Semin Reprod Med 2015; 33(06): 410-421
DOI: 10.1055/s-0035-1567826
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Autologous Germline Mitochondrial Energy Transfer (AUGMENT) in Human Assisted Reproduction

Dori C. Woods
1   Laboratory of Aging and Infertility Research (LAIR), Department of Biology, Northeastern University, Boston, Massachusetts
,
Jonathan L. Tilly
1   Laboratory of Aging and Infertility Research (LAIR), Department of Biology, Northeastern University, Boston, Massachusetts
2   Office of the Dean, College of Science, Northeastern University, Boston, Massachusetts
› Institutsangaben
Weitere Informationen

Publikationsverlauf

Publikationsdatum:
17. November 2015 (online)

Abstract

Ovarian aging is characterized by a decline in both the total number and overall quality of oocytes, the latter of which has been experimentally tied to mitochondrial dysfunction. Clinical studies in the late 1990s demonstrated that transfer of cytoplasm aspirated from eggs of young female donors into eggs of infertile women at the time of intracytoplasmic sperm injection improved pregnancy success rates. However, donor mitochondria were identified in offspring, and the United States Food and Drug Administration raised questions about delivery of foreign genetic material into human eggs at the time of fertilization. Accordingly, heterologous cytoplasmic transfer, while promising, was in effect shut down as a clinical protocol. The recent discovery of adult oogonial (oocyte-generating) stem cells in mice, and subsequently in women, has since re-opened the prospects of delivering a rich source of pristine and patient-matched germline mitochondria to boost egg health and embryonic developmental potential without the need for young donor eggs to obtain cytoplasm. Herein we overview the science behind this new protocol, which has been patented and termed autologous germline mitochondrial energy transfer, and its use to date in clinical studies for improving pregnancy success in women with a prior history of assisted reproduction failure.

Note

During the final preparation of this article, additional key studies of central importance to verifying the ability of isolated mouse OSCs to generate fertilization competent eggs and viable offspring,[97] and to identifying the existence of OSCs in adult baboon ovaries,[98] were published; additionally, a third clinical site reporting its experience with AUGMENT in human assisted reproduction was published.[99]


