Mosaicism between trophectoderm and inner cell mass Antonio Capalbo, Ph.D.a,b and Laura Rienzi, M.Sc.a,b a GENERA, Centre for Reproductive Medicine, Clinica Valle Giulia, Rome; and b GENETYX, Molecular Genetics Laboratory, Vicenza, Italy

Defining the actual incidence and prevalence of mosaicism in human blastocysts still remains a difficult task. The small amount of evidence generated by animal and human studies does not support the existence of mechanisms involved in developmental arrest, clonal depletion, or aneuploidy rescue for abnormal cells in euploid/aneuploid embryos during preimplantation development. However, studies in humans are mainly descriptive and lack functional evidence. Understanding the biological mechanisms that beset preimplantation differentiation holds the potential to reveal the role of aneuploidies and gene dosage imbalances in cell fate decision, providing important clues on the origin and evolution of embryonic mosaicism. The evidence on human blastocysts suggests that a mosaic euploid/aneuploid configuration is detected in around 5% of embryos. This figure supports the extremely low level of mosaicism reported in natural and IVF pregnancies. Similarly, the clinical management of patterns consistent with the presence of mosaicism in a trophectoderm biopsy during preimplantation genetic diagnosis cycles (PGD-A) is still a controversial issue. Despite the facts that some contemporary comprehensive chromosomal screening platforms can detect mosaic samples in cell mixture models with variable accuracy and many reproductive genetics laboratories are now routinely including embryonic mosaicism on their genetic reports, a diagnosis of certainty for mosaicism in PGD-A cycles is conceptually impracticable. Indeed, several technical and biological sources of errors clearly exist when trying to estimate mosaicism from a single trophectoderm biopsy in PGD-A cycles and must be understood to adequately guide patients during clinical care. (Fertil SterilÒ 2017;107:1098–106. Ó2017 by American Society for Reproductive Medicine.) Key Words: Chromosomal mosaicism, blastocyst, inner cell mass, trophectoderm, aneuploidies, preimplantation genetic screening Discuss: You can discuss this article with its authors and with other ASRM members at https://www.fertstertdialog.com/users/ 16110-fertility-and-sterility/posts/15693-23856

T

he unbalanced transmission of chromosomes in human gametes and early preimplantation embryos causes aneuploidy, which is a major cause of infertility, pregnancy loss, and intellectual disability in humans (1). In the preimplantation and prenatal setting, chromosome abnormalities span a wide range of genomic imbalances, from polyploidy, to whole chromosome and large structural aneuploidies, down to submicroscopic deletions and duplications. Whole chromosome aneuploidies, monosomies and trisomies for the entire chromosomes, are the far more prevalent abnormalities and have been extensively investigated due to their high incidence in human conceptions and their clear association with clinical phenotypes and infertility. Undoubtedly, they represent

the single most common form of aneuploidy and the primary cause for implantation failure in IVF cycles and miscarriages in human pregnancies. Another well-defined characteristic of human aneuploidies is their strict correlation with female age. As women age, oocytes become increasingly susceptible to chromosome segregation errors during the meiotic process. Extensive analysis of main autosomal trisomies in clinical pregnancies revealed that more than 90% had a meiotic origin. The majority are due to maternal errors, with >75% due to errors in meiosis I and 50%) have full developmental potential, while when the proportion of normal blastomeres was decreased to one third, there was a partial rescue of embryonic lethality. In humans, both functional and prospective nonselection studies aimed at modeling the clinical fate and the reproductive potential of embryos showing intermediate copy number profiles are lacking. Prospective cohort studies in PGD-A cycles are also of poor value to determine the predictive value of patterns consistent with mosaicism as these are subjected to a patient and embryo selection bias. In conclusion, despite the fact that some contemporary CCS platforms can detect mosaic samples in cell mixture models with variable accuracy (59–61), a diagnosis of certainty for mosaicism in PGD-A cycles is conceptually impracticable. Many reproductive genetics laboratories are now routinely including embryonic mosaicism on their diagnostic reports. However, for the arguments discussed above, both biological and technological limitations exist and must be understood in order to adequately counsel patients during clinical care. Furthermore, it should be stressed that the chromosomal variations observed in a TE biopsy will not unequivocally represent the mosaicism rate present within the whole embryo. The existence of a sample bias has to be acknowledged in patients consent forms as a limitation for mosaicism detection in PGD-A cycles. In the light of the arguments discussed above, mosaicism should be recognized as a biological limitation for PGD-A programs as it is for prenatal diagnosis. In addition, it appears reasonable to avoid reporting mosaicism in PGD-A cycles

