#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Partially Redundant Enhancers Cooperatively Maintain Mammalian Expression Above a Critical Functional Threshold


The stability of animal form and function in the face of genetic and environmental variation relies on consistent gene expression. Multiple enhancers, each specifying a unique regulatory domain, control the precise spatiotemporal expression of many genes. However, in some genes apparently redundant enhancers regulate expression in overlapping cell-specific patterns. Although this arrangement has been shown to be important for developmental robustness in invertebrates, the role of apparently redundant enhancers in vertebrate species and in genes functioning in adulthood is poorly understood. Here, we show that expression of the mammalian Pomc gene is controlled in a tissue-specific manner by two such apparently redundant enhancers. We used targeted deletion of the individual enhancers to delineate their respective contributions to Pomc expression in the brain. Since Pomc expression from its intact locus exceeds the sum of the individual enhancer contributions to Pomc mRNA levels in embryonic mice, we infer a synergistic action between the enhancers during development. In contrast, the interaction between the enhancers is additive in adult mice. Deletion of both enhancers simultaneously almost completely abolished Pomc expression and the mutant mice displayed extreme obesity and metabolic dysfunction, while deletion of the individual enhancers had a modest or no phenotypic effect. Together, our results demonstrate that the two enhancers cooperatively maintain Pomc expression above a critical functional threshold.


Vyšlo v časopise: Partially Redundant Enhancers Cooperatively Maintain Mammalian Expression Above a Critical Functional Threshold. PLoS Genet 11(2): e32767. doi:10.1371/journal.pgen.1004935
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004935

Souhrn

The stability of animal form and function in the face of genetic and environmental variation relies on consistent gene expression. Multiple enhancers, each specifying a unique regulatory domain, control the precise spatiotemporal expression of many genes. However, in some genes apparently redundant enhancers regulate expression in overlapping cell-specific patterns. Although this arrangement has been shown to be important for developmental robustness in invertebrates, the role of apparently redundant enhancers in vertebrate species and in genes functioning in adulthood is poorly understood. Here, we show that expression of the mammalian Pomc gene is controlled in a tissue-specific manner by two such apparently redundant enhancers. We used targeted deletion of the individual enhancers to delineate their respective contributions to Pomc expression in the brain. Since Pomc expression from its intact locus exceeds the sum of the individual enhancer contributions to Pomc mRNA levels in embryonic mice, we infer a synergistic action between the enhancers during development. In contrast, the interaction between the enhancers is additive in adult mice. Deletion of both enhancers simultaneously almost completely abolished Pomc expression and the mutant mice displayed extreme obesity and metabolic dysfunction, while deletion of the individual enhancers had a modest or no phenotypic effect. Together, our results demonstrate that the two enhancers cooperatively maintain Pomc expression above a critical functional threshold.


Zdroje

1. Levine M (2010) Transcriptional enhancers in animal development and evolution. Curr Biol 20: R754–63. 20833320

2. Rubinstein M, de Souza FSJ (2013) Evolution of transcriptional enhancers and animal diversity. Philos Trans R Soc Lond B Biol Sci 368: 20130017. 24218630

3. Shen Y, Yue F, McCleary DF, Ye Z, Edsall L, et al. (2012) A map of the cis-regulatory sequences in the mouse genome. Nature 488: 116–120. 22763441

4. Thurman RE, Rynes E, Humbert R, Vierstra J, Maurano MT, et al. (2012) The accessible chromatin landscape of the human genome. Nature 489: 75–82. 22955617

5. Sanyal A, Lajoie BR, Jain G, Dekker J (2012) The long-range interaction landscape of gene promoters. Nature 489: 109–113. 22955621

6. Andersson R, Gebhard C, Miguel-Escalada I, Hoof I, Bornholdt J, et al. (2014) An atlas of active enhancers across human cell types and tissues. Nature 507: 455–461. doi: 10.1038/nature12787 24670763

7. Goto T, Macdonald P, Maniatis T (1989) Early and late periodic patterns of even skipped expression are controlled by distinct regulatory elements that respond to different spatial cues. Cell 57: 413–422. 2720776

8. Yuh CH, Davidson EH (1996) Modular cis-regulatory organization of Endo16, a gut-specific gene of the sea urchin embryo. Development 122: 1069–1082. 8620834

9. Jeong Y, El-Jaick K, Roessler E, Muenke M, Epstein DJ (2006) A functional screen for sonic hedgehog regulatory elements across a 1 Mb interval identifies long-range ventral forebrain enhancers. Development 133: 761–772. 16407397

10. Werner T, Hammer A, Wahlbuhl M, Bösl MR, Wegner M (2007) Multiple conserved regulatory elements with overlapping functions determine Sox10 expression in mouse embryogenesis. Nucleic Acids Res 35: 6526–6538. 17897962

11. Wilkins AS (1997) Canalization: a molecular genetic perspective. Bioessays 19: 257–262. 9080776

12. Diss G, Ascencio D, Deluna A, Landry CR (2013) Molecular mechanisms of paralogous compensation and the robustness of cellular networks. J Exp Zool B Mol Dev Evol.

13. Nowak MA, Boerlijst MC, Cooke J, Smith JM (1997) Evolution of genetic redundancy. Nature 388: 167–171.

14. Barbaric I, Miller G, Dear TN (2007) Appearances can be deceiving: phenotypes of knockout mice. Brief Funct Genomic Proteomic 6: 91–103. 17584761

15. Brookfield JF (1997) Genetic redundancy. Adv Genet 36: 137–155.

16. Zhang J (2012) Genetic redundancies and their evolutionary maintenance. Adv Exp Med Biol 751: 279–300. 22821463

17. Hong J-W, Hendrix DA, Levine MS (2008) Shadow enhancers as a source of evolutionary novelty. Science 321: 1314. doi: 10.1126/science.1160631 18772429

18. Hobert O (2010) Gene regulation: enhancers stepping out of the shadow. Curr Biol 20: R697–9. doi: 10.1016/j.cub.2010.07.035 20833307

19. Barolo S (2012) Shadow enhancers: frequently asked questions about distributed cis-regulatory information and enhancer redundancy. Bioessays 34: 135–141. doi: 10.1002/bies.201100121 22083793

20. Frankel N, Davis GK, Vargas D, Wang S, Payre F, et al. (2010) Phenotypic robustness conferred by apparently redundant transcriptional enhancers. Nature 466: 490–493. doi: 10.1038/nature09158 20512118

21. Perry MW, Boettiger AN, Bothma JP, Levine M (2010) Shadow enhancers foster robustness of Drosophila gastrulation. Curr Biol 20: 1562–1567. doi: 10.1016/j.cub.2010.07.043 20797865

22. Perry MW, Boettiger AN, Levine M (2011) Multiple enhancers ensure precision of gap gene-expression patterns in the Drosophila embryo. Proc Natl Acad Sci U S A 108: 13570–13575. doi: 10.1073/pnas.1109873108 21825127

23. Waddington CH (1942) Canalization of development and the inheritance of acquired characters. Nature 150: 563–565. 14986860

24. Yaswen L, Diehl N, Brennan MB, Hochgeschwender U (1999) Obesity in the mouse model of pro-opiomelanocortin deficiency responds to peripheral melanocortin. Nat Med 5: 1066–1070. 10470087

25. Krude H, Biebermann H, Luck W, Horn R, Brabant G, et al. (1998) Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet 19: 155–157. 9620771

26. Bumaschny VF, Yamashita M, Casas-Cordero R, Otero-Corchón V, de Souza FSJ, et al. (2012) Obesity-programmed mice are rescued by early genetic intervention. J Clin Invest 122: 4203–4212. doi: 10.1172/JCI62543 23093774

27. De Souza FSJ, Santangelo AM, Bumaschny V, Avale ME, Smart JL, et al. (2005) Identification of neuronal enhancers of the proopiomelanocortin gene by transgenic mouse analysis and phylogenetic footprinting. Mol Cell Biol 25: 3076–3086. 15798195

28. Franchini LF, López-Leal R, Nasif S, Beati P, Gelman DM, et al. (2011) Convergent evolution of two mammalian neuronal enhancers by sequential exaptation of unrelated retroposons. Proc Natl Acad Sci U S A 108: 15270–15275. doi: 10.1073/pnas.1104997108 21876128

29. Santangelo AM, de Souza FSJ, Franchini LF, Bumaschny VF, Low MJ, et al. (2007) Ancient exaptation of a CORE-SINE retroposon into a highly conserved mammalian neuronal enhancer of the proopiomelanocortin gene. PLoS Genet 3: 1813–1826. 17922573

30. Langlais D, Couture C, Sylvain-Drolet G, Drouin J (2011) A pituitary-specific enhancer of the POMC gene with preferential activity in corticotrope cells. Mol Endocrinol 25: 348–359. doi: 10.1210/me.2010-0422 21193556

31. Elkabes S, Loh YP, Nieburgs A, Wray S (1989) Prenatal ontogenesis of pro-opiomelanocortin in the mouse central nervous system and pituitary gland: an in situ hybridization and immunocytochemical study. Brain Res Dev Brain Res 46: 85–95. 2706773

32. Japón MA, Rubinstein M, Low MJ (1994) In situ hybridization analysis of anterior pituitary hormone gene expression during fetal mouse development. J Histochem Cytochem 42: 1117–1125.

33. Zhan C, Zhou J, Feng Q, Zhang J-E, Lin S, et al. (2013) Acute and long-term suppression of feeding behavior by POMC neurons in the brainstem and hypothalamus, respectively. J Neurosci 33: 3624–3632. doi: 10.1523/JNEUROSCI.2742-12.2013 23426689

34. Conant GC, Wolfe KH (2008) Turning a hobby into a job: how duplicated genes find new functions. Nat Rev Genet 9: 938–950. doi: 10.1038/nrg2482 19015656

35. De Souza FSJ, Nasif S, López-Leal R, Levi DH, Low MJ, et al. (2011) The estrogen receptor α colocalizes with proopiomelanocortin in hypothalamic neurons and binds to a conserved motif present in the neuron-specific enhancer nPE2. Eur J Pharmacol 660: 181–187. doi: 10.1016/j.ejphar.2010.10.114 21211522

36. Smart JL, Tolle V, Low MJ (2006) Glucocorticoids exacerbate obesity and insulin resistance in neuron-specific proopiomelanocortin-deficient mice. J Clin Invest 116: 495–505. 16440060

37. Padilla SL, Carmody JS, Zeltser LM (2010) Pomc-expressing progenitors give rise to antagonistic neuronal populations in hypothalamic feeding circuits. Nat Med 16: 403–405. doi: 10.1038/nm.2126 20348924

38. Dvornyk V, Vinogradova O, Nevo E (2003) Origin and evolution of circadian clock genes in prokaryotes. Proc Natl Acad Sci U S A 100: 2495–2500. 12604787

39. Speakman JR (2013) Evolutionary perspectives on the obesity epidemic: adaptive, maladaptive, and neutral viewpoints. Annu Rev Nutr 33: 289–317. doi: 10.1146/annurev-nutr-071811-150711 23862645

40. Hammoud AO, Gibson M, Peterson CM, Meikle AW, Carrell DT (2008) Impact of male obesity on infertility: a critical review of the current literature. Fertil Steril 90: 897–904. doi: 10.1016/j.fertnstert.2008.08.026 18929048

41. Brewer CJ, Balen AH (2010) The adverse effects of obesity on conception and implantation. Reproduction 140: 347–364. 20395425

42. Sakabe NJ, Nobrega MA (2013) Beyond the ENCODE project: using genomics and epigenomics strategies to study enhancer evolution. Philos Trans R Soc Lond B Biol Sci 368: 20130022. doi: 10.1098/rstb.2013.0022 24218635

43. Sakabe NJ, Savic D, Nobrega MA (2012) Transcriptional enhancers in development and disease. Genome Biol 13: 238.

44. Smemo S, Tena JJ, Kim K-H, Gamazon ER, Sakabe NJ, et al. (2014) Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature 507: 371–375. doi: 10.1038/nature13138 24646999

45. Graff M, Ngwa JS, Workalemahu T, Homuth G, Schipf S, et al. (2013) Genome-wide analysis of BMI in adolescents and young adults reveals additional insight into the effects of genetic loci over the life course. Hum Mol Genet 22: 3597–3607. doi: 10.1093/hmg/ddt205 23669352

46. Cousminer DL, Berry DJ, Timpson NJ, Ang W, Thiering E, et al. (2013) Genome-wide association and longitudinal analyses reveal genetic loci linking pubertal height growth, pubertal timing and childhood adiposity. Hum Mol Genet 22: 2735–2747. doi: 10.1093/hmg/ddt104 23449627

47. Speliotes EK, Willer CJ, Berndt SI, Monda KL, Thorleifsson G, et al. (2010) Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat Genet 42: 937–948. 20935630

48. Queitsch C, Carlson KD, Girirajan S (2012) Lessons from model organisms: phenotypic robustness and missing heritability in complex disease. PLoS Genet 8: e1003041. doi: 10.1371/journal.pgen.1003041 23166511

49. Symmons O, Uslu VV, Tsujimura T, Ruf S, Nassari S, et al. (2014) Functional and topological characteristics of mammalian regulatory domains. Genome Res 24: 390–400. doi: 10.1101/gr.163519.113 24398455

50. Marinić M, Aktas T, Ruf S, Spitz F (2013) An integrated holo-enhancer unit defines tissue and gene specificity of the Fgf8 regulatory landscape. Dev Cell 24: 530–542. doi: 10.1016/j.devcel.2013.01.025 23453598

51. Mali P, Yang L, Esvelt KM, Aach J, Guell M, et al. (2013) RNA-guided human genome engineering via Cas9. Science 339: 823–826. 23287722

52. Gaj T, Gersbach CA, Barbas CF (2013) ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 31: 397–405.

53. Larkin MA, Blackshields G, Brown NP, Chenna R, McGettigan PA, et al. (2007) Clustal W and Clustal X version 2.0. Bioinformatics 23: 2947–2948. 17846036

54. Cowley MA, Smart JL, Rubinstein M, Cerdán MG, Diano S, et al. (2001) Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411: 480–484. 11373681

55. Li E, Bestor TH, Jaenisch R (1992) Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69: 915–926. 1606615

56. Schwenk F, Baron U, Rajewsky K (1995) A cre-transgenic mouse strain for the ubiquitous deletion of loxP-flanked gene segments including deletion in germ cells. Nucleic Acids Res 23: 5080–5081. 8559668

57. Savontaus E, Breen TL, Kim A, Yang LM, Chua SC, et al. (2004) Metabolic effects of transgenic melanocyte-stimulating hormone overexpression in lean and obese mice. Endocrinology 145: 3881–3891. 15117873

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 2
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#