TGF-β1/Smad3 and p38/PPARα pathways were detected using qRT-PCR, immunohistochemical staining, and western blotting. Results Our findings confirmed that apigenin could decrease the levels of ALT and AST, suppress the generation of ECM, inhibit the activation of HSCs, regulate the balance of MMP2 and TIMP1, reduce the expression of autophagy-linked protein, and restrain the TGF-β1/Smad3 and p38/PPARα pathways. Conclusion Apigenin could alleviate liver fibrosis by inhibiting hepatic stellate cell activation and autophagy via TGF-β1/Smad3 and p38/PPARα pathways.
Collapse
|
6
|
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
|
7
|
Ji J, Wu L, Feng J, Mo W, Wu J, Yu Q, Li S, Zhang J, Dai W, Xu X, Mao Y, Xu S, Chen K, Li J, Guo C. Cafestol preconditioning attenuates apoptosis and autophagy during hepatic ischemia-reperfusion injury by inhibiting ERK/PPARγ pathway. Int Immunopharmacol 2020; 84:106529. [PMID: 32344356 DOI: 10.1016/j.intimp.2020.106529] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/01/2020] [Accepted: 04/18/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The study was aimed to explore the hepatocellular protective functions of cafestol during hepatic ischemia-reperfusion injury and the possible mechanisms. METHODS Ninety male Balb/c mice were randomly divided into seven groups, including normal control group, L-cafestol(20mg/kg) group, H-cafestol(40mg/kg) group, sham group, IR group, L-cafestol(20mg/kg) + IR group, H-cafestol(40mg/kg) + IR group. Serum liver enzymes (ALT, AST), inflammation mediators, proteins associated with apoptosis and autophagy, indicators linked with ERK/PPARγ pathway, and liver histopathology were measured using ELISA, qRT-PCR, immunohistochemical staining, and western blotting at 2, 8, and 24 hours after reperfusion. RESULTS Our findings confirmed that cafestol preconditioning groups could reduce the levels of ALT and AST, alleviate liver pathological damage, suppress the release of inflammation mediators, inhibit the production of pro-apoptosis protein including caspase-3, caspase-9 and Bax, decrease the expression of autophagy-linked protein including Beclin-1 and LC3, increase anti-apoptosis protein Bcl-2, and restrain the activation of ERK and PPARγ. CONCLUSION Cafestol preconditioning could attenuate inflammatory response, apoptosis and autophagy on hepatic ischemia reperfusion injury by suppressing ERK/PPARγ pathway.
Collapse
Affiliation(s)
- Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Wenhui Mo
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China; Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai 200032, China; Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China; Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China
| | - Xuanfu Xu
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, China
| | - Yuqing Mao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Shizan Xu
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Jinshan, Shanghai 201508, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
8
|
Peroxisome proliferator-activated receptor β/δ and γ agonists differentially affect prostaglandin E2 and cytokine synthesis and nutrient transporter expression in porcine trophoblast cells during implantation. Theriogenology 2020; 152:36-46. [PMID: 32361305 DOI: 10.1016/j.theriogenology.2020.04.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/24/2020] [Accepted: 04/18/2020] [Indexed: 01/09/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor family of ligand-dependent transcription factors. PPARs have been shown to be important regulators of female reproductive functions, including conceptus development and placenta formation. This study examines the effect of PPARβ/δ and PPARγ agonists and antagonists on (1) the synthesis of prostaglandin (PG) E2, interleukin (IL) 6, interferon (IFN) γ, and tumor necrosis factor (TNF) α and (2) the mRNA expression of genes encoding nutrient transporters and/or binding proteins in Day 15 conceptus trophoblast cells. The study also examines whether PPAR agonist-modulated IL6, IFNγ, and TNFα secretion is mediated via mitogen-activated protein kinase (MAPK) pathways. Trophoblast cells were exposed to L-165,041 (a PPARβ/δ agonist) or rosiglitazone (a PPARγ agonist) in the presence or absence of GSK3787 (a PPARβ/δ antagonist) or GW9662 (a PPARγ antagonist) or in the presence or absence of U0126 (a MAPK inhibitor). Rosiglitazone stimulated PGE synthase and IFNG mRNA expression in trophoblast cells and enhanced PGE2 concentrations in the incubation medium. Moreover, cells treated with rosiglitazone exhibited increased abundance of the solute carrier organic anion transporter family member 2A1 (SLCO2A1, a PG transporter) and of fatty acid binding protein (FABP) 5 transcripts. All these effects were abolished by the addition of GW9662, which indicates that the action of rosiglitazone is PPARγ-dependent in the studied cells. L-165,041 inhibited TNFα synthesis and decreased the mRNA expression of FABP3 and IL6 in trophoblast cells. However, this effect was not abolished by the addition of GSK3787 into the incubation medium, suggesting that L-165,041 action is independent of PPARβ/δ. The inhibitory effect of L-165,041 on TNFα concentration and the stimulatory effect of rosiglitazone on IFNγ accumulation in the medium were not observed in the presence of the MAPK inhibitor, suggesting that the action of both agonists may be mediated by MAPKs. In conclusion, PPARβ/δ and PPARγ agonists are differentially involved in the trophoblast expression of genes related to conceptus development and implantation in pigs. Furthermore, L-165,041 and rosiglitazone may have PPAR-dependent and -independent effects in conceptus trophoblast cells.
Collapse
|
9
|
Cheng V, Dasgupta S, Reddam A, Volz DC. Ciglitazone-a human PPARγ agonist-disrupts dorsoventral patterning in zebrafish. PeerJ 2019; 7:e8054. [PMID: 31741801 PMCID: PMC6858815 DOI: 10.7717/peerj.8054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/17/2019] [Indexed: 01/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-activated transcription factor that regulates lipid/glucose homeostasis and adipocyte differentiation. While the role of PPARγ in adipogenesis and diabetes has been extensively studied, little is known about PPARγ function during early embryonic development. Within zebrafish, maternally-loaded pparγ transcripts are present within the first 6 h post-fertilization (hpf), and de novo transcription of zygotic pparγ commences at ~48 hpf. Since maternal pparγ transcripts are elevated during a critical window of cell fate specification, the objective of this study was to test the hypothesis that PPARγ regulates gastrulation and dorsoventral patterning during zebrafish embryogenesis. To accomplish this objective, we relied on (1) ciglitazone as a potent PPARγ agonist and (2) a splice-blocking, pparγ-specific morpholino to knockdown pparγ. We found that initiation of ciglitazone-a potent human PPARγ agonist-exposure by 4 hpf resulted in concentration-dependent effects on dorsoventral patterning in the absence of epiboly defects during gastrulation, leading to ventralized embryos by 24 hpf. Interestingly, ciglitazone-induced ventralization was reversed by co-exposure with dorsomorphin, a bone morphogenetic protein signaling inhibitor that induces strong dorsalization within zebrafish embryos. Moreover, mRNA-sequencing revealed that lipid- and cholesterol-related processes were affected by exposure to ciglitazone. However, pparγ knockdown did not block ciglitazone-induced ventralization, suggesting that PPARγ is not required for dorsoventral patterning nor involved in ciglitazone-induced toxicity within zebrafish embryos. Our findings point to a novel, PPARγ-independent mechanism of action and phenotype following ciglitazone exposure during early embryonic development.
Collapse
Affiliation(s)
- Vanessa Cheng
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| | - Subham Dasgupta
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| | - Aalekhya Reddam
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| | - David C Volz
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| |
Collapse
|