Altered Transcription Factor Binding and Gene Bivalency in Islets of Intrauterine Growth Retarded Rats
Abstract
:1. Introduction
2. Materials and Methods
2.1. Animal Model
2.2. Islet Isolation
2.3. Chromatin Preparation and Chromatin Immunoprecipitation (ChIP)
2.4. ChIP-Seq and Data Analysis
3. Results
3.1. ChIP-Seq Analysis
3.2. IUGR-Induced Histone Modification Changes in Islets Correlated with Transcription Activities and Phenotypes
3.3. IUGR Altered Histone Modifications at Critical Transcription Factor Binding Motifs
3.3.1. Two-Week-Old Animals
3.3.2. Ten-Week-Old Animals
3.3.3. Persistent Changes from Two- to Ten-Week-Old Animals
3.4. Poised States of Potential Bivalent Genes Were Altered in IUGR Islets
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Newsome, C.A.; Shiell, A.W.; Fall, C.H.; Phillips, D.I.; Shier, R.; Law, C.M. Is birth weight related to later glucose and insulin metabolism?—A systematic review. Diabet. Med. 2003, 20, 339–348. [Google Scholar] [CrossRef]
- Bianco-Miotto, T.; Craig, J.M.; Gasser, Y.P.; van Dijk, S.J.; Ozanne, S.E. Epigenetics and DOHaD: From basics to birth and beyond. J. Dev. Orig. Health Dis. 2017, 8, 513–519. [Google Scholar] [CrossRef] [PubMed]
- Simmons, R.A.; Templeton, L.J.; Gertz, S.J. Intrauterine growth retardation leads to the development of type 2 diabetes in the rat. Diabetes 2001, 50, 2279–2286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gatford, K.L.; Simmons, R.A.; De Blasio, M.J.; Robinson, J.S.; Owens, J.A. Review: Placental programming of postnatal diabetes and impaired insulin action after IUGR. Placenta 2010, 31, S60–S65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ogata, E.S.; Bussey, M.E.; Finley, S. Altered gas exchange, limited glucose and branched chain amino acids, and hypoinsulinism retard fetal growth in the rat. Metabolism 1986, 35, 970–977. [Google Scholar] [CrossRef]
- Jaeckle Santos, L.J.; Li, C.; Doulias, P.T.; Ischiropoulos, H.; Worthen, G.S.; Simmons, R.A. Neutralizing Th2 inflammation in neonatal islets prevents β-cell failure in adult IUGR rats. Diabetes 2014, 63, 1672–1684. [Google Scholar] [CrossRef] [Green Version]
- Stoffers, D.A.; Desai, B.M.; DeLeon, D.D.; Simmons, R.A. Neonatal exendin-4 prevents the development of diabetes in the intrauterine growth retarded rat. Diabetes 2003, 52, 734–740. [Google Scholar] [CrossRef] [Green Version]
- Ham, J.N.; Crutchlow, M.F.; Desai, B.M.; Simmons, R.A.; Stoffers, D.A. Exendin-4 normalizes islet vascularity in intrauterine growth restricted rats: Potential role of VEGF. Pediatr. Res. 2009, 66, 42–46. [Google Scholar] [CrossRef] [Green Version]
- Rashid, C.S.; Lien, Y.C.; Bansal, A.; Jaeckle-Santos, L.J.; Li, C.; Won, K.J.; Simmons, R.A. Transcriptomic analysis reveals novel mechanisms mediating islet dysfunction in the intrauterine growth-restricted rat. Endocrinology 2018, 159, 1035–1049. [Google Scholar] [CrossRef] [Green Version]
- Li, B.; Carey, M.; Workman, J.L. The role of chromatin during transcription. Cell 2007, 128, 707–719. [Google Scholar] [CrossRef] [Green Version]
- Bernstein, B.E.; Meissner, A.; Lander, E.S. The mammalian epigenome. Cell 2007, 128, 669–681. [Google Scholar] [CrossRef] [Green Version]
- Mellor, J. The dynamics of chromatin remodeling at promoters. Mol. Cell. 2005, 19, 147–157. [Google Scholar] [CrossRef] [PubMed]
- Pinney, S.E.; Simmons, R.A. Epigenetic mechanisms in the development of type 2 diabetes. Trends Endocrinol. Metab. 2010, 21, 223–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sterns, J.D.; Smith, C.B.; Steele, J.R.; Stevenson, K.L.; Gallicano, G.I. Epigenetics and type II diabetes mellitus: Underlying mechanisms of prenatal predisposition. Front. Cell Dev. Biol. 2014, 2, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosen, E.D.; Kaestner, K.H.; Natarajan, R.; Patti, M.E.; Sallari, R.; Sander, M.; Susztak, K. Epigenetics and epigenomics: Implications for diabetes and obesity. Diabetes 2018, 67, 1923–1931. [Google Scholar] [CrossRef] [Green Version]
- Smail, H.O. The epigenetics of diabetes, obesity, overweight and cardiovascular disease. AIMS Genet. 2019, 6, 36–45. [Google Scholar] [CrossRef]
- Ling, C.; Rönn, T. Epigenetics in human obesity and type 2 diabetes. Cell Metab. 2019, 29, 1028–1044. [Google Scholar] [CrossRef] [Green Version]
- Park, J.H.; Stoffers, D.A.; Nicholls, R.D.; Simmons, R.A. Development of type 2 diabetes following intrauterine growth retardation in rats is associated with progressive epigenetic silencing of Pdx1. J. Clin. Investig. 2008, 118, 2316–2324. [Google Scholar] [CrossRef] [Green Version]
- Pinney, S.E.; Jaeckle Santos, L.J.; Han, Y.; Stoffers, D.A.; Simmons, R.A. Exendin-4 increases histone acetylase activity and reverses epigenetic modifications that silence Pdx1 in the intrauterine growth retarded rat. Diabetologia 2011, 54, 2606–2614. [Google Scholar] [CrossRef] [Green Version]
- Allis, C.D.; Jenuwein, T. The molecular hallmarks of epigenetic control. Nat. Rev. Genet. 2016, 17, 487–500. [Google Scholar] [CrossRef]
- Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernstein, E.; Allis, C.D. RNA meets chromatin. Genes Dev. 2005, 19, 1635–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thompson, R.F.; Fazzari, M.J.; Niu, H.; Barzilai, N.; Simmons, R.A.; Greally, J.M. Experimental intrauterine growth restriction induces alterations in DNA methylation and gene expression in pancreatic islets of rats. J. Biol. Chem. 2010, 285, 15111–15118. [Google Scholar] [CrossRef] [Green Version]
