Semin Reprod Med 2019; 37(04): 166-173
DOI: 10.1055/s-0039-3400965
Review Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Evolutionary Conservation of MKRN3 and Other Makorins and Their Roles in Puberty Initiation and Endocrine Functions

Lydie Naulé
1   Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
,
Ursula B. Kaiser
1   Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
› Author Affiliations
Further Information

Publication History

Publication Date:
23 January 2020 (online)

Abstract

Puberty is a critical period of development regulated by genetic, nutritional, and environmental factors. The role of makorin ring finger protein 3 (MKRN3) in the regulation of pubertal timing was revealed when loss-of-function mutations were identified in patients with central precocious puberty (CPP). To date, MKRN3 mutations are the most common known genetic cause of CPP. MKRN3 is a member of the makorin family of ubiquitin ligases, together with MKRN1 and MKRN2. The Mkrn genes have been identified in both vertebrates and invertebrates and show high evolutionary conservation of their gene and protein structures. While the existence of Mkrn orthologues in a wide spectrum of species suggests a vital cellular role of the makorins, their role in puberty initiation and endocrine functions is just beginning to be investigated. In this review, we discuss recent studies that have shown the involvement of Mkrn3 and other makorins in the regulation of pubertal development and other endocrine functions, including metabolism and fertility, as well as their underlying mechanisms of action.