1103

1104

until its impact on clinical treatment has been clarified. A more conservative approach for reporting this type of result may be therefore preferred until evidence showing correlation between intermediate copy number profiles and mosaicism in embryos is available (i.e., ‘‘analysis showing a pattern that is consistent with the potential presence of mosaicism’’). Similarly, if future nonselection studies and randomized controlled trials show a lower reproductive potential for blastocyst with intermediate copy number profiles, clinically validated criteria should be introduced into clinical practice. In this case, on top of their genetic diagnosis, additional viability scores could be assigned to embryos. Nonetheless, extensive genetic counseling detailing all the specific limitations should be given to patients.

CONCLUDING REMARKS AND FUTURE PERSPECTIVES

Capalbo. Blastocyst mosaicism. Fertil Steril 2017.

92/181 (51.7) 77/181 (42.8) 41/51 (80) 9/51 (18) 4/10 (40) 6/10 (60) 29/50 (58) 13/50 (26)

Note: Abnormal cells in euploid-aneuploid mosaic embryos are similarly distributed between ICM and TE, indicating no sign of preferential allocation or confinement of chromosomally abnormal cells. The fact that results were almost identical for samples from the TE and ICM indicates that data obtained from a clinical TE sample can generally be considered diagnostic of the ICM chromosomal complement. CGH ¼ comparative genomic hybridization.

– – Not applicable

TE-confined mosaicism (%) ICM-confined mosaicism (%) Mosaic blastocysts (both ICM and TE) (%) Most frequent mosaic chromosomes Fully euploid blastocyst (%) Fully aneuploid blastocyst (%)

chr16; chr22

19/70 (70) 49/70 (27)

4/181 (2.2) 2/181 (1.1) 5/181 (2.8) 0/51 (0) 1/51 (2) 0/51 (0)

chr16; chr17; chr22; chr21

– 181 71 Range, 26–42 – SNP array with parental support 51 17 31 Donated blastocysts

FISH þ CGH reanalysis 10 4 33 Blastocysts diagnosed as aneuploid by FISH-based blastomere analysis 0/10 (0) 0/10 (0) 0/10 (0) Method No. of samples No. of patients Mean female age Origin of embryos

FISH þ SNP array reanalysis 50 24 35 Blastocysts diagnosed as aneuploid by FISH-based blastomere analysis 4/50 (8) 1/50 (2) 3/50 (6)

aCGH þ FISH reanalysis 70 26 36 Blastocysts diagnosed as aneuploid by aCGH-based TE analysis 0/70 (0) 0/70 (0) 2/70 (3)

Overall Johnson et al. (2010) Capalbo et al. (2013) Fragouli et al. (2008) Northrop et al. (2010)

Summary table of methodology and main findings from relevant studies on the cytogenetic constitution of ICM and TE samples from disaggregated human blastocysts.

TABLE 1

VIEWS AND REVIEWS

The small amount of evidence gathered from animal and human studies suggests the absence of mechanisms involved in embryo arrest, clonal depletion, or aneuploidy rescue for abnormal cells in euploid/aneuploid embryos during preimplantation development and blastocyst differentiation. Thus, it is likely that abnormal cells in mosaic diploid/aneuploid blastocysts are randomly allocated to the ICM and TE. However, our understanding of human preimplantation development is limited due to ethical and legal restrictions on embryo research and scarcity of materials. Studies in humans are mainly descriptive and lack functional evidence. Most of the information on embryo development is inferred from animal models and embryonic stem cell cultures, which should therefore be applied to the human model with caution. Understanding the biological mechanisms that beset preimplantation differentiation might reveal how aneuploidies and related gene dosage imbalances impact cell fate decisions and provide important clues on the biology of mosaicism in preimplantation embryos. The lack of correction for expected false positives and the use of nonstandardized criteria for classification of embryos as mosaic have likely resulted in an overestimation of mosaicism in IVF embryos. The little evidence available on humans suggests that mosaic euploid/aneuploid configuration is a relatively uncommon feature and should not impact the effectiveness of PGD-A programs appreciably. Further studies are needed to corroborate these findings and to better characterize at the single-cell level the incidence and prevalence of mosaicism at the blastocyst stage. Undoubtedly, novel CCS technologies able to distinguish between meiotic and mitotic errors in embryonic biopsies will deliver powerful research and clinical tools for the comprehensive analysis of aneuploidies in human blastocyst and consequent PGD-A protocols improvement. At the present, reports of mosaicism should be avoided in blastocyst PGD-A cycles due to the several technical and biological issues present when attempting this type of diagnosis and the lack of evidence from nonselection studies. The observation of intermediate chromosome copy number profiles would be better reported as a ‘‘pattern consistent with the presence of mosaicism,’’ and genetic counseling should be provided to guide patients' decisions. VOL. 107 NO. 5 / MAY 2017

Fertility and Sterility®

REFERENCES 1. 2. 3.

4.

5. 6.

7.

8. 9.

10.

11.

12.

13.

14.

15. 16. 17. 18.

19.

20.

21.

22.

23.

Hassold T, Hunt P. To err (meiotically) is human: the genesis of human aneuploidy. Nat Rev Genet 2001;2:280–91. Chiang T, Schultz RM, Lampson MA. Meiotic origins of maternal age-related aneuploidy. Biol Reprod 2012;86:1–7. Ottolini CS, Newnham LJ, Capalbo A, Natesan SA, Joshi HA, Cimadomo D, et al. Genome-wide maps of recombination and chromosome segregation in human oocytes and embryos show selection for maternal recombination rates. Nat Genet 2015;47:727–35. Franasiak JM, Olcha M, Shastri S, Molinaro TA, Congdon H, Treff NR, et al. Embryonic aneuploidy does not differ among genetic ancestry according to continental origin as determined by ancestry informative markers. Hum Reprod 2016;31:2391–5. Vermeesch JR, Voet T, Devriendt K. Prenatal and pre-implantation genetic diagnosis. Nat Rev Genet 2016;17:643–56. Chen M, Wei S, Hu J, Quan S. Can comprehensive chromosome screening technology improve IVF/ICSI outcomes? A meta-analysis. PLoS One 2015; 10:e0140779. McCoy RC, Demko ZP, Ryan A, Banjevic M, Hill M, Sigurjonsson S, et al. Evidence of selection against complex mitotic-origin aneuploidy during preimplantation development. PLoS Genet 2015;11:e1005601. Cohen J. Sorting out chromosome errors. Science 2002;296:2164–6. Johnson DS, Cinnioglu C, Ross R, Filby A, Gemelos G, Hill M, et al. Comprehensive analysis of karyotypic mosaicism between trophectoderm and inner cell mass. Mol Hum Reprod 2010;16:944–9. Wapner RJ, Simpson JL, Golbus MS, Zachary JM, Ledbetter DH, Desnick RJ, et al. Chorionic mosaicism: association with fetal loss but not with adverse perinatal outcome. Prenat Diagn 1992;12:347–55. Wilkins-Haug L, Roberts DJ, Morton CC. Confined placental mosaicism and intrauterine growth retardation: a case-control analysis of placentas at delivery. Am J Obstet Gynecol 1995;172:44–50. Malvestiti F, Agrati C, Grimi B, Pompilii E, Izzi C, Martinoni L, et al. Interpreting mosaicism in chorionic villi: results of a monocentric series of 1001 mosaics in chorionic villi with follow-up amniocentesis. Prenat Diagn 2015;35: 1117–27. Taylor TH, Gitlin SA, Patrick JL, Crain JL, Wilson JM, Griffin DK. The origin, mechanisms, incidence and clinical consequences of chromosomal mosaicism in humans. Hum Reprod Update 2014;20:571–81. Capalbo A, Ubaldi FM, Rienzi L, Scott R, Treff N. Detecting mosaicism in trophectoderm biopsies: current challenges and future possibilities. Hum Reprod 2017;32:492–8. Munne S, Grifo J, Wells D. Mosaicism: ‘‘Survival of the fittest’’ versus ‘‘no embryo left behind’’. Fertil Steril 2016;105:1146–9. Scott RT Jr, Galliano D. The challenge of embryonic mosaicism in preimplantation genetic screening. Fertil Steril 2016;105:1150–2. Mastenbroek S, Repping S. Preimplantation genetic screening: back to the future. Hum Reprod 2014;29:1846–50. Braude P, Bolton V, Moore S. Human gene expression first occurs between the four- and eight-cell stages of preimplantation development. Nature 1988;332:459–61. Petropoulos S, Edsgard D, Reinius B, Deng Q, Panula SP, Codeluppi S, et al. Single-cell RNA-Seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 2016;167:285. van Echten-Arends J, Mastenbroek S, Sikkema-Raddatz B, Korevaar JC, Heineman MJ, van der Veen F, et al. Chromosomal mosaicism in human preimplantation embryos: a systematic review. Hum Reprod Update 2011;17: 620–7. Zamani Esteki M, Dimitriadou E, Mateiu L, Melotte C, Van der Aa N, Kumar P, et al. Concurrent whole-genome haplotyping and copy-number profiling of single cells. Am J Hum Genet 2015;96:894–912. Bazrgar M, Gourabi H, Valojerdi MR, Yazdi PE, Baharvand H. Self-correction of chromosomal abnormalities in human preimplantation embryos and embryonic stem cells. Stem Cells Dev 2013;22:2449–56. Mertzanidou A, Spits C, Nguyen HT, Van de Velde H, Sermon K. Evolution of aneuploidy up to day 4 of human preimplantation development. Hum Reprod 2013;28:1716–24.

VOL. 107 NO. 5 / MAY 2017

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34. 35. 36.

37. 38.

39.

40.

41.

42.

43. 44.

45.

