Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Genetics and imaging to assess oocyte and preimplantation embryo health

C. M. Warner A D , J. A. Newmark A , M. Comiskey A , S. R. De Fazio A , D. M. O’Malley A , M. Rajadhyaksha B , D. J. Townsend B , S. McKnight B , B. Roysam C , P. J. Dwyer B and C. A. DiMarzio B
+ Author Affiliations
- Author Affiliations

A Department of Biology, Northeastern University, Boston, MA 02115, USA.

B Department of Electrical and Computer Engineering, Northeastern University, Boston, MA 02115, USA.

C Department of Electrical, Computer, and Systems Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.

D To whom correspondence should be addressed. email: c.warner@neu.edu

Reproduction, Fertility and Development 16(7) 729-741 https://doi.org/10.1071/RD04088
Submitted: 10 August 2004  Accepted: 19 October 2004   Published: 9 December 2004

Abstract

Two major criteria are currently used in human assisted reproductive technologies (ART) to evaluate oocyte and preimplantation embryo health: (1) rate of preimplantation embryonic development; and (2) overall morphology. A major gene that regulates the rate of preimplantation development is the preimplantation embryo development (Ped) gene, discovered in our laboratory. In mice, presence of the Ped gene product, Qa-2 protein, results in a fast rate of preimplantation embryonic development, compared with a slow rate of preimplantation embryonic development for embryos that are lacking Qa-2 protein. Moreover, mice that express Qa-2 protein have an overall reproductive advantage that extends beyond the preimplantation period, including higher survival to birth, higher birthweight, and higher survival to weaning. Data are presented that suggest that Qa-2 increases the rate of development of early embryos by acting as a cell-signalling molecule and that phosphatidylinositol-3′ kinase is involved in the cell-signalling pathway. The most likely human homologue of Qa-2 has recently been identified as human leukocyte antigen (HLA)-G. Data are presented which show that HLA-G, like Qa-2, is located in lipid rafts, implying that HLA-G also acts as a signalling molecule. In order to better evaluate the second criterion used in ART (i.e. overall morphology), a unique and innovative imaging microscope has been constructed, the Keck 3-D fusion microscope (Keck 3DFM). The Keck 3DFM combines five different microscopic modes into a single platform, allowing multi-modal imaging of the specimen. One of the modes, the quadrature tomographic microscope (QTM), creates digital images of non-stained transparent cells by measuring changes in the index of refraction. Quadrature tomographic microscope images of oocytes and preimplantation mouse embryos are presented for the first time. The digital information from the QTM images should allow the number of cells in a preimplantation embryo to be counted non-invasively. The Keck 3DFM is also being used to assess mitochondrial distribution in mouse oocytes and embryos by using the k-means clustering algorithm. Both the number of cells in preimplantation embryos and mitochondrial distribution are related to oocyte and embryo health. New imaging data obtained from the Keck 3DFM, combined with genetic and biochemical approaches, have the promise of being able to distinguish healthy from unhealthy oocytes and embryos in a non-invasive manner. The goal is to apply the information from our mouse model system to the clinic in order to identify one and only one healthy embryo for transfer back to the mother undergoing an ART procedure. This approach has the potential to increase the success rate of ART and to decrease the high, and undesirable, multiple birth rate presently associated with ART.

Extra keywords: human leukocyte antigen-G, k-means clustering algorithm, mitochondria, Ped gene.


References

Barker, D. J. (1994). Maternal and fetal origins of coronary heart disease. J. R. Coll. Physicians Lond. 28, 544–551.
PubMed | Duda R. O., Hart P. E., and Stork D. G. (2001). ‘Pattern Classification.’ (Wiley-Interscience: New York, USA.)

Dykstra, M. , Cherukuri, A. , Sohn, H. W. , Tzeng, S. J. , and Pierce, S. K. (2003). Location is everything: lipid rafts and immune cell signaling. Annu. Rev. Immunol. 21, 457–481.
Crossref | GoogleScholarGoogle Scholar | PubMed | Squirrell J. M. (2002). Multiphoton microscopy for imaging mammalian embryos. In ‘Assessment of Mammalian Embryo Quality: Invasive and Non-invasive Techniques’. (Eds A. Van Soom and M. L. Boerjan.) pp. 195–217. (Kluwer Academic Publishers: Boston, MA.)

Squirrell, J. M. , Schramm, R. D. , Paprocki, A. M. , Wokosin, D. L. , and Bavister, B. D. (2003). Imaging mitochondrial organization in living primate oocytes and embryos using multiphoton microscopy. Microsc. Microanal. 9, 190–201.
Crossref | GoogleScholarGoogle Scholar | PubMed | Sutcliffe A. G. (2002). ‘IVF Children: The First Generation.’ (Parthenon Publishing: New York.)

Tesarik, J. , and Greco, E. (1999). The probability of abnormal preimplantation development can be predicted by a single static observation on pronuclear stage morphology. Hum. Reprod. 14, 1318–1323.
Crossref | GoogleScholarGoogle Scholar | PubMed | Verbanac K. M., and Warner C. M. (1981). Role of the major histocompatibility complex in the timing of early mammalian development. In ‘Cellular and Molecular Aspects of Implantation’. (Eds S. R. Glasser and D. W. Bullock.) pp. 467–470. (Plenum Publishers: New York, NY, USA.)

Visser, D. S. , and Fourie, F. R. (1993). The applicability of cumulative embryo score system for embryo selection and quality control in an in-vitro fertilization/embryo transfer programme. Hum. Reprod. 8, 1719–1722.
PubMed | Warner C. M., and Brenner C. A. (2001). Genetic regulation of preimplantation embryo survival. In ‘Current Topics in Developmental Biology. Vol. 52’. (Ed. G. P. Schatten.) pp. 151–192. (Academic Press: San Diego, CA, USA.)

Warner, C. M. , Brownell, M. S. , and Rothschild, M. F. (1991). Analysis of litter size and weight in mice differing in Ped gene phenotype and the Q region of the H-2 complex. J. Reprod. Immunol. 19, 303–313.
Crossref | GoogleScholarGoogle Scholar | PubMed |

Warner, C. M. , Panda, P. , Almquist, C. D. , and Xu, Y. (1993). Preferential survival of mice expressing the Qa-2 antigen. J. Reprod. Fertil. 99, 145–147.
Crossref | GoogleScholarGoogle Scholar | PubMed |

Wennerholm, U. B. , Bergh, C. , Hamberger, L. , Lundin, K. , Nilsson, L. , Wikland, M. , and Kallen, B. (2000). Incidence of congenital malformations in children born after ICSI. Hum. Reprod. 15, 944–948.
Crossref | GoogleScholarGoogle Scholar | PubMed |

Wharf, E. , Dimitrakopoulos, A. , Khalaf, Y. , and Pickering, S. (2004). Early embryo development is an indicator of implantation potential. Reprod. Biomed. Online 8, 212–218.
PubMed |

Wilding, M. , Dale, B. , Marino, M. , di Matteo, L. , Alviggi, C. , Pisaturo, M. L. , Lombardi, L. , and De Placido, G. (2001). Mitochondrial aggregation patterns and activity in human oocytes and preimplantation embryos. Hum. Reprod. 16, 909–917.
Crossref | GoogleScholarGoogle Scholar | PubMed |

Wittemer, C. , Bettahar-Lebugle, K. , Ohl, J. , Rongieres, C. , Nisand, I. , and Gerlinger, P. (2000). Zygote evaluation: an efficient tool for embryo selection. Hum. Reprod. 15, 2591–2597.
Crossref | GoogleScholarGoogle Scholar | PubMed |

Young, L. E. , Sinclair, K. D. , and Wilmut, I. (1998). Large offspring syndrome in cattle and sheep. Rev. Reprod. 3, 155–163.
Crossref | GoogleScholarGoogle Scholar | PubMed |

Ziebe, S. , Petersen, K. , Lindenberg, S. , Andersen, A. G. , Gabrielsen, A. , and Andersen, A. N. (1997). Embryo morphology or cleavage stage: how to select the best embryos for transfer after in-vitro fertilization. Hum. Reprod. 12, 1545–1549.
Crossref | GoogleScholarGoogle Scholar | PubMed |