 
  • References

  • 1 Abreu AP, Kaiser UB. Pubertal development and regulation. Lancet Diabetes Endocrinol 2016; 4 (03) 254-264
  • 2 Plant TM. Neuroendocrine control of the onset of puberty. Front Neuroendocrinol 2015; 38: 73-88
  • 3 Avendaño MS, Vazquez MJ, Tena-Sempere M. Disentangling puberty: novel neuroendocrine pathways and mechanisms for the control of mammalian puberty. Hum Reprod Update 2017; 23 (06) 737-763
  • 4 Palmert MR, Boepple PA. Variation in the timing of puberty: clinical spectrum and genetic investigation. J Clin Endocrinol Metab 2001; 86 (06) 2364-2368
  • 5 Schwanzel-Fukuda M, Bick D, Pfaff DW. Luteinizing hormone-releasing hormone (LHRH)-expressing cells do not migrate normally in an inherited hypogonadal (Kallmann) syndrome. Brain Res Mol Brain Res 1989; 6 (04) 311-326
  • 6 Bianco SDC, Kaiser UB. The genetic and molecular basis of idiopathic hypogonadotropic hypogonadism. Nat Rev Endocrinol 2009; 5 (10) 569-576
  • 7 Clarke SA, Dhillo WS. Kisspeptin across the human lifespan: evidence from animal studies and beyond. J Endocrinol 2016; 229 (03) R83-R98
  • 8 Seminara SB, Messager S, Chatzidaki EE. , et al. The GPR54 gene as a regulator of puberty. N Engl J Med 2003; 349 (17) 1614-1627
  • 9 de Roux N, Genin E, Carel J-C, Matsuda F, Chaussain J-L, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A 2003; 100 (19) 10972-10976
  • 10 Teles MG, Bianco SDC, Brito VN. , et al. A GPR54-activating mutation in a patient with central precocious puberty. N Engl J Med 2008; 358 (07) 709-715
  • 11 Silveira LG, Noel SD, Silveira-Neto AP. , et al. Mutations of the KISS1 gene in disorders of puberty. J Clin Endocrinol Metab 2010; 95 (05) 2276-2280
  • 12 Topaloglu AK, Reimann F, Guclu M. , et al. TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for Neurokinin B in the central control of reproduction. Nat Genet 2009; 41 (03) 354-358
  • 13 Young J, Bouligand J, Francou B. , et al. TAC3 and TACR3 defects cause hypothalamic congenital hypogonadotropic hypogonadism in humans. J Clin Endocrinol Metab 2010; 95 (05) 2287-2295
  • 14 Gianetti E, Tusset C, Noel SD. , et al. TAC3/TACR3 mutations reveal preferential activation of gonadotropin-releasing hormone release by neurokinin B in neonatal life followed by reversal in adulthood. J Clin Endocrinol Metab 2010; 95 (06) 2857-2867
  • 15 Navarro VM, Gottsch ML, Chavkin C, Okamura H, Clifton DK, Steiner RA. Regulation of gonadotropin-releasing hormone secretion by kisspeptin/dynorphin/neurokinin B neurons in the arcuate nucleus of the mouse. J Neurosci 2009; 29 (38) 11859-11866
  • 16 Lehman MN, Coolen LM, Goodman RL. Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion. Endocrinology 2010; 151 (08) 3479-3489
  • 17 Clarkson J, Herbison AE. Development of GABA and glutamate signaling at the GnRH neuron in relation to puberty. Mol Cell Endocrinol 2006; 254-255: 32-38
  • 18 Abreu AP, Dauber A, Macedo DB. , et al. Central precocious puberty caused by mutations in the imprinted gene MKRN3. N Engl J Med 2013; 368 (26) 2467-2475
  • 19 Dauber A, Cunha-Silva M, Macedo DB. , et al. Paternally inherited DLK1 deletion associated with familial central precocious puberty. J Clin Endocrinol Metab 2017; 102 (05) 1557-1567
  • 20 Gomes LG, Cunha-Silva M, Crespo RP. , et al. DLK1 is a novel link between reproduction and metabolism. J Clin Endocrinol Metab 2019; 104 (06) 2112-2120
  • 21 Settas N, Dacou-Voutetakis C, Karantza M, Kanaka-Gantenbein C, Chrousos GP, Voutetakis A. Central precocious puberty in a girl and early puberty in her brother caused by a novel mutation in the MKRN3 gene. J Clin Endocrinol Metab 2014; 99 (04) E647-E651
  • 22 Schreiner F, Gohlke B, Hamm M, Korsch E, Woelfle J. MKRN3 mutations in familial central precocious puberty. Horm Res Paediatr 2014; 82 (02) 122-126
  • 23 de Vries L, Gat-Yablonski G, Dror N, Singer A, Phillip M. A novel MKRN3 missense mutation causing familial precocious puberty. Hum Reprod 2014; 29 (12) 2838-2843
  • 24 Macedo DB, Abreu AP, Reis ACS. , et al. Central precocious puberty that appears to be sporadic caused by paternally inherited mutations in the imprinted gene makorin ring finger 3. J Clin Endocrinol Metab 2014; 99 (06) E1097-E1103
  • 25 Grandone A, Cantelmi G, Cirillo G. , et al. A case of familial central precocious puberty caused by a novel mutation in the makorin RING finger protein 3 gene. BMC Endocr Disord 2015; 15: 60