Kort DH, Chia G, Treff NR, Tanaka AJ, Xing T, Vensand LB, et al. Human embryos commonly form abnormal nuclei during development: a mechanism of DNA damage, embryonic aneuploidy, and developmental arrest. Hum Reprod 2016;31:312–23. Vega M, Breborowicz A, Moshier EL, McGovern PG, Keltz MD. Blastulation rates decline in a linear fashion from euploid to aneuploid embryos with single versus multiple chromosomal errors. Fertil Steril 2014;102:394–8. Bisignano A, Wells D, Harton G, Munne S. PGD and aneuploidy screening for 24 chromosomes: advantages and disadvantages of competing platforms. Reprod Biomed Online 2011;23:677–85. Handyside AH. PGD and aneuploidy screening for 24 chromosomes by genome-wide SNP analysis: seeing the wood and the trees. Reprod Biomed Online 2011;23:686–91. Capalbo A, Rienzi L, Cimadomo D, Maggiulli R, Elliott T, Wright G, et al. Correlation between standard blastocyst morphology, euploidy and implantation: an observational study in two centers involving 956 screened blastocysts. Hum Reprod 2014;29:1173–81. Franasiak JM, Forman EJ, Hong KH, Werner MD, Upham KM, Treff NR, et al. The nature of aneuploidy with increasing age of the female partner: a review of 15,169 consecutive trophectoderm biopsies evaluated with comprehensive chromosomal screening. Fertil Steril 2014;101:656–63.e1. Rienzi L, Capalbo A, Stoppa M, Romano S, Maggiulli R, Albricci L, et al. No evidence of association between blastocyst aneuploidy and morphokinetic assessment in a selected population of poor-prognosis patients: a longitudinal cohort study. Reprod Biomed Online 2015;30:57–66. Carbone L, Chavez SL. Mammalian pre-implantation chromosomal instability: species comparison, evolutionary considerations, and pathological correlations. Syst Biol Reprod Med 2015;61:321–35. Bedzhov I, Graham SJ, Leung CY, Zernicka-Goetz M. Developmental plasticity, cell fate specification and morphogenesis in the early mouse embryo. Philos Trans R Soc Lond B Biol Sci 2014:369. Morris SA, Teo RT, Li H, Robson P, Glover DM, Zernicka-Goetz M. Origin and formation of the first two distinct cell types of the inner cell mass in the mouse embryo. Proc Natl Acad Sci U S A 2010;107:6364–9. Fleming TP. A quantitative analysis of cell allocation to trophectoderm and inner cell mass in the mouse blastocyst. Dev Biol 1987;119:520–31. Zernicka-Goetz M, Huang S. Stochasticity versus determinism in development: a false dichotomy? Nat Rev Genet 2010;11:743–4. Biggins JS, Royer C, Watanabe T, Srinivas S. Towards understanding the roles of position and geometry on cell fate decisions during preimplantation development. Semin Cell Dev Biol 2015;47–48:74–9. Ajduk A, Zernicka-Goetz M. Polarity and cell division orientation in the cleavage embryo: from worm to human. Mol Hum Reprod 2016;10:691–703. Hirate Y, Hirahara S, Inoue K, Suzuki A, Alarcon VB, Akimoto K, et al. Polarity-dependent distribution of angiomotin localizes Hippo signaling in preimplantation embryos. Curr Biol 2013;23:1181–94. Leung CY, Zernicka-Goetz M. Angiomotin prevents pluripotent lineage differentiation in mouse embryos via Hippo pathway-dependent and -independent mechanisms. Nat Commun 2013;4:2251. Nishioka N, Inoue K, Adachi K, Kiyonari H, Ota M, Ralston A, et al. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell 2009;16:398–410. Morin-Kensicki EM, Boone BN, Howell M, Stonebraker JR, Teed J, Alb JG, et al. Defects in yolk sac vasculogenesis, chorioallantoic fusion, and embryonic axis elongation in mice with targeted disruption of Yap65. Mol Cell Biol 2006;26:77–87. Makita R, Uchijima Y, Nishiyama K, Amano T, Chen Q, Takeuchi T, et al. Multiple renal cysts, urinary concentration defects, and pulmonary emphysematous changes in mice lacking TAZ. Am J Physiol Renal Physiol 2008;294:F542–53. Chazaud C, Rossant J. Disruption of early proximodistal patterning and AVE formation in Apc mutants. Development 2006;133:3379–87. Wicklow E, Blij S, Frum T, Hirate Y, Lang RA, Sasaki H, et al. HIPPO pathway members restrict SOX2 to the inner cell mass where it promotes ICM fates in the mouse blastocyst. PLoS Genet 2014;10:e1004618. Nichols J, Zevnik B, Anastassiadis K, Niwa H, Klewe-Nebenius D, Chambers I, et al. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 1998;95:379–91.

1105

VIEWS AND REVIEWS 46.

47.

48.

49.

50. 51.

52.

53.

Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, et al. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 2003;113:631–42. Nishioka N, Yamamoto S, Kiyonari H, Sato H, Sawada A, Ota M, et al. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos. Mech Dev 2008;125:270–83. Kang M, Piliszek A, Artus J, Hadjantonakis AK. FGF4 is required for lineage restriction and salt-and-pepper distribution of primitive endoderm factors but not their initial expression in the mouse. Development 2013;140:267–79. Wong CC, Loewke KE, Bossert NL, Behr B, De Jonge CJ, Baer TM, et al. Noninvasive imaging of human embryos before embryonic genome activation predicts development to the blastocyst stage. Nat Biotechnol 2010;28:1115–21. Kiessling AA. Timing is everything in the human embryo. Nat Biotechnol 2010;28:1025–6. Bolton H, Graham SJ, Van der Aa N, Kumar P, Theunis K, Fernandez Gallardo E, et al. Mouse model of chromosome mosaicism reveals lineage-specific depletion of aneuploid cells and normal developmental potential. Nat Commun 2016;7:11165. Capalbo A, Wright G, Elliott T, Ubaldi FM, Rienzi L, Nagy ZP. FISH reanalysis of inner cell mass and trophectoderm samples of previously array-CGH screened blastocysts shows high accuracy of diagnosis and no major diagnostic impact of mosaicism at the blastocyst stage. Hum Reprod 2013;28:2298–307. Gueye NA, Devkota B, Taylor D, Pfundt R, Scott RT Jr, Treff NR. Uniparental disomy in the human blastocyst is exceedingly rare. Fertil Steril 2014;101: 232–6.

1106

54.

55.

56.

57.

58.

59. 60.

61.

Van der Aa N, Cheng J, Mateiu L, Zamani Esteki M, Kumar P, Dimitriadou E, et al. Genome-wide copy number profiling of single cells in S-phase reveals DNA-replication domains. Nucleic Acids Res 2013;41:e66. Treff NR, Franasiak JM. Detection of segmental aneuploidy and mosaicism in the human preimplantation embryo: technical considerations and limitations. Fertil Steril 2017;107:27–31. Fragouli E, Lenzi M, Ross R, Katz-Jaffe M, Schoolcraft WB, Wells D. Comprehensive molecular cytogenetic analysis of the human blastocyst stage. Hum Reprod 2008;23:2596–608. Northrop LE, Treff NR, Levy B, Scott RT Jr. SNP microarray-based 24 chromosome aneuploidy screening demonstrates that cleavage-stage FISH poorly predicts aneuploidy in embryos that develop to morphologically normal blastocysts. Mol Hum Reprod 2010;16:590–600. Huang A, Adusumalli J, Patel S, Liem J, Williams J 3rd, Pisarska MD. Prevalence of chromosomal mosaicism in pregnancies from couples with infertility. Fertil Steril 2009;91:2355–60. Greco E, Minasi MG, Fiorentino F. Healthy babies after intrauterine transfer of mosaic aneuploid blastocysts. N Engl J Med 2015;373:2089–90. Mamas T, Gordon A, Brown A, Harper J, Sengupta S. Detection of aneuploidy by array comparative genomic hybridization using cell lines to mimic a mosaic trophectoderm biopsy. Fertil Steril 2012;97:943–7. Goodrich D, Tao X, Bohrer C, Lonczak A, Xing T, Zimmerman R, et al. A randomized and blinded comparison of qPCR and NGS-based detection of aneuploidy in a cell line mixture model of blastocyst biopsy mosaicism. J Assist Reprod Genet 2016;33:1473–80.

VOL. 107 NO. 5 / MAY 2017

Mosaicism between trophectoderm and inner cell mass.

Defining the actual incidence and prevalence of mosaicism in human blastocysts still remains a difficult task. The small amount of evidence generated ...
440KB Sizes 1 Downloads 14 Views