- Bramswig, N.C.; Everett, L.J.; Schug, J.; Dorrell, C.; Liu, C.; Luo, Y.; Streeter, P.R.; Naji, A.; Grompe, M.; Kaestner, K.H. Epigenomic plasticity enables human pancreatic α to β cell reprogramming. J. Clin. Investig. 2013, 123, 1275–1284. [Google Scholar] [CrossRef] [Green Version]
- Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef] [PubMed]
- Li, H. A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data. Bioinformatics 2011, 27, 2987–2993. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Liu, T.; Meyer, C.A.; Eeckhoute, J.; Johnson, D.S.; Bernstein, B.E.; Nusbaum, C.; Myers, R.M.; Brown, M.; Li, W.; et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 2008, 9, R137. [Google Scholar] [CrossRef] [Green Version]
- Shen, L.; Shao, N.Y.; Liu, X.; Maze, I.; Feng, J.; Nestler, E.J. diffReps: Detecting differential chromatin modification sites from ChIP-seq data with biological replicates. PLoS ONE 2013, 8, e65598. [Google Scholar] [CrossRef] [Green Version]
- Heinz, S.; Benner, C.; Spann, N.; Bertolino, E.; Lin, Y.C.; Laslo, P.; Cheng, J.X.; Murre, C.; Singh, H.; Glass, C.K. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 2010, 38, 576–589. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.; Cheung, T.H.; Charville, G.W.; Hurgo, B.M.; Leavitt, T.; Shih, J.; Brunet, A.; Rando, T.A. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 2013, 4, 189–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Creyghton, M.P.; Cheng, A.W.; Welstead, G.G.; Kooistra, T.; Carey, B.W.; Steine, E.J.; Hanna, J.; Lodato, M.A.; Frampton, G.M.; Sharp, P.A.; et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 2010, 107, 21931–21936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rada-Iglesias, A.; Bajpai, R.; Swigut, T.; Brugmann, S.A.; Flynn, R.A.; Wysocka, J. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 2011, 470, 279–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zentner, G.E.; Tesar, P.J.; Scacheri, P.C. Epigenetic signatures distinguish multiple classes of enhancers with distinct cellular functions. Genome Res. 2011, 21, 1273–1283. [Google Scholar] [CrossRef] [Green Version]
- Colsoul, B.; Schraenen, A.; Lemaire, K.; Quintens, R.; Van Lommel, L.; Segal, A.; Owsianik, G.; Talavera, K.; Voets, T.; Margolskee, R.F.; et al. Loss of high-frequency glucose-induced Ca2+ oscillations in pancreatic islets correlates with impaired glucose tolerance in Trpm5-/- mice. Proc. Natl. Acad. Sci. USA 2010, 107, 5208–5213. [Google Scholar] [CrossRef] [Green Version]
- Brixel, L.R.; Monteilh-Zoller, M.K.; Ingenbrandt, C.S.; Fleig, A.; Penner, R.; Enklaar, T.; Zabel, B.U.; Prawitt, D. TRPM5 regulates glucose-stimulated insulin secretion. Pflug. Arch. 2010, 460, 69–76. [Google Scholar] [CrossRef] [Green Version]
- Campbell, C.T.; Kolesar, J.E.; Kaufman, B.A. Mitochondrial transcription factor A regulates mitochondrial transcription initiation, DNA packaging, and genome copy number. Biochim. Biophys. Acta 2012, 1819, 921–929. [Google Scholar] [CrossRef]
- Gauthier, B.R.; Wiederkehr, A.; Baquié, M.; Dai, C.; Powers, A.C.; Kerr-Conte, J.; Pattou, F.; MacDonald, R.J.; Ferrer, J.; Wollheim, C.B. PDX1 deficiency causes mitochondrial dysfunction and defective insulin secretion through TFAM suppression. Cell Metab. 2009, 10, 110–118. [Google Scholar] [CrossRef] [Green Version]
- Nile, D.L.; Brown, A.E.; Kumaheri, M.A.; Blair, H.R.; Heggie, A.; Miwa, S.; Cree, L.M.; Payne, B.; Chinnery, P.F.; Brown, L.; et al. Age-related mitochondrial DNA depletion and the impact on pancreatic Beta cell function. PLoS ONE 2014, 9, e115433. [Google Scholar] [CrossRef] [Green Version]
- Simmons, R.A.; Suponitsky-Kroyter, I.; Selak, M.A. Progressive accumulation of mitochondrial DNA mutations and decline in mitochondrial function lead to beta-cell failure. J. Biol. Chem. 2005, 280, 28785–28791. [Google Scholar] [CrossRef] [Green Version]
- Horii, Y.; Beeler, J.F.; Sakaguchi, K.; Tachibana, M.; Miki, T. A novel oncogene, ost, encodes a guanine nucleotide exchange factor that potentially links Rho and Rac signaling pathways. EMBO J. 1994, 13, 4776–4786. [Google Scholar] [CrossRef] [PubMed]
- Kowluru, A. Role of G-proteins in islet function in health and diabetes. Diabetes Obes. Metab. 2017, 19, 63–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Møller, L.L.V.; Klip, A.; Sylow, L. Rho GTPases-Emerging regulators of glucose homeostasis and metabolic health. Cells 2019, 8, 434. [Google Scholar] [CrossRef] [Green Version]
- Che, M.; Ortiz, D.F.; Arias, I.M. Primary structure and functional expression of a cDNA encoding the bile canalicular, purine-specific Na(+)-nucleoside cotransporter. J. Biol. Chem. 1995, 270, 13596–13599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antonioli, L.; Blandizzi, C.; Csóka, B.; Pacher, P.; Haskó, G. Adenosine signalling in diabetes mellitus—Pathophysiology and therapeutic considerations. Nat. Rev. Endocrinol. 2015, 11, 228–241. [Google Scholar] [CrossRef] [PubMed]
- Faulhaber-Walter, R.; Jou, W.; Mizel, D.; Li, L.; Zhang, J.; Kim, S.M.; Huang, Y.; Chen, M.; Briggs, J.P.; Gavrilova, O.; et al. Impaired glucose tolerance in the absence of adenosine A1 receptor signaling. Diabetes 2011, 60, 2578–2587. [Google Scholar] [CrossRef] [Green Version]
- Shy, M.E. Peripheral neuropathies caused by mutations in the myelin protein zero. J. Neurol. Sci. 2006, 242, 55–66. [Google Scholar] [CrossRef] [PubMed]
- Woods, S.C.; Porte, D. Neural control of the endocrine pancreas. Physiol. Rev. 1974, 54, 596–619. [Google Scholar] [CrossRef]
- Kohnert, K.D.; Axcrona, U.M.; Hehmke, B.; Klöting, I.; Sundler, F.; Ahrén, B. Islet neuronal abnormalities associated with impaired insulin secretion in type 2 diabetes in the Chinese hamster. Regul. Pept. 1999, 82, 71–79. [Google Scholar] [CrossRef]
- Ebato, C.; Uchida, T.; Arakawa, M.; Komatsu, M.; Ueno, T.; Komiya, K.; Azuma, K.; Hirose, T.; Tanaka, K.; Kominami, E.; et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab. 2008, 8, 325–332. [Google Scholar] [CrossRef] [Green Version]
- Lu, T.; Zhu, Z.; Wu, J.; She, H.; Han, R.; Xu, H.; Qin, Z.H. DRAM1 regulates autophagy and cell proliferation via inhibition of the phosphoinositide 3-kinase-Akt-mTOR-ribosomal protein S6 pathway. Cell Commun. Signal. 2019, 17, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, M.; Gwak, J.; Hwang, S.; Yang, S.; Jeong, S.M. Mitochondrial GPT2 plays a pivotal role in metabolic adaptation to the perturbation of mitochondrial glutamine metabolism. Oncogene 2019, 38, 4729–4738. [Google Scholar] [CrossRef] [PubMed]
- Ding, J.; Kuo, M.L.; Su, L.; Xue, L.; Luh, F.; Zhang, H.; Wang, J.; Lin, T.G.; Zhang, K.; Chu, P.; et al. Human mitochondrial pyrroline-5-carboxylate reductase 1 promotes invasiveness and impacts survival in breast cancers. Carcinogenesis 2017, 38, 519–531. [Google Scholar] [CrossRef]
- Smirnov, A.; Comte, C.; Mager-Heckel, A.M.; Addis, V.; Krasheninnikov, I.A.; Martin, R.P.; Entelis, N.; Tarassov, I. Mitochondrial enzyme rhodanese is essential for 5 S ribosomal RNA import into human mitochondria. J. Biol. Chem. 2010, 285, 30792–30803. [Google Scholar] [CrossRef] [Green Version]
- Morton, N.M.; Beltram, J.; Carter, R.N.; Michailidou, Z.; Gorjanc, G.; McFadden, C.; Barrios-Llerena, M.E.; Rodriguez-Cuenca, S.; Gibbins, M.T.; Aird, R.E.; et al. Genetic identification of thiosulfate sulfurtransferase as an adipocyte-expressed antidiabetic target in mice selected for leanness. Nat. Med. 2016, 22, 771–779. [Google Scholar] [CrossRef]
- Diaferia, G.R.; Cirulli, V.; Biunno, I. SEL1L regulates adhesion, proliferation and secretion of insulin by affecting integrin signaling. PLoS ONE 2013, 8, e79458. [Google Scholar] [CrossRef]
- Shen, W.; Scearce, L.M.; Brestelli, J.E.; Sund, N.J.; Kaestner, K.H. Foxa3 (hepatocyte nuclear factor 3gamma) is required for the regulation of hepatic GLUT2 expression and the maintenance of glucose homeostasis during a prolonged fast. J. Biol. Chem. 2001, 276, 42812–42817. [Google Scholar] [CrossRef] [Green Version]
- Chen, G.; Korfhagen, T.R.; Karp, C.L.; Impey, S.; Xu, Y.; Randell, S.H.; Kitzmiller, J.; Maeda, Y.; Haitchi, H.M.; Sridharan, A.; et al. Foxa3 induces goblet cell metaplasia and inhibits innate antiviral immunity. Am. J. Respir. Crit. Care Med. 2014, 189, 301–313. [Google Scholar] [CrossRef] [Green Version]
- Lee, C.H.; Olson, P.; Hevener, A.; Mehl, I.; Chong, L.W.; Olefsky, J.M.; Gonzalez, F.J.; Ham, J.; Kang, H.; Peters, J.M.; et al. PPARdelta regulates glucose metabolism and insulin sensitivity. Proc. Natl. Acad. Sci. USA 2006, 103, 3444–3449. [Google Scholar] [CrossRef] [Green Version]
- Mitsui, K.; Tokuzawa, Y.; Itoh, H.; Segawa, K.; Murakami, M.; Takahashi, K.; Maruyama, M.; Maeda, M.; Yamanaka, S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 2003, 113, 631–642. [Google Scholar] [CrossRef] [Green Version]
- Shi, G.; Jin, Y. Role of Oct4 in maintaining and regaining stem cell pluripotency. Stem Cell Res. Ther. 2010, 1, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keller, A.D.; Maniatis, T. Identification and characterization of a novel repressor of beta-interferon gene expression. Genes Dev. 1991, 5, 868–879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turner, C.A.; Mack, D.H.; Davis, M.M. Blimp-1, a novel zinc finger-containing protein that can drive the maturation of B lymphocytes into immunoglobulin-secreting cells. Cell 1994, 77, 297–306. [Google Scholar] [CrossRef]
- Gutierrez-Hartmann, A.; Duval, D.L.; Bradford, A.P. ETS transcription factors in endocrine systems. Trends Endocrinol. Metab. 2007, 18, 150–158. [Google Scholar] [CrossRef]
- Maroulakou, I.G.; Bowe, D.B. Expression and function of Ets transcription factors in mammalian development: A regulatory network. Oncogene 2000, 19, 6432–6442. [Google Scholar] [CrossRef] [Green Version]
- Kobberup, S.; Nyeng, P.; Juhl, K.; Hutton, J.; Jensen, J. ETS-family genes in pancreatic development. Dev. Dyn. 2007, 236, 3100–3110. [Google Scholar] [CrossRef]
- Chen, F.; Sha, M.; Wang, Y.; Wu, T.; Shan, W.; Liu, J.; Zhou, W.; Zhu, Y.; Sun, Y.; Shi, Y.; et al. Transcription factor Ets-1 links glucotoxicity to pancreatic beta cell dysfunction through inhibiting PDX-1 expression in rodent models. Diabetologia 2016, 59, 316–324. [Google Scholar] [CrossRef] [Green Version]
- Cichowski, K.; Jacks, T. NF1 tumor suppressor gene function: Narrowing the GAP. Cell 2001, 104, 593–604. [Google Scholar] [CrossRef] [Green Version]
- De Vas, M.G.; Kopp, J.L.; Heliot, C.; Sander, M.; Cereghini, S.; Haumaitre, C. Hnf1b controls pancreas morphogenesis and the generation of Ngn3+ endocrine progenitors. Development 2015, 142, 871–882. [Google Scholar] [CrossRef] [Green Version]
- Welters, H.J.; Senkel, S.; Klein-Hitpass, L.; Erdmann, S.; Thomas, H.; Harries, L.W.; Pearson, E.R.; Bingham, C.; Hattersley, A.T.; Ryffel, G.U.; et al. Conditional expression of hepatocyte nuclear factor-1beta, the maturity-onset diabetes of the young-5 gene product, influences the viability and functional competence of pancreatic beta-cells. J. Endocrinol. 2006, 190, 171–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- LaPensee, C.R.; Lin, G.; Dent, A.L.; Schwartz, J. Deficiency of the transcriptional repressor B cell lymphoma 6 (Bcl6) is accompanied by dysregulated lipid metabolism. PLoS ONE 2014, 9, e97090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Igoillo-Esteve, M.; Gurzov, E.N.; Eizirik, D.L.; Cnop, M. The transcription factor B-cell lymphoma (BCL)-6 modulates pancreatic {beta}-cell inflammatory responses. Endocrinology 2011, 152, 447–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moore, F.; Santin, I.; Nogueira, T.C.; Gurzov, E.N.; Marselli, L.; Marchetti, P.; Eizirik, D.L. The transcription factor C/EBP delta has anti-apoptotic and anti-inflammatory roles in pancreatic beta cells. PLoS ONE 2012, 7, e31062. [Google Scholar] [CrossRef] [Green Version]
- Matsuda, T.; Takahashi, H.; Mieda, Y.; Shimizu, S.; Kawamoto, T.; Matsuura, Y.; Takai, T.; Suzuki, E.; Kanno, A.; Koyanagi-Kimura, M.; et al. Regulation of Pancreatic β cell mass by cross-interaction between CCAAT enhancer binding protein β induced by endoplasmic reticulum stress and AMP-activated protein kinase activity. PLoS ONE 2015, 10, e0130757. [Google Scholar] [CrossRef]
- Lu, M.; Seufert, J.; Habener, J.F. Pancreatic beta-cell-specific repression of insulin gene transcription by CCAAT/enhancer-binding protein beta. Inhibitory interactions with basic helix-loop-helix transcription factor E47. J. Biol. Chem. 1997, 272, 28349–28359. [Google Scholar] [CrossRef] [Green Version]
- Wegner, M. From head to toes: The multiple facets of Sox proteins. Nucleic Acids Res. 1999, 27, 1409–1420. [Google Scholar] [CrossRef]
- Kormish, J.D.; Sinner, D.; Zorn, A.M. Interactions between SOX factors and Wnt/beta-catenin signaling in development and disease. Dev. Dyn. 2010, 239, 56–68. [Google Scholar] [CrossRef] [Green Version]
- She, Z.Y.; Yang, W.X. SOX family transcription factors involved in diverse cellular events during development. Eur. J. Cell Biol. 2015, 94, 547–563. [Google Scholar] [CrossRef]
- Mastracci, T.L.; Evans-Molina, C. Pancreatic and islet development and function: The role of thyroid hormone. J. Endocrinol. Diabetes Obes. 2014, 2, 1044. [Google Scholar]
- Treiber, T.; Mandel, E.M.; Pott, S.; Györy, I.; Firner, S.; Liu, E.T.; Grosschedl, R. Early B cell factor 1 regulates B cell gene networks by activation, repression, and transcription- independent poising of chromatin. Immunity 2010, 32, 714–725. [Google Scholar] [CrossRef] [Green Version]
- Garcia-Dominguez, M.; Poquet, C.; Garel, S.; Charnay, P. Ebf gene function is required for coupling neuronal differentiation and cell cycle exit. Development 2003, 130, 6013–6025. [Google Scholar] [CrossRef] [Green Version]
- Mazur, M.A.; Winkler, M.; Ganic, E.; Colberg, J.K.; Johansson, J.K.; Bennet, H.; Fex, M.; Nuber, U.A.; Artner, I. Microphthalmia transcription factor regulates pancreatic β-cell function. Diabetes 2013, 62, 2834–2842. [Google Scholar] [CrossRef] [Green Version]
- Hobert, O.; Westphal, H. Functions of LIM-homeobox genes. Trends Genet. 2000, 16, 75–83. [Google Scholar] [CrossRef]
- Bethea, M.; Liu, Y.; Wade, A.K.; Mullen, R.; Gupta, R.; Gelfanov, V.; DiMarchi, R.; Bhatnagar, S.; Behringer, R.; Habegger, K.M.; et al. The islet-expressed Lhx1 transcription factor interacts with Islet-1 and contributes to glucose homeostasis. Am. J. Physiol. Endocrinol. Metab. 2019, 316, E397–E409. [Google Scholar] [CrossRef]
- Prieto-Ruiz, J.A.; Alis, R.; García-Benlloch, S.; Sáez-Atiénzar, S.; Ventura, I.; Hernández-Andreu, J.M.; Hernández-Yago, J.; Blesa, J.R. Expression of the human TIMM23 and TIMM23B genes is regulated by the GABP transcription factor. Biochim. Biophys. Acta Gene Regul. Mech. 2018, 1861, 80–94. [Google Scholar] [CrossRef]
- Yang, Z.F.; Drumea, K.; Mott, S.; Wang, J.; Rosmarin, A.G. GABP transcription factor (nuclear respiratory factor 2) is required for mitochondrial biogenesis. Mol. Cell. Biol. 2014, 34, 3194–3201. [Google Scholar] [CrossRef] [Green Version]
- Sanvito, F.; Herrera, P.L.; Huarte, J.; Nichols, A.; Montesano, R.; Orci, L.; Vassalli, J.D. TGF-beta 1 influences the relative development of the exocrine and endocrine pancreas in vitro. Development 1994, 120, 3451–3462. [Google Scholar]
- Jiang, Y.; Fischbach, S.; Xiao, X. The Role of the TGFβ receptor signaling pathway in adult beta cell proliferation. Int. J. Mol. Sci. 2018, 19, 3136. [Google Scholar] [CrossRef] [Green Version]
- Hisa, T.; Spence, S.E.; Rachel, R.A.; Fujita, M.; Nakamura, T.; Ward, J.M.; Devor-Henneman, D.E.; Saiki, Y.; Kutsuna, H.; Tessarollo, L.; et al. Hematopoietic, angiogenic and eye defects in Meis1 mutant animals. EMBO J. 2004, 23, 450–459. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Rowan, S.; Yue, Y.; Heaney, S.; Pan, Y.; Brendolan, A.; Selleri, L.; Maas, R.L. Pax6 is regulated by Meis and Pbx homeoproteins during pancreatic development. Dev. Biol. 2006, 300, 748–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalkhoff, R.K. Metabolic effects of progesterone. Am. J. Obstet. Gynecol. 1982, 142, 735–738. [Google Scholar] [CrossRef]
- Zhou, R.; Yao, X.; Xu, X.; Wang, G.; Zhu, Z.; Chen, J.; Chen, L.; Shen, X. Blockage of progesterone receptor effectively protects pancreatic islet beta cell viability. Steroids 2013, 78, 987–995. [Google Scholar] [CrossRef] [PubMed]
- Picard, F.; Wanatabe, M.; Schoonjans, K.; Lydon, J.; O’Malley, B.W.; Auwerx, J. Progesterone receptor knockout mice have an improved glucose homeostasis secondary to beta -cell proliferation. Proc. Natl. Acad. Sci. USA 2002, 99, 15644–15648. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Q.; Choi, G.; Anderson, D.J. The bHLH transcription factor Olig2 promotes oligodendrocyte differentiation in collaboration with Nkx2.2. Neuron 2001, 31, 791–807. [Google Scholar] [CrossRef] [Green Version]
- Kuhn, S.; Gritti, L.; Crooks, D.; Dombrowski, Y. Oligodendrocytes in development, myelin generation and beyond. Cells 2019, 8, 1424. [Google Scholar] [CrossRef] [Green Version]
- Buffo, A.; Vosko, M.R.; Ertürk, D.; Hamann, G.F.; Jucker, M.; Rowitch, D.; Götz, M. Expression pattern of the transcription factor Olig2 in response to brain injuries: Implications for neuronal repair. Proc. Natl. Acad. Sci. USA 2005, 102, 18183–18188. [Google Scholar] [CrossRef] [Green Version]
- Vastenhouw, N.L.; Schier, A.F. Bivalent histone modifications in early embryogenesis. Curr. Opin. Cell Biol. 2012, 24, 374–386. [Google Scholar] [CrossRef] [Green Version]
- Bernstein, B.E.; Mikkelsen, T.S.; Xie, X.; Kamal, M.; Huebert, D.J.; Cuff, J.; Fry, B.; Meissner, A.; Wernig, M.; Plath, K.; et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 2006, 125, 315–326. [Google Scholar] [CrossRef] [Green Version]
- Voigt, P.; Tee, W.W.; Reinberg, D. A double take on bivalent promoters. Genes Dev. 2013, 27, 1318–1338. [Google Scholar] [CrossRef] [Green Version]
- Maupetit-Méhouas, S.; Montibus, B.; Nury, D.; Tayama, C.; Wassef, M.; Kota, S.K.; Fogli, A.; Cerqueira Campos, F.; Hata, K.; Feil, R.; et al. Imprinting control regions (ICRs) are marked by mono-allelic bivalent chromatin when transcriptionally inactive. Nucleic Acids Res. 2016, 44, 621–635. [Google Scholar] [CrossRef] [Green Version]
- Bernhart, S.H.; Kretzmer, H.; Holdt, L.M.; Jühling, F.; Ammerpohl, O.; Bergmann, A.K.; Northoff, B.H.; Doose, G.; Siebert, R.; Stadler, P.F.; et al. Changes of bivalent chromatin coincide with increased expression of developmental genes in cancer. Sci. Rep. 2016, 6, 37393. [Google Scholar] [CrossRef] [PubMed]
- Zsindely, N.; Bodai, L. Histone methylation in Huntington’s disease: Are bivalent promoters the critical targets? Neural Regen. Res. 2018, 13, 1191–1192. [Google Scholar] [CrossRef] [PubMed]
- Lu, T.T.; Heyne, S.; Dror, E.; Casas, E.; Leonhardt, L.; Boenke, T.; Yang, C.H.; Sagar Arrigoni, L.; Dalgaard, K. The Polycomb-Dependent Epigenome Controls β Cell Dysfunction, Dedifferentiation, and Diabetes. Cell Metab. 2018, 27, 1294–1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Persson, K.; Pacini, G.; Sundler, F.; Ahrén, B. Islet function phenotype in gastrin-releasing peptide receptor gene-deficient mice. Endocrinology 2002, 143, 3717–3726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Persson, K.; Gingerich, R.L.; Nayak, S.; Wada, K.; Wada, E.; Ahrén, B. Reduced GLP-1 and insulin responses and glucose intolerance after gastric glucose in GRP receptor-deleted mice. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E956–E962. [Google Scholar] [CrossRef]
- Seidah, N.G.; Mayer, G.; Zaid, A.; Rousselet, E.; Nassoury, N.; Poirier, S.; Essalmani, R.; Prat, A. The activation and physiological functions of the proprotein convertases. Int. J. Biochem. Cell Biol. 2008, 40, 1111–1125. [Google Scholar] [CrossRef]
- Seidah, N.G.; Awan, Z.; Chrétien, M.; Mbikay, M. PCSK9: A key modulator of cardiovascular health. Circ. Res. 2014, 114, 1022–1036. [Google Scholar] [CrossRef]
- Da Dalt, L.; Ruscica, M.; Bonacina, F.; Balzarotti, G.; Dhyani, A.; Di Cairano, E.; Baragetti, A.; Arnaboldi, L.; De Metrio, S.; Pellegatta, F.; et al. PCSK9 deficiency reduces insulin secretion and promotes glucose intolerance: The role of the low-density lipoprotein receptor. Eur. Heart J. 2019, 40, 357–368. [Google Scholar] [CrossRef]
- Maravillas-Montero, J.L.; Santos-Argumedo, L. The myosin family: Unconventional roles of actin-dependent molecular motors in immune cells. J. Leukoc. Biol. 2012, 91, 35–46. [Google Scholar] [CrossRef] [Green Version]
- Shestenko, O.P.; Nikonov, S.D.; Mertvetsov, N.P. Angiogenin and its role in angiogenesis. Mol. Biol. Mosk. 2001, 35, 349–371. [Google Scholar] [CrossRef] [PubMed]
- Ambrosini, A.; Bresciani, L.; Fracchia, S.; Brunello, N.; Racagni, G. Metabotropic glutamate receptors negatively coupled to adenylate cyclase inhibit N-methyl-D-aspartate receptor activity and prevent neurotoxicity in mesencephalic neurons in vitro. Mol. Pharmacol. 1995, 47, 1057–1064. [Google Scholar] [PubMed]
- Otter, S.; Lammert, E. Exciting times for pancreatic islets: Glutamate signaling in endocrine cells. Trends Endocrinol. Metab. 2016, 27, 177–188. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Hagedorn, C.H.; Wang, L. Role of nuclear receptor SHP in metabolism and cancer. Biochim. Biophys. Acta 2011, 1812, 893–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suh, Y.H.; Kim, S.Y.; Lee, H.Y.; Jang, B.C.; Bae, J.H.; Sohn, J.N.; Suh, S.I.; Park, J.W.; Lee, K.U.; Song, D.K. Overexpression of short heterodimer partner recovers impaired glucose-stimulated insulin secretion of pancreatic beta-cells overexpressing UCP2. J. Endocrinol. 2004, 183, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Zhang, P.; Wang, C.; Han, C.; Meng, J.; Liu, X.; Xu, S.; Li, N.; Wang, Q.; Shi, X.; et al. Immune responsive gene 1 (IRG1) promotes endotoxin tolerance by increasing A20 expression in macrophages through reactive oxygen species. J. Biol. Chem. 2013, 288, 16225–16234. [Google Scholar] [CrossRef] [Green Version]
- Degrandi, D.; Hoffmann, R.; Beuter-Gunia, C.; Pfeffer, K. The proinflammatory cytokine-induced IRG1 protein associates with mitochondria. J. Interferon Cytokine Res. 2009, 29, 55–67. [Google Scholar] [CrossRef]
- Lampropoulou, V.; Sergushichev, A.; Bambouskova, M.; Nair, S.; Vincent, E.E.; Loginicheva, E.; Cervantes-Barragan, L.; Ma, X.; Huang, S.C.; Griss, T.; et al. Itaconate Links Inhibition of Succinate Dehydrogenase with Macrophage Metabolic Remodeling and Regulation of Inflammation. Cell Metab. 2016, 24, 158–166. [Google Scholar] [CrossRef]
- Kharitonenkov, A.; Shiyanova, T.L.; Koester, A.; Ford, A.M.; Micanovic, R.; Galbreath, E.J.; Sandusky, G.E.; Hammond, L.J.; Moyers, J.S.; Owens, R.A.; et al. FGF-21 as a novel metabolic regulator. J. Clin. Investig. 2005, 115, 1627–1635. [Google Scholar] [CrossRef] [Green Version]
- Ge, X.; Chen, C.; Hui, X.; Wang, Y.; Lam, K.S.; Xu, A. Fibroblast growth factor 21 induces glucose transporter-1 expression through activation of the serum response factor/Ets-like protein-1 in adipocytes. J. Biol. Chem. 2011, 286, 34533–34541. [Google Scholar] [CrossRef] [Green Version]
- Singhal, G.; Fisher, F.M.; Chee, M.J.; Tan, T.G.; El Ouaamari, A.; Adams, A.C.; Najarian, R.; Kulkarni, R.N.; Benoist, C.; Flier, J.S.; et al. Fibroblast Growth Factor 21 (FGF21) Protects against High Fat Diet Induced Inflammation and Islet Hyperplasia in Pancreas. PLoS ONE 2016, 11, e0148252. [Google Scholar] [CrossRef] [PubMed]
- Pan, Y.; Wang, B.; Zheng, J.; Xiong, R.; Fan, Z.; Ye, Y.; Zhang, S.; Li, Q.; Gong, F.; Wu, C.; et al. Pancreatic fibroblast growth factor 21 protects against type 2 diabetes in mice by promoting insulin expression and secretion in a PI3K/Akt signaling-dependent manner. J. Cell. Mol. Med. 2019, 23, 1059–1071. [Google Scholar] [CrossRef] [PubMed]
- So, W.Y.; Cheng, Q.; Xu, A.; Lam, K.S.; Leung, P.S. Loss of fibroblast growth factor 21 action induces insulin resistance, pancreatic islet hyperplasia and dysfunction in mice. Cell Death Dis. 2015, 6, e1707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heit, C.; Jackson, B.C.; McAndrews, M.; Wright, M.W.; Thompson, D.C.; Silverman, G.A.; Nebert, D.W.; Vasiliou, V. Update of the human and mouse SERPIN gene superfamily. Hum. Genom. 2013, 7, 22. [Google Scholar] [CrossRef] [Green Version]
- Wendeler, M.W.; Praus, M.; Jung, R.; Hecking, M.; Metzig, C.; Gessner, R. Ksp-cadherin is a functional cell-cell adhesion molecule related to LI-cadherin. Exp. Cell Res. 2004, 294, 345–355. [Google Scholar] [CrossRef] [PubMed]
- Ng, A.C.; Eisenberg, J.M.; Heath, R.J.; Huett, A.; Robinson, C.M.; Nau, G.J.; Xavier, R.J. Human leucine-rich repeat proteins: A genome-wide bioinformatic categorization and functional analysis in innate immunity. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. 1), 4631–4638. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Aulia, S.; Li, L.; Tang, B.L. AMIGO and friends: An emerging family of brain-enriched, neuronal growth modulating, type I transmembrane proteins with leucine-rich repeats (LRR) and cell adhesion molecule motifs. Brain Res. Rev. 2006, 51, 265–274. [Google Scholar] [CrossRef]
- Sandovici, I.; Smith, N.H.; Nitert, M.D.; Ackers-Johnson, M.; Uribe-Lewis, S.; Ito, Y.; Jones, R.H.; Marquez, V.E.; Cairns, W.; Tadayyon, M.; et al. Maternal diet and aging alter the epigenetic control of a promoter-enhancer interaction at the Hnf4a gene in rat pancreatic islets. Proc. Natl. Acad. Sci. USA 2011, 108, 5449–5454. [Google Scholar] [CrossRef] [Green Version]
- Quilter, C.R.; Cooper, W.N.; Cliffe, K.M.; Skinner, B.M.; Prentice, P.M.; Nelson, L.; Bauer, J.; Ong, K.K.; Constância, M.; Lowe, W.L.; et al. Impact on offspring methylation patterns of maternal gestational diabetes mellitus and intrauterine growth restraint suggest common genes and pathways linked to subsequent type 2 diabetes risk. FASEB J. 2014, 28, 4868–4879. [Google Scholar] [CrossRef] [Green Version]
- Zeng, Y.; Gu, P.; Liu, K.; Huang, P. Maternal protein restriction in rats leads to reduced PGC-1a expression via altered DNA methylation in skeletal muscle. Mol. Med. Rep. 2013, 7, 306–312. [Google Scholar] [CrossRef] [Green Version]
- Raychaudhuri, N.; Raychaudhuri, S.; Thamotharan, M.; Devaskar, S.U. Histone code modifications repress glucose transporter 4 expression in the intrauterine growth-restricted offspring. J. Biol. Chem. 2008, 283, 13611–13626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fung, C.M.; Yang, Y.; Fu, Q.; Brown, A.S.; Yu, B.; Callaway, C.W.; Li, J.; Lane, R.H.; McKnight, R.A. IUGR prevents IGF-1 upregulation in juvenile male mice by perturbing postnatal IGF-1 chromatin remodeling. Pediatr. Res. 2015, 78, 14–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burdge, G.C.; Slater-Jefferies, J.; Torrens, C.; Phillips, E.S.; Hanson, M.A.; Lillycrop, K.A. Dietary protein restriction of pregnant rats in the F0 generation induces altered methylation of hepatic gene promoters in the adult male offspring in the F1 and F2 generations. Br. J. Nutr. 2007, 97, 435–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martínez, D.; Pentinat, T.; Ribó, S.; Daviaud, C.; Bloks, V.W.; Cebrià, J.; Villalmanzo, N.; Kalko, S.G.; Ramón-Krauel, M.; Díaz, R.; et al. In utero undernutrition in male mice programs liver lipid metabolism in the second-generation offspring involving altered Lxra DNA methylation. Cell Metab. 2014, 19, 941–951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hardikar, A.A.; Satoor, S.N.; Karandikar, M.S.; Joglekar, M.V.; Puranik, A.S.; Wong, W.; Kumar, S.; Limaye, A.; Bhat, D.S.; Januszewski, A.S.; et al. Multigenerational Undernutrition Increases Susceptibility to Obesity and Diabetes that Is Not Reversed after Dietary Recuperation. Cell Metab. 2015, 22, 312–319. [Google Scholar] [CrossRef] [Green Version]
- Paauw, N.D.; Lely, A.T.; Joles, J.A.; Franx, A.; Nikkels, P.G.; Mokry, M.; van Rijn, B.B. H3K27 acetylation and gene expression analysis reveals differences in placental chromatin activity in fetal growth restriction. Clin. Epigenet. 2018, 10, 85. [Google Scholar] [CrossRef]
- Simmons, R.A. Developmental origins of beta-cell failure in type 2 diabetes: The role of epigenetic mechanisms. Pediatr Res. 2007, 61, 64–67. [Google Scholar] [CrossRef] [Green Version]
- Akbari, M.; Hassan-Zadeh, V. The inflammatory effect of epigenetic factors and modifications in type 2 diabetes. Inflammopharmacology 2019. [Google Scholar] [CrossRef]
- Eliasson, L.; Regazzi, R. Micro(RNA) management and mismanagement of the islet. J. Mol. Biol. 2019. [Google Scholar] [CrossRef]
- De Jesus, D.F.; Kulkarni, R.N. “Omics” and “epi-omics” underlying the β-cell adaptation to insulin resistance. Mol. Metab. 2019, 27, 42–48. [Google Scholar] [CrossRef]
- Young, M.D.; Willson, T.A.; Wakefield, M.J.; Trounson, E.; Hilton, D.J.; Blewitt, M.E.; Oshlack, A.; Majewski, I.J. ChIP-seq analysis reveals distinct H3K27me3 profiles that correlate with transcriptional activity. Nucleic Acids Res. 2011, 39, 7415–7427. [Google Scholar] [CrossRef] [PubMed]
- Salomon, D.; Meda, P. Heterogeneity and contact-dependent regulation of hormone secretion by individual B cells. Exp. Cell Res. 1986, 162, 507–520. [Google Scholar] [CrossRef]
- Santos-Silva, J.C.; Carvalho, C.P.; de Oliveira, R.B.; Boschero, A.C.; Collares-Buzato, C.B. Cell-to-cell contact dependence and junctional protein content are correlated with in vivo maturation of pancreatic beta cells. Can. J. Physiol. Pharmacol. 2012, 90, 837–850. [Google Scholar] [CrossRef] [PubMed]
- Collares-Buzato, C.B.; Carvalho, C.P.; Furtado, A.G.; Boschero, A.C. Upregulation of the expression of tight and adherens junction-associated proteins during maturation of neonatal pancreatic islets in vitro. J. Mol. Histol. 2004, 35, 811–822. [Google Scholar] [CrossRef]
- Meda, P. Protein-mediated interactions of pancreatic islet cells. Scientifica 2013, 2013, 621249. [Google Scholar] [CrossRef]
- Elghazi, L.; Gould, A.P.; Weiss, A.J.; Barker, D.J.; Callaghan, J.; Opland, D.; Myers, M.; Cras-Méneur, C.; Bernal-Mizrachi, E. Importance of β-Catenin in glucose and energy homeostasis. Sci. Rep. 2012, 2, 693. [Google Scholar] [CrossRef] [Green Version]
- Calderon, B.; Carrero, J.A.; Ferris, S.T.; Sojka, D.K.; Moore, L.; Epelman, S.; Murphy, K.M.; Yokoyama, W.M.; Randolph, G.J.; Unanue, E.R. The pancreas anatomy conditions the origin and properties of resident macrophages. J. Exp. Med. 2015, 212, 1497–1512. [Google Scholar] [CrossRef] [Green Version]
- Geutskens, S.B.; Otonkoski, T.; Pulkkinen, M.A.; Drexhage, H.A.; Leenen, P.J. Macrophages in the murine pancreas and their involvement in fetal endocrine development in vitro. J. Leukoc. Biol. 2005, 78, 845–852. [Google Scholar] [CrossRef]
- Radenkovic, M.; Uvebrant, K.; Skog, O.; Sarmiento, L.; Avartsson, J.; Storm, P.; Vickman, P.; Bertilsson, P.A.; Fex, M.; Korgsgren, O.; et al. Characterization of resident lymphocytes in human pancreatic islets. Clin. Exp. Immunol. 2017, 187, 418–427. [Google Scholar] [CrossRef] [Green Version]
- Georges, A.; Auguste, A.; Bessière, L.; Vanet, A.; Todeschini, A.L.; Veitia, R.A. FOXL2: A central transcription factor of the ovary. J. Mol. Endocrinol. 2014, 52, R17–R33. [Google Scholar] [CrossRef]
- Stocco, D.M. StAR protein and the regulation of steroid hormone biosynthesis. Annu. Rev. Physiol. 2001, 63, 193–213. [Google Scholar] [CrossRef]
- Li, J.; Cao, Y.; Wu, Y.; Chen, W.; Yuan, Y.; Ma, X.; Huang, G. The expression profile analysis of NKX2-5 knock-out embryonic mice to explore the pathogenesis of congenital heart disease. J. Cardiol. 2015, 66, 527–531. [Google Scholar] [CrossRef] [Green Version]
- Konishi, M.; Mikami, T.; Yamasaki, M.; Miyake, A.; Itoh, N. Fibroblast growth factor-16 is a growth factor for embryonic brown adipocytes. J. Biol. Chem. 2000, 275, 12119–12122. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Jin, Y.; Cattini, P.A. Expression of the Cardiac Maintenance and Survival Factor FGF-16 Gene Is Regulated by Csx/Nkx2.5 and Is an Early Target of Doxorubicin Cardiotoxicity. DNA Cell Biol. 2017, 36, 117–126. [Google Scholar] [CrossRef]
- Basch, M.L.; Bronner-Fraser, M.; García-Castro, M.I. Specification of the neural crest occurs during gastrulation and requires Pax7. Nature 2006, 441, 218–222. [Google Scholar] [CrossRef]
- Von Maltzahn, J.; Jones, A.E.; Parks, R.J.; Rudnicki, M.A. Pax7 is critical for the normal function of satellite cells in adult skeletal muscle. Proc. Natl. Acad. Sci. USA 2013, 110, 16474–16479. [Google Scholar] [CrossRef] [Green Version]
2-wk | 10-wk | |||||
---|---|---|---|---|---|---|
Histone Mark | Total Gene # | Up-Regulated Gene # | Down-Regulated Gene # | Total Gene # | Up-Regulated Gene # | Down-Regulated Gene # |
H3K4me3 | 41 | 29 | 12 | 638 | 344 | 294 |
H3K27me3 | 61 | 23 | 38 | 357 | 24 | 333 |
H3K27Ac | 148 | 52 | 96 | 401 | 29 | 372 |
All 3 marks | 0 | 0 | 0 | 6 | 3 | 3 |
Gene | Gene Name | H3K4me3 logFC | H3K27me3 logFC | H3K27Ac logFC | RNASeq logFC | RNASeq FDR |
---|---|---|---|---|---|---|
Trpm5 | transient receptor potential cation channel, subfamily M, member 5 | 1.27 | −0.78 | 0.96 | 0.8302 | 0.0215 |
Tfam | transcription factor A, mitochondrial | 0.90 | −0.95 | 1.10 | 0.5014 | 0.0148 |
Mcf2l | MCF.2 cell line derived transforming sequence-like | 1.17 | −1.06 | 1.19 | 0.4669 | 0.0500 |
Slc28a2 | solute carrier family 28 member 2 | −1.05 | 1.77 | −0.75 | −1.2177 | 0.0040 |
Tnf | tumor necrosis factor | −1.47 | 1.01 | −0.95 | −2.1019 | 0.0020 |
Mpz | myelin protein zero | −0.55 | 0.42 | −0.56 | −2.4936 | 0.0027 |
2-wk Islets | 10-wk Islets | |||||
---|---|---|---|---|---|---|
Genes | RNAseq logFC | RNAseq FDR | H3K27Ac logFC | RNAseq logFC | RNAseq FDR | H3K27Ac logFC |
Bcat2 | −1.08 | 1.30 × 10−2 | −0.98 | −1.37 | 1.74 × 10−4 | −0.88 |