  • 26 Simon D, Ba I, Mekhail N. , et al. Mutations in the maternally imprinted gene MKRN3 are common in familial central precocious puberty. Eur J Endocrinol 2016; 174 (01) 1-8
  • 27 Simsek E, Demiral M, Ceylaner S, Kırel B. Two frameshift mutations in MKRN3 in Turkish patients with familial central precocious puberty. Horm Res Paediatr 2017; 87 (06) 405-411
  • 28 Lee HS, Jin HS, Shim YS. , et al. Low frequency of MKRN3 mutations in central precocious puberty among Korean girls. Horm Metab Res 2016; 48 (02) 118-122
  • 29 Ortiz-Cabrera NV, Riveiro-Álvarez R, López-Martínez MÁ. , et al. Clinical exome sequencing reveals MKRN3 pathogenic variants in familial and nonfamilial idiopathic central precocious puberty. Horm Res Paediatr 2017; 87 (02) 88-94
  • 30 Nishioka J, Shima H, Fukami M. , et al. The first Japanese case of central precocious puberty with a novel MKRN3 mutation. Hum Genome Var 2017; 4: 17017
  • 31 Bessa DS, Macedo DB, Brito VN. , et al. High frequency of MKRN3 mutations in male central precocious puberty previously classified as idiopathic. Neuroendocrinology 2017; 105 (01) 17-25
  • 32 Stecchini MF, Macedo DB, Reis ACS. , et al. Time course of central precocious puberty development caused by an MKRN3 gene mutation: a prismatic case. Horm Res Paediatr 2016; 86 (02) 126-130
  • 33 Valadares LP, Meireles CG, De Toledo IP. , et al. MKRN3 mutations in central precocious puberty: a systematic review and meta-analysis. J Endocr Soc 2019; 3 (05) 979-995
  • 34 Rosenfield RL, Lipton RB, Drum ML. Thelarche, pubarche, and menarche attainment in children with normal and elevated body mass index. Pediatrics 2009; 123 (01) 84-88
  • 35 Day FR, Elks CE, Murray A, Ong KK, Perry JRB. Puberty timing associated with diabetes, cardiovascular disease and also diverse health outcomes in men and women: the UK Biobank study. Sci Rep 2015; 5: 11208
  • 36 Day FR, Thompson DJ, Helgason H. , et al; LifeLines Cohort Study; InterAct Consortium; kConFab/AOCS Investigators; Endometrial Cancer Association Consortium; Ovarian Cancer Association Consortium; PRACTICAL consortium. Genomic analyses identify hundreds of variants associated with age at menarche and support a role for puberty timing in cancer risk. Nat Genet 2017; 49 (06) 834-841
  • 37 Lakshman R, Forouhi NG, Sharp SJ. , et al. Early age at menarche associated with cardiovascular disease and mortality. J Clin Endocrinol Metab 2009; 94 (12) 4953-4960
  • 38 Perry JRB, Day F, Elks CE. , et al; Australian Ovarian Cancer Study; GENICA Network; kConFab; LifeLines Cohort Study; InterAct Consortium; Early Growth Genetics (EGG) Consortium. Parent-of-origin-specific allelic associations among 106 genomic loci for age at menarche. Nature 2014; 514 (7520): 92-97
  • 39 Jong MT, Gray TA, Ji Y. , et al. A novel imprinted gene, encoding a RING zinc-finger protein, and overlapping antisense transcript in the Prader-Willi syndrome critical region. Hum Mol Genet 1999; 8 (05) 783-793
  • 40 Jong MT, Carey AH, Caldwell KA. , et al. Imprinting of a RING zinc-finger encoding gene in the mouse chromosome region homologous to the Prader-Willi syndrome genetic region. Hum Mol Genet 1999; 8 (05) 795-803
  • 41 Hall TMT. Multiple modes of RNA recognition by zinc finger proteins. Curr Opin Struct Biol 2005; 15 (03) 367-373
  • 42 Deshaies RJ, Joazeiro CAP. RING domain E3 ubiquitin ligases. Annu Rev Biochem 2009; 78: 399-434
  • 43 Gray TA, Hernandez L, Carey AH. , et al. The ancient source of a distinct gene family encoding proteins featuring RING and C(3)H zinc-finger motifs with abundant expression in developing brain and nervous system. Genomics 2000; 66 (01) 76-86
  • 44 Zhang QH, Ye M, Wu XY. , et al. Cloning and functional analysis of cDNAs with open reading frames for 300 previously undefined genes expressed in CD34+ hematopoietic stem/progenitor cells. Genome Res 2000; 10 (10) 1546-1560
  • 45 Gray TA, Azama K, Whitmore K, Min A, Abe S, Nicholls RD. Phylogenetic conservation of the makorin-2 gene, encoding a multiple zinc-finger protein, antisense to the RAF1 proto-oncogene. Genomics 2001; 77 (03) 119-126
  • 46 Böhne A, Darras A, D'Cotta H, Baroiller JF, Galiana-Arnoux D, Volff JN. The vertebrate makorin ubiquitin ligase gene family has been shaped by large-scale duplication and retroposition from an ancestral gonad-specific, maternal-effect gene. BMC Genomics 2010; 11: 721