 
  • References

  • 1 Zuckerman S. The number of oocytes in the mature ovary. Recent Prog Horm Res 1951; 6: 63-108
  • 2 Navot D, Bergh PA, Williams MA , et al. Poor oocyte quality rather than implantation failure as a cause of age-related decline in female fertility. Lancet 1991; 337 (8754) 1375-1377
  • 3 Broekmans FJ, Soules MR, Fauser BC. Ovarian aging: mechanisms and clinical consequences. Endocr Rev 2009; 30 (5) 465-493
  • 4 Johnson J, Canning J, Kaneko T, Pru JK, Tilly JL. Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature 2004; 428 (6979) 145-150
  • 5 Zou K, Yuan Z, Yang Z , et al. Production of offspring from a germline stem cell line derived from neonatal ovaries. Nat Cell Biol 2009; 11 (5) 631-636
  • 6 Pacchiarotti J, Maki C, Ramos T , et al. Differentiation potential of germ line stem cells derived from the postnatal mouse ovary. Differentiation 2010; 79 (3) 159-170
  • 7 Zhang Y, Yang Z, Yang Y , et al. Production of transgenic mice by random recombination of targeted genes in female germline stem cells. J Mol Cell Biol 2011; 3 (2) 132-141
  • 8 Zou K, Hou L, Sun K, Xie W, Wu J. Improved efficiency of female germline stem cell purification using fragilis-based magnetic bead sorting. Stem Cells Dev 2011; 20 (12) 2197-2204
  • 9 White YAR, Woods DC, Takai Y, Ishihara O, Seki H, Tilly JL. Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nat Med 2012; 18 (3) 413-421
  • 10 Woods DC, Tilly JL. An evolutionary perspective on adult female germline stem cell function from flies to humans. Semin Reprod Med 2013; 31 (1) 24-32
  • 11 Woods DC, Tilly JL. Isolation, characterization and propagation of mitotically active germ cells from adult mouse and human ovaries. Nat Protoc 2013; 8 (5) 966-988
  • 12 Imudia AN, Wang N, Tanaka Y, White YA, Woods DC, Tilly JL. Comparative gene expression profiling of adult mouse ovary-derived oogonial stem cells supports a distinct cellular identity. Fertil Steril 2013; 100 (5) 1451-1458
  • 13 Park ES, Woods DC, Tilly JL. Bone morphogenetic protein 4 promotes mammalian oogonial stem cell differentiation via Smad1/5/8 signaling. Fertil Steril 2013; 100 (5) 1468-1475
  • 14 Zhou L, Wang L, Kang JX , et al. Production of fat-1 transgenic rats using a post-natal female germline stem cell line. Mol Hum Reprod 2014; 20 (3) 271-281
  • 15 Xie W, Wang H, Wu J. Similar morphological and molecular signatures shared by female and male germline stem cells. Sci Rep 2014; 4: 5580
  • 16 Park ES, Tilly JL. Use of DEAD-box polypeptide-4 (Ddx4) gene promoter-driven fluorescent reporter mice to identify mitotically active germ cells in post-natal mouse ovaries. Mol Hum Reprod 2015; 21 (1) 58-65
  • 17 Khosravi-Farsani S, Amidi F, Habibi Roudkenar M, Sobhani A. Isolation and enrichment of mouse female germ line stem cells. Cell J 2015; 16 (4) 406-415
  • 18 Grieve KM, McLaughlin M, Dunlop CE, Telfer EE, Anderson RA. The controversial existence and functional potential of oogonial stem cells. Maturitas 2015;
  • 19 Hassold T, Chiu D. Maternal age-specific rates of numerical chromosome abnormalities with special reference to trisomy. Hum Genet 1985; 70 (1) 11-17
  • 20 Battaglia DE, Goodwin P, Klein NA, Soules MR. Influence of maternal age on meiotic spindle assembly in oocytes from naturally cycling women. Hum Reprod 1996; 11 (10) 2217-2222
  • 21 Van Blerkom J. Mitochondrial function in the human oocyte and embryo and their role in developmental competence. Mitochondrion 2011; 11 (5) 797-813
  • 22 Tilly JL, Sinclair DA. Germline energetics, aging, and female infertility. Cell Metab 2013; 17 (6) 838-850
  • 23 Tarín JJ, Pérez-Albalá S, Cano A. Oral antioxidants counteract the negative effects of female aging on oocyte quantity and quality in the mouse. Mol Reprod Dev 2002; 61 (3) 385-397
  • 24 Selesniemi K, Lee HJ, Muhlhauser A, Tilly JL. Prevention of maternal aging-associated oocyte aneuploidy and meiotic spindle defects in mice by dietary and genetic strategies. Proc Natl Acad Sci USA 2011; 108 (30) 12319-12324
  • 25 Selesniemi K, Lee HJ, Tilly JL. Moderate caloric restriction initiated in rodents during adulthood sustains function of the female reproductive axis into advanced chronological age. Aging Cell 2008; 7 (5) 622-629
  • 26 Ben-Meir A, Burstein E, Borrego-Alvarez A , et al. Coenzyme Q10 restores oocyte mitochondrial function and fertility during reproductive aging. Aging Cell 2015; 14 (5) 887-895
  • 27 Liu M, Yin Y, Ye X , et al. Resveratrol protects against age-associated infertility in mice. Hum Reprod 2013; 28 (3) 707-717
  • 28 Dumollard R, Duchen M, Carroll J. The role of mitochondrial function in the oocyte and embryo. Curr Top Dev Biol 2007; 77: 21-49
  • 29 Bentov Y, Esfandiari N, Burstein E, Casper RF. The use of mitochondrial nutrients to improve the outcome of infertility treatment in older patients. Fertil Steril 2010; 93 (1) 272-275
  • 30 Bentov Y, Yavorska T, Esfandiari N, Jurisicova A, Casper RF. The contribution of mitochondrial function to reproductive aging. J Assist Reprod Genet 2011; 28 (9) 773-783
  • 31 Van Blerkom J, Davis PW, Lee J. ATP content of human oocytes and developmental potential and outcome after in-vitro fertilization and embryo transfer. Hum Reprod 1995; 10 (2) 415-424
  • 32 Thouas GA, Trounson AO, Wolvetang EJ, Jones GM. Mitochondrial dysfunction in mouse oocytes results in preimplantation embryo arrest in vitro. Biol Reprod 2004; 71 (6) 1936-1942
  • 33 Wai T, Ao A, Zhang X, Cyr D, Dufort D, Shoubridge EA. The role of mitochondrial DNA copy number in mammalian fertility. Biol Reprod 2010; 83 (1) 52-62
  • 34 Cohen J, Scott R, Schimmel T, Levron J, Willadsen S. Birth of infant after transfer of anucleate donor oocyte cytoplasm into recipient eggs. Lancet 1997; 350 (9072) 186-187
  • 35 Cohen J, Scott R, Alikani M , et al. Ooplasmic transfer in mature human oocytes. Mol Hum Reprod 1998; 4 (3) 269-280
  • 36 Lanzendorf SE, Mayer JF, Toner J, Oehninger S, Saffan DS, Muasher S. Pregnancy following transfer of ooplasm from cryopreserved-thawed donor oocytes into recipient oocytes. Fertil Steril 1999; 71 (3) 575-577
  • 37 Huang CC, Cheng TC, Chang HH , et al. Birth after the injection of sperm and the cytoplasm of tripronucleate zygotes into metaphase II oocytes in patients with repeated implantation failure after assisted fertilization procedures. Fertil Steril 1999; 72 (4) 702-706
  • 38 Dale B, Wilding M, Botta G , et al. Pregnancy after cytoplasmic transfer in a couple suffering from idiopathic infertility: case report. Hum Reprod 2001; 16 (7) 1469-1472
  • 39 Barritt J, Willadsen S, Brenner C, Cohen J. Cytoplasmic transfer in assisted reproduction. Hum Reprod Update 2001; 7 (4) 428-435
  • 40 Tilly JL, Woods DC. Compositions and Methods for Autologous Germline Mitochondrial Energy Transfer. United States Patent Number 8,642,329. United States Patent and Trademark Office; 2014
  • 41 Tilly JL, Woods DC. Compositions and Methods for Autologous Germline Mitochondrial Energy Transfer. United States Patent Number 8,647,869. United States Patent and Trademark Office; 2014
  • 42 Reynier P, May-Panloup P, Chrétien MF , et al. Mitochondrial DNA content affects the fertilizability of human oocytes. Mol Hum Reprod 2001; 7 (5) 425-429
  • 43 May-Panloup P, Chrétien MF, Jacques C, Vasseur C, Malthièry Y, Reynier P. Low oocyte mitochondrial DNA content in ovarian insufficiency. Hum Reprod 2005; 20 (3) 593-597
  • 44 Santos TA, El Shourbagy S, St John JC. Mitochondrial content reflects oocyte variability and fertilization outcome. Fertil Steril 2006; 85 (3) 584-591
  • 45 Duran HE, Simsek-Duran F, Oehninger SC, Jones Jr HW, Castora FJ. The association of reproductive senescence with mitochondrial quantity, function, and DNA integrity in human oocytes at different stages of maturation. Fertil Steril 2011; 96 (2) 384-388
  • 46 Murakoshi Y, Sueoka K, Takahashi K , et al. Embryo developmental capability and pregnancy outcome are related to the mitochondrial DNA copy number and ooplasmic volume. J Assist Reprod Genet 2013; 30 (10) 1367-1375
  • 47 Simsek-Duran F, Li F, Ford W, Swanson RJ, Jones Jr HW, Castora FJ. Age-associated metabolic and morphologic changes in mitochondria of individual mouse and hamster oocytes. PLoS One 2013; 8 (5) e64955
  • 48 Kushnir VA, Ludaway T, Russ RB, Fields EJ, Koczor C, Lewis W. Reproductive aging is associated with decreased mitochondrial abundance and altered structure in murine oocytes. J Assist Reprod Genet 2012; 29 (7) 637-642
  • 49 Fragouli E, Spath K, Alfarawati S , et al. Altered levels of mitochondrial DNA are associated with female age, aneuploidy, and provide an independent measure of embryonic implantation potential. PLoS Genet 2015; 11 (6) e1005241
  • 50 Larsson NG, Wang J, Wilhelmsson H , et al. Mitochondrial transcription factor A is necessary for mtDNA maintenance and embryogenesis in mice. Nat Genet 1998; 18 (3) 231-236
  • 51 St John JC, Facucho-Oliveira J, Jiang Y, Kelly R, Salah R. Mitochondrial DNA transmission, replication and inheritance: a journey from the gamete through the embryo and into offspring and embryonic stem cells. Hum Reprod Update 2010; 16 (5) 488-509
  • 52 Muggleton-Harris A, Whittingham DG, Wilson L. Cytoplasmic control of preimplantation development in vitro in the mouse. Nature 1982; 299 (5882) 460-462
  • 53 Barritt JA, Brenner CA, Malter HE, Cohen J. Mitochondria in human offspring derived from ooplasmic transplantation. Hum Reprod 2001; 16 (3) 513-516
  • 54 Brenner CA, Barritt JA, Willadsen S, Cohen J. Mitochondrial DNA heteroplasmy after human ooplasmic transplantation. Fertil Steril 2000; 74 (3) 573-578
  • 55 Zoon KC. Human cells used in therapy involving the transfer of genetic material by means other than the union of gamete nuclei. United States FDA; 2001; Available at: http://www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/ucm105852.htm
  • 56 Bratic A, Larsson NG. The role of mitochondria in aging. J Clin Invest 2013; 123 (3) 951-957
  • 57 Tzeng CR, Hsieh RH, Au HK, Yen YH, Chang SJ, Cheng YF. Mitochondria transfer (MIT) into oocyte from autologous cumulus granulosa cells (cGCs). Fertil Steril 2004; 82 (Suppl. 02) S53
  • 58 Harvey AJ, Gibson TC, Quebedeaux TM, Brenner CA. Impact of assisted reproductive technologies: a mitochondrial perspective of cytoplasmic transplantation. Curr Top Dev Biol 2007; 77: 229-249
  • 59 El Shourbagy SH, Spikings EC, Freitas M, St John JC. Mitochondria directly influence fertilisation outcome in the pig. Reproduction 2006; 131 (2) 233-245
  • 60 Yi YC, Chen MJ, Ho JYP, Guu HF, Ho ES. Mitochondria transfer can enhance the murine embryo development. J Assist Reprod Genet 2007; 24 (10) 445-449
  • 61 Proceedings from the Workshop on Experts in Egg Health: Advancing Fertility Care, 31st Annual Meeting of the European Society of Human Reproduction and Embryology (ESHRE), Lisbon, Portugal 2015; Available at: http://www.ovascience.com/files/ESHRE_Symposium_2015_Final_For_Website_Posting_FINAL-2.pdf
  • 62 Acton BM, Lai I, Shang X, