Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2018, Vol. 12 Issue (5) : 525-532    https://doi.org/10.1007/s11684-017-0577-9
RESEARCH ARTICLE
Role of chemerin/CMKLR1 in the maintenance of early pregnancy
Xuezhou Yang1, Junning Yao2, Qipeng Wei1, Jinhai Ye1, Xiaofang Yin1, Xiaozhen Quan1, Yanli Lan1, Hui Xing1()
1. Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China
2. Department of Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
 Download: PDF(239 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Chemerin is a cytokine that attracts much attention in the reproductive process. This study aimed to explore the effects of chemerin and its receptor chemokine-like receptor 1 (CMKLR1) on the maintenance of early pregnancy. The expression levels of chemerin and CMKLR1 in the decidua tissues of 20 early normal pregnant women and 20 early spontaneous abortion women were examined by Western blot and real-time polymerase chain reaction analyses. CMKLR1 receptor antagonist (α-NETA) was then intrauterinely injected into normal pregnant mice model to assess its effect on the outcome of pregnancy and the phosphorylation rate of ERK1/2 in decidua tissues. We found that the expression level of chemerin in women who had experienced early spontaneous abortion was lower than in those who had experienced normal early pregnancy (P<0.01); conversely, CMKLR1 expression was higher in the former than in the latter (P<0.01). In a pregnant-mouse model, the embryo resorption rate of α-NETA group was higher than that in the negative control group (61.5% vs. 10.8%) (P<0.001). Compared with the control group, ERK1/2 phosphorylation in decidua tissues decreased in the α-NETA-treated group (P<0.01). These results suggested that the inhibition of the chemerin/CMKLR1 signaling pathway can lead to the abortion of mouse embryos, and that chemerin/CMKLR1 may play an important role in the maintenance of early pregnancy possibly by regulating ERK1/2 phosphorylation.

Keywords chemerin      CMKLR1      early abortion     
Corresponding Author(s): Hui Xing   
Just Accepted Date: 27 December 2017   Online First Date: 20 March 2018    Issue Date: 29 September 2018
 Cite this article:   
Xuezhou Yang,Junning Yao,Qipeng Wei, et al. Role of chemerin/CMKLR1 in the maintenance of early pregnancy[J]. Front. Med., 2018, 12(5): 525-532.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-017-0577-9
https://academic.hep.com.cn/fmd/EN/Y2018/V12/I5/525
Gene Primers Sequence (5′–3′)
Chemerin F 5′-GAAGAAACCCGAGTGCAAAG-3′
R 5′-CTTGGAGAAGGCGACTGTC-3′
CMKLR1 F 5′-CGGGTGATAGGGGTGTTCCA-3′
R 5′-GCACACCCTGGCTTCTTTCT-3′
ACTB F 5′-CATGTACGTTGCTACCGC-3′
R 5′-CTCCTTAATGTCACGCACGAT-3′
Tab.1  Primer sequences (all 5′–3′) used in real-time-PCR
Fig.1  Expression of chemerin and CMKLR1 in decidua tissues of early spontaneous abortion women and normal early pregnancy women. (A) Relative mRNA levels of chemerin in the decidua tissues of early spontaneous abortion women and normal early pregnant women were tested by real-time PCR. ** P<0.01. (B) Quantitative presentation of data shown in Fig.1E. ** P<0.01. (C) Relative mRNA levels of CMKLR1 in the decidua tissues of early spontaneous abortion women and normal early pregnancy women were tested by real-time PCR. * P<0.05. (D) Quantitative presentation of data shown in Fig.1E. *P<0.05. (E) Protein levels of chemerin and CMKLR1 in the decidua tissues of early spontaneous abortion women and normal early pregnancy women were examined by Western blot. GAPDH was used as a control.
Fig.2  Effect of antagonist of CMKLR1 on the continued pregnancy of mice. (A) Abortion rate of different groups. *** P<0.001. (B) Representative pictures of embryos attached to the uterus in different groups. Picture α-NETA showed the uteri injected with α-NETA and the ▾ site was the abortion embryo. Picture PBS showed the uteri received PBS injection, whereas None represented the uteri, which were injected with nothing.
Fig.3  Effect of antagonist of CMKLR1 on the phosphorylation of ERK1/2. (A) Level of phosphorylated ERK1/2 (phos ERK1/2) in decidua tissues in different groups by Western blot. (B) Quantitative presentation of data shown in Fig.3A. ** P<0.01.