Bhlha15 | −2.12 | 8.77 × 10−5 | −0.66 | −1.42 | 5.75 × 10−3 | −0.48 |
Cyp2t1 | −0.99 | 1.14 × 10−2 | −0.63 | −0.78 | 3.54 × 10−2 | −0.56 |
Dram1 | −1.42 | 1.41 × 10−3 | −0.12 | −1.92 | 5.29 × 10−7 | −0.76 |
Foxa3 | −0.98 | 3.16 × 10−2 | −0.67 | −0.92 | 2.37 × 10−2 | −0.47 |
Gpt2 | −1.39 | 2.83 × 10−4 | −0.59 | −0.91 | 1.67 × 10−2 | −0.77 |
Mast3 | −1.10 | 2.69 × 10−4 | −0.56 | −0.83 | 4.53 × 10−3 | −0.77 |
Ppard | −1.36 | 5.15 × 10−5 | −0.06 | −1.97 | 7.32 × 10−11 | −0.74 |
Pycr1 | −1.85 | 1.82 × 10−4 | −0.87 | −1.18 | 2.05 × 10−2 | −0.82 |
Reep5 | −0.92 | 4.49 × 10−2 | −0.41 | −1.48 | 2.62 × 10−5 | −0.47 |
Tex49 | −2.10 | 3.54 × 10−3 | −0.33 | −1.93 | 2.48 × 10−2 | −1.35 |
Rnh1 | −1.15 | 1.39 × 10−2 | −0.28 | −1.97 | 1.32 × 10−7 | −0.56 |
Rnpepl1 | −1.03 | 1.17 × 10−3 | −0.64 | −1.37 | 9.37 × 10−7 | −0.48 |
Sel1l | −1.61 | 3.84 × 10−3 | −0.38 | −1.86 | 9.29 × 10−5 | −0.95 |
Sfxn2 | −1.42 | 2.34 × 10−3 | −0.51 | −0.97 | 3.91 × 10−2 | −0.90 |
Tnip2 | −0.61 | 3.89 × 10−2 | −0.55 | −0.59 | 2.96 × 10−2 | −0.59 |
Tst | −1.26 | 1.41 × 10−2 | −0.14 | −3.13 | 9.28 × 10−14 | −0.54 |
Ulk1 | −1.03 | 2.87 × 10−5 | −0.41 | −0.70 | 4.26 × 10−3 | −0.66 |
Wdtc1 | −0.92 | 2.98 × 10−3 | −1.01 | −0.90 | 1.26 × 10−3 | −0.51 |
Xbp1 | −1.14 | 1.64 × 10−2 | −0.56 | −1.16 | 4.90 × 10−3 | −0.84 |
Histone Mark | Ingenuity Canonical Pathways | p-Value |
---|---|---|
H3K4me3 | Type II Diabetes Mellitus signaling | 1.32 × 10−4 |
antiproliferative role of TOB in T- cell signaling | 7.24 × 10−4 | |
Tight junction signaling | 1.00 × 10−3 | |
RAR activation | 1.12 × 10−3 | |
PI3K signaling in B lymphocytes | 1.23 × 10−3 | |
Factors promoting cardiogenesis in vertebrates | 1.23 × 10−3 | |
BMP signaling pathway | 1.41 × 10−3 | |
Neurotrophin/TRK signaling | 1.51 × 10−3 | |
PPAR signaling | 1.55 × 10−3 | |
H3K27me3 | Cellular stress and injury | 2.24 × 10−7 |
Hepatic fibrosis / hepatic stellate cell activation | 1.41 × 10−6 | |
cAMP-mediated signaling | 1.32 × 10−5 | |
Macrophage function | 2.95 × 10−5 | |
Mitochondrial function / Autophagy | 3.55 × 10−5 | |
Citrulline-nitric oxide cycle | 3.63 × 10−5 | |
Wnt/β-catenin signaling | 8.13 × 10−5 | |
Axonal guidance signaling | 9.12 × 10−5 | |
eNOS signaling | 3.63 × 10−4 | |
H3K27Ac | Polyamine regulation | 1.78 × 10−6 |
Cell migration signaling | 1.02 × 10−5 | |
Production of nitric oxide and reactive oxygen species in Macrophages | 1.05 × 10−5 | |
IL-9 signaling | 1.35 × 10−5 | |
PDGF signaling | 3.24 × 10−5 | |
PPAR signaling | 4.90 × 10−5 | |
Unfolded protein response | 5.13 × 10−5 | |
ERK/MAPK signaling | 6.46 × 10−5 | |
Phospholipase C signaling | 1.12 × 10−4 |
H3K4me3 | |||||
---|---|---|---|---|---|
Motif Name | Consensus | 2w_IUGR p-Value | 2w_Ctrl p-Value | 10w_IUGR p-Value | 10w_Ctrl p-Value |
ELF3 | ANCAGGAAGT | 1.00 × 10−43 | 0.1 | 1.00 × 10−9 | 1 |
ELF5 | ACVAGGAAGT | 1.00 × 10−34 | 0.1 | 1.00 × 10−7 | 1 |
ERG | ACAGGAAGTG | 1.00 × 10−71 | 0.1 | 1.00 × 10−8 | 1 |
ETS1 | ACAGGAAGTG | 1.00 × 10−48 | 1 | 1.00 × 10−16 | 0.1 |
ETV1 | AACCGGAAGT | 1.00 × 10−80 | 0.1 | 1.00 × 10−11 | 1 |
ETV2 | NNAYTTCCTGHN | 1.00 × 10−62 | 1 | 1.00 × 10−9 | 0.1 |
EWS:ERG | ATTTCCTGTN | 1.00 × 10−69 | 0.1 | 1.00 × 10−8 | 1 |
GABPA | RACCGGAAGT | 1.00 × 10−58 | 0.1 | 1.00 × 10−8 | 0.1 |
SMAD4 | VBSYGTCTGG | 1.00 × 10−24 | 1 | 1.00 × 10−6 | 1 |
H3K27me3 | |||||
Motif name | Consensus | 2w_IUGR p-Value | 2w_Ctrl p-Value | 10w_IUGR p-Value | 10w_Ctrl p-Value |
MEIS1 | VGCTGWCAVB | 1 | 1.00 × 10−4 | 1 | 1.00 × 10−17 |
PR | VAGRACAKNCTGTBC | 0.1 | 1.00 × 10−11 | 1 | 1.00 × 10−20 |
OLIG2 | RCCATMTGTT | 1 | 1.00 × 10−5 | 0.1 | 1.00 × 10−10 |
2-wk | |||
---|---|---|---|
Upstream Regulator | Activation z-Score | p-Value | Target Gene Number |
FOXL2 | 2.06 | 7.02 × 10−3 | 7 |
ELK1 | 0.74 | 1.77 × 10−3 | 7 |
ETS1 | 0.36 | 1.65 × 10−2 | 14 |
NKX2-5 | −0.39 | 4.13 × 10−2 | 4 |
BCL6 | −1.24 | 5.37 × 10−3 | 12 |
CEBPA | 1.89 | 1.87 × 10−7 | 37 |
CEBPB | 2.73 | 1.94 × 10−7 | 36 |
PAX7 | 1.14 | 8.16 × 10−4 | 9 |
SOX4 | 1.89 | 5.68 × 10−4 | 16 |
HNF1B | −1.63 | 5.26 × 10−5 | 14 |
10-wk | |||
Upstream Regulator | Activation z-Score | p-Value | Target Gene Number |
FLI1 | 0.45 | 5.42 × 10−4 | 14 |
CDX2 | −0.84 | 1.29 × 10−4 | 26 |
SOX10 | −0.84 | 1.80 × 10−6 | 13 |
EBF1 | −1.23 | 1.09 × 10−5 | 30 |
SOX9 | −0.43 | 3.72 × 10−2 | 11 |
MITF | −3.25 | 8.96 × 10−4 | 39 |
NANOG | −0.24 | 2.62 × 10−3 | 22 |
TCF | −2.00 | 9.48 × 10−8 | 27 |
THRB | 0.42 | 2.38 × 10−8 | 47 |
ELF3 | −2.07 | 5.52 × 10−3 | 8 |
ERG | −4.45 | 1.40 × 10−10 | 50 |
Gene | Gene Name | Bivalency | |||
---|---|---|---|---|---|
2-wk | 10-wk | ||||
Control | IUGR | Control | IUGR | ||
Acod1 | aconitate decarboxylase 1 | + | + | + | |
Fgf21 | fibroblast growth factor 21 | + | + | + | |
Serpina11 | serpin family A member 11 | + | + | + | |
Cdh16 | cadherin 16 | + | |||
Lrrc27 | leucine-rich repeat-containing 27 | + | |||
Lrrc66 | leucine-rich repeat-containing 66 | + |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lien, Y.-C.; Wang, P.Z.; Lu, X.M.; Simmons, R.A. Altered Transcription Factor Binding and Gene Bivalency in Islets of Intrauterine Growth Retarded Rats. Cells 2020, 9, 1435. https://doi.org/10.3390/cells9061435
Lien Y-C, Wang PZ, Lu XM, Simmons RA. Altered Transcription Factor Binding and Gene Bivalency in Islets of Intrauterine Growth Retarded Rats. Cells. 2020; 9(6):1435. https://doi.org/10.3390/cells9061435
Chicago/Turabian StyleLien, Yu-Chin, Paul Zhiping Wang, Xueqing Maggie Lu, and Rebecca A. Simmons. 2020. "Altered Transcription Factor Binding and Gene Bivalency in Islets of Intrauterine Growth Retarded Rats" Cells 9, no. 6: 1435. https://doi.org/10.3390/cells9061435