  • 47 Rapkins RW, Hore T, Smithwick M. , et al. Recent assembly of an imprinted domain from non-imprinted components. PLoS Genet 2006; 2 (10) e182
  • 48 Gray TA, Wilson A, Fortin PJ, Nicholls RD. The putatively functional Mkrn1-p1 pseudogene is neither expressed nor imprinted, nor does it regulate its source gene in trans. Proc Natl Acad Sci U S A 2006; 103 (32) 12039-12044
  • 49 Kim JH, Park SM, Kang MR. , et al. Ubiquitin ligase MKRN1 modulates telomere length homeostasis through a proteolysis of hTERT. Genes Dev 2005; 19 (07) 776-781
  • 50 Lee E-W, Lee MS, Camus S. , et al. Differential regulation of p53 and p21 by MKRN1 E3 ligase controls cell cycle arrest and apoptosis. EMBO J 2009; 28 (14) 2100-2113
  • 51 Kim JH, Park KW, Lee EW. , et al. Suppression of PPARγ through MKRN1-mediated ubiquitination and degradation prevents adipocyte differentiation. Cell Death Differ 2014; 21 (04) 594-603
  • 52 Lee MS, Han HJ, Han SY. , et al. Loss of the E3 ubiquitin ligase MKRN1 represses diet-induced metabolic syndrome through AMPK activation. Nat Commun 2018; 9 (01) 3404
  • 53 Omwancha J, Zhou XF, Chen SY. , et al. Makorin RING finger protein 1 (MKRN1) has negative and positive effects on RNA polymerase II-dependent transcription. Endocrine 2006; 29 (02) 363-373
  • 54 Miroci H, Schob C, Kindler S. , et al. Makorin ring zinc finger protein 1 (MKRN1), a novel poly(A)-binding protein-interacting protein, stimulates translation in nerve cells. J Biol Chem 2012; 287 (02) 1322-1334
  • 55 Cassar PA, Carpenedo RL, Samavarchi-Tehrani P. , et al. Integrative genomics positions MKRN1 as a novel ribonucleoprotein within the embryonic stem cell gene regulatory network. EMBO Rep 2015; 16 (10) 1334-1357
  • 56 Qian X, Wang L, Zheng B. , et al. Deficiency of Mkrn2 causes abnormal spermiogenesis and spermiation, and impairs male fertility. Sci Rep 2016; 6: 39318
  • 57 Yang PH, Cheung WKC, Peng Y. , et al. Makorin-2 is a neurogenesis inhibitor downstream of phosphatidylinositol 3-kinase/Akt (PI3K/Akt) signal. J Biol Chem 2008; 283 (13) 8486-8495
  • 58 Cheung WKC, Yang PH, Huang QH. , et al. Identification of protein domains required for makorin-2-mediated neurogenesis inhibition in Xenopus embryos. Biochem Biophys Res Commun 2010; 394 (01) 18-23
  • 59 Shin C, Ito Y, Ichikawa S, Tokunaga M, Sakata-Sogawa K, Tanaka T. MKRN2 is a novel ubiquitin E3 ligase for the p65 subunit of NF-κB and negatively regulates inflammatory responses. Sci Rep 2017; 7: 46097
  • 60 Kanber D, Giltay J, Wieczorek D. , et al. A paternal deletion of MKRN3, MAGEL2 and NDN does not result in Prader-Willi syndrome. Eur J Hum Genet 2009; 17 (05) 582-590
  • 61 Driscoll DJ, Waters MF, Williams CA. , et al. A DNA methylation imprint, determined by the sex of the parent, distinguishes the Angelman and Prader-Willi syndromes. Genomics 1992; 13 (04) 917-924
  • 62 Hershko A, Razin A, Shemer R. Imprinted methylation and its effect on expression of the mouse Zfp127 gene. Gene 1999; 234 (02) 323-327
  • 63 Nicholls RD, Saitoh S, Horsthemke B. Imprinting in Prader-Willi and Angelman syndromes. Trends Genet 1998; 14 (05) 194-200
  • 64 Liu H, Kong X, Chen F. Mkrn3 functions as a novel ubiquitin E3 ligase to inhibit Nptx1 during puberty initiation. Oncotarget 2017; 8 (49) 85102-85109
  • 65 Lund C, Pulli K, Yellapragada V. , et al. Development of gonadotropin-releasing hormone-secreting neurons from human pluripotent stem cells. Stem Cell Reports 2016; 7 (02) 149-157
  • 66 Yellapragada V, Liu X, Lund C. , et al. MKRN3 interacts with several proteins implicated in puberty timing but does not influence GNRH1 expression. Front Endocrinol (Lausanne) 2019; 10: 48
  • 67 Tran HT, Cho E, Jeong S. , et al. Makorin 1 regulates developmental timing in drosophila. Mol Cells 2018; 41 (12) 1024-1032
  • 68 Jeong EB, Jeong SS, Cho E, Kim EY. Makorin 1 is required for Drosophila oogenesis by regulating insulin/Tor signaling. PLoS One 2019; 14 (04) e0215688
  • 69 Herrera RA, Kiontke K, Fitch DHA. Makorin ortholog LEP-2 regulates LIN-28 stability to promote the juvenile-to-adult transition in Caenorhabditis elegans . Development 2016; 143 (05) 799-809
  • 70 Lawson H, Vuong E, Miller RM, Kiontke K, Fitch DH, Portman DS. The Makorin lep-2 and the lncRNA lep-5 regulate lin-28 to schedule sexual maturation of the C. elegans nervous system. eLife 2019; 8: e43660