1 Rai R, Regan L. Recurrent miscarriage. Lancet 2006; 368(9535): 601–611
https://doi.org/10.1016/S0140-6736(06)69204-0 pmid: 16905025
2 Saravelos SH, Cocksedge KA, Li TC. The pattern of pregnancy loss in women with congenital uterine anomalies and recurrent miscarriage. Reprod Biomed Online 2010; 20(3): 416–422
https://doi.org/10.1016/j.rbmo.2009.11.021 pmid: 20093084
3 Branch DW, Gibson M, Silver RM. Recurrent miscarriage. N Engl J Med 2010; 363(18): 1740–1747
https://doi.org/10.1056/NEJMcp1005330 pmid: 20979474
4 Sugiura-Ogasawara M, Aoki K, Fujii T, Fujita T, Kawaguchi R, Maruyama T, Ozawa N, Sugi T, Takeshita T, Saito S. Subsequent pregnancy outcomes in recurrent miscarriage patients with a paternal or maternal carrier of a structural chromosome rearrangement. J Hum Genet 2008; 53(7): 622–628
https://doi.org/10.1007/s10038-008-0290-2 pmid: 18414779
5 Su MT, Lin SH, Chen YC. Genetic association studies of angiogenesis- and vasoconstriction-related genes in women with recurrent pregnancy loss: a systematic review and meta-analysis. Hum Reprod Update 2011; 17(6): 803–812
https://doi.org/10.1093/humupd/dmr027 pmid: 21642294
6 Franssen MT, Musters AM, van der Veen F, Repping S, Leschot NJ, Bossuyt PM, Goddijn M, Korevaar JC. Reproductive outcome after PGD in couples with recurrent miscarriage carrying a structural chromosome abnormality: a systematic review. Hum Reprod Update 2011; 17(4): 467–475
https://doi.org/10.1093/humupd/dmr011 pmid: 21504961
7 Arredondo F, Noble LS. Endocrinology of recurrent pregnancy loss. Semin Reprod Med 2006; 24(1): 33–39
https://doi.org/10.1055/s-2006-931799 pmid: 16418976
8 Saravelos SH, Cocksedge KA, Li TC. Prevalence and diagnosis of congenital uterine anomalies in women with reproductive failure: a critical appraisal. Hum Reprod Update 2008; 14(5): 415–429
https://doi.org/10.1093/humupd/dmn018 pmid: 18539641
9 Saravelos SH, Yan J, Rehmani H, Li TC. The prevalence and impact of fibroids and their treatment on the outcome of pregnancy in women with recurrent miscarriage. Hum Reprod 2011; 26(12): 3274–3279
https://doi.org/10.1093/humrep/der293 pmid: 21954281
10 Alijotas-Reig J, Ferrer-Oliveras R; EUROAPS Study Group. The European Registry on Obstetric Antiphospholipid Syndrome (EUROAPS): a preliminary first year report. Lupus 2012; 21(7): 766–768
https://doi.org/10.1177/0961203312440058 pmid: 22635227
11 Danza A, Ruiz-Irastorza G, Khamashta M. Antiphospohlipid syndrome in obstetrics. Best Pract Res Clin Obstet Gynaecol 2012; 26(1): 65–76
https://doi.org/10.1016/j.bpobgyn.2011.10.006 pmid: 22079775
12 Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature 2012; 490(7418): 102–106
https://doi.org/10.1038/nature11462 pmid: 23023128
13 Tang AW, Alfirevic Z, Quenby S. Natural killer cells and pregnancy outcomes in women with recurrent miscarriage and infertility: a systematic review. Hum Reprod 2011; 26(8): 1971–1980
https://doi.org/10.1093/humrep/der164 pmid: 21613313
14 Davenport WB, Kutteh WH. Inherited thrombophilias and adverse pregnancy outcomes: a review of screening patterns and recommendations. Obstet Gynecol Clin North Am 2014; 41(1): 133–144
https://doi.org/10.1016/j.ogc.2013.10.005 pmid: 24491988
15 Ernst MC, Sinal CJ. Chemerin: at the crossroads of inflammation and obesity. Trends Endocrinol Metab 2010; 21(11): 660–667
https://doi.org/10.1016/j.tem.2010.08.001 pmid: 20817486
16 Corona-Meraz FI, Navarro-Hernandez RE, Ruiz-Quezada SL, Madrigal-Ruiz PM, Castro-Albarran J, Chavarria-Avila E, Guzman-Ornelas MO, Gomez-Banuelos E, Petri MH, Ramirez-Cedano JI, Aguilar-Aldrete ME, Rios-Ibarra C, Vazquez-Del Mercado M. Inverse relationship of the CMKLR1 relative expression and chemerin serum levels in obesity with dysmetabolic phenotype and insulin resistance. Mediators Inflamm 2016; 2016:3085390
https://doi.org/10.1155/2016/3085390 pmid: 27239101
17 Becker M, Rabe K, Lebherz C, Zugwurst J, Göke B, Parhofer KG, Lehrke M, Broedl UC. Expression of human chemerin induces insulin resistance in the skeletal muscle but does not affect weight, lipid levels, and atherosclerosis in LDL receptor knockout mice on high-fat diet. Diabetes 2010; 59(11): 2898–2903
https://doi.org/10.2337/db10-0362 pmid: 20724582
18 Sell H, Laurencikiene J, Taube A, Eckardt K, Cramer A, Horrighs A, Arner P, Eckel J. Chemerin is a novel adipocyte-derived factor inducing insulin resistance in primary human skeletal muscle cells. Diabetes 2009; 58(12): 2731–2740
https://doi.org/10.2337/db09-0277 pmid: 19720798
19 Kennedy AJ, Yang P, Read C, Kuc RE, Yang L, Taylor EJ, Taylor CW, Maguire JJ, Davenport AP. Chemerin elicits potent constrictor actions via chemokine-like receptor 1 (CMKLR1), not G-protein-coupled receptor 1 (GPR1), in human and rat vasculature. J Am Heart Assoc 2016; 5(10): e004421
https://doi.org/10.1161/JAHA.116.004421 pmid: 27742615
20 Ferland DJ, Watts SW. Chemerin: a comprehensive review elucidating the need for cardiovascular research. Pharmacol Res 2015; 99: 351–361
https://doi.org/10.1016/j.phrs.2015.07.018 pmid: 26211950
21 Dessein PH, Tsang L, Woodiwiss AJ, Norton GR, Solomon A. Circulating concentrations of the novel adipokine chemerin are associated with cardiovascular disease risk in rheumatoid arthritis. J Rheumatol 2014; 41(9): 1746–1754
https://doi.org/10.3899/jrheum.140122 pmid: 25028378
22 Samson M, Edinger AL, Stordeur P, Rucker J, Verhasselt V, Sharron M, Govaerts C, Mollereau C, Vassart G, Doms RW, Parmentier M. ChemR23, a putative chemoattractant receptor, is expressed in monocyte-derived dendritic cells and macrophages and is a coreceptor for SIV and some primary HIV-1 strains. Eur J Immunol 1998; 28(5): 1689–1700
https://doi.org/10.1002/(SICI)1521-4141(199805)28:05<1689::AID-IMMU1689>3.0.CO;2-I pmid: 9603476
23 Parolini S, Santoro A, Marcenaro E, Luini W, Massardi L, Facchetti F, Communi D, Parmentier M, Majorana A, Sironi M, Tabellini G, Moretta A, Sozzani S. The role of chemerin in the colocalization of NK and dendritic cell subsets into inflamed tissues. Blood 2007; 109(9): 3625–3632
https://doi.org/10.1182/blood-2006-08-038844 pmid: 17202316
24 Luangsay S, Wittamer V, Bondue B, De Henau O, Rouger L, Brait M, Franssen JD, de Nadai P, Huaux F, Parmentier M. Mouse ChemR23 is expressed in dendritic cell subsets and macrophages, and mediates an anti-inflammatory activity of chemerin in a lung disease model. J Immunol 2009; 183(10): 6489–6499
https://doi.org/10.4049/jimmunol.0901037 pmid: 19841182
25 Garces MF, Sanchez E, Ruíz-Parra AI, Rubio-Romero JA, Angel-Müller E, Suarez MA, Bohórquez LF, Bravo SB, Nogueiras R, Diéguez C, Caminos JE. Serum chemerin levels during normal human pregnancy. Peptides 2013; 42: 138–143
https://doi.org/10.1016/j.peptides.2013.01.003 pmid: 23313148
26 Boutsikou T, Briana DD, Boutsikou M, Kafalidis G, Stamati L, Baka S, Hassiakos D, Gourgiotis D, Malamitsi-Puchner A. Cord blood chemerin and obestatin levels in large for gestational age infants. J Matern Fetal Neonatal Med 2013; 26(2): 123–126
https://doi.org/10.3109/14767058.2012.728648 pmid: 22963327
27 Garces MF, Sanchez E, Acosta BJ, Angel E, Ruíz AI, Rubio-Romero JA, Diéguez C, Nogueiras R, Caminos JE. Expression and regulation of chemerin during rat pregnancy. Placenta 2012; 33(5): 373–378
https://doi.org/10.1016/j.placenta.2012.02.007 pmid: 22369992
28 Carlino C, Trotta E, Stabile H, Morrone S, Bulla R, Soriani A, Iannitto ML, Agostinis C, Mocci C, Minozzi M, Aragona C, Perniola G, Tedesco F, Sozzani S, Santoni A, Gismondi A. Chemerin regulates NK cell accumulation and endothelial cell morphogenesis in the decidua during early pregnancy. J Clin Endocrinol Metab 2012; 97(10): 3603–3612
https://doi.org/10.1210/jc.2012-1102 pmid: 22791765
29 Duan DM, Niu JM, Lei Q, Lin XH, Chen X. Serum levels of the adipokine chemerin in preeclampsia. J Perinat Med 2011; 40(2): 121–127
pmid: 22117111
30 Wang Q, Leader A, Tsang BK. Inhibitory roles of prohibitin and chemerin in FSH-induced rat granulosa cell steroidogenesis. Endocrinology 2013; 154(2): 956–967
https://doi.org/10.1210/en.2012-1836 pmid: 23254195
31 Lee KJ, Shim SH, Kang KM, Kang JH, Park DY, Kim SH, Farina A, Shim SS, Cha DH. Global gene expression changes induced in the human placenta during labor. Placenta 2010; 31(8): 698–704
https://doi.org/10.1016/j.placenta.2010.05.006 pmid: 20554320
32 Trowsdale J, Betz AG. Mother’s little helpers: mechanisms of maternal-fetal tolerance. Nat Immunol 2006; 7(3): 241–246
https://doi.org/10.1038/ni1317 pmid: 16482172
33 Kyurkchiev D, Ivanova-Todorova E, Hayrabedyan S, Altankova I, Kyurkchiev S. Female sex steroid hormones modify some regulatory properties of monocyte-derived dendritic cells. Am J Reprod Immunol 2007; 58(5): 425–433
https://doi.org/10.1111/j.1600-0897.2007.00526.x pmid: 17922695
34 Ivanova E, Kyurkchiev D, Altankova I, Dimitrov J, Binakova E, Kyurkchiev S. CD83 monocyte-derived dendritic cells are present in human decidua and progesterone induces their differentiation in vitro. Am J Reprod Immunol 2005; 53(4): 199–205
https://doi.org/10.1111/j.1600-0897.2005.00266.x pmid: 15760381
35 Hashino M, Tachibana M, Nishida T, Hara H, Tsuchiya K, Mitsuyama M, Watanabe K, Shimizu T, Watarai M. Inactivation of the MAPK signaling pathway by Listeria monocytogenes infection promotes trophoblast giant cell death. Front Microbiol 2015; 6: 1145
https://doi.org/10.3389/fmicb.2015.01145 pmid: 26528279
36 Wang S, Cao C, Piao H, Li Y, Tao Y, Zhang X, Zhang D, Sun C, Zhu R, Wang Y, Yuan M, Li D, Du M. Tim-3 protects decidual stromal cells from toll-like receptor-mediated apoptosis and inflammatory reactions and promotes Th2 bias at the maternal-fetal interface. Sci Rep 2015; 5(1): 9013
https://doi.org/10.1038/srep09013 pmid: 25757669
37 Hu WT, Li MQ, Liu W, Jin LP, Li DJ, Zhu XY. IL-33 enhances proliferation and invasiveness of decidual stromal cells by up-regulation of CCL2/CCR2 via NF-kB and ERK1/2 signaling. Mol Hum Reprod 2014; 20(4): 358–372
https://doi.org/10.1093/molehr/gat094 pmid: 24344240
38 Zhu R, Huang YH, Tao Y, Wang SC, Sun Ch, Piao HL, Wang XQ, Du MR, Li DJ. Hyaluronan up-regulates growth and invasion of trophoblasts in an autocrine manner via PI3K/AKT and MAPK/ERK1/2 pathways in early human pregnancy. Placenta 2013; 34(9): 784–791
https://doi.org/10.1016/j.placenta.2013.05.009 pmid: 23806178
39 Xie KM, Hou XF, Li MQ, Li DJ. NME1 at the human maternal-fetal interface downregulates titin expression and invasiveness of trophoblast cells via MAPK pathway in early pregnancy. Reproduction 2010; 139(4): 799–808
https://doi.org/10.1530/REP-09-0490 pmid: 20145075
40 Laird SM, Tuckerman EM, Cork BA, Linjawi S, Blakemore AI, Li TC. A review of immune cells and molecules in women with recurrent miscarriage. Hum Reprod Update 2003; 9(2): 163–174
https://doi.org/10.1093/humupd/dmg013 pmid: 12751778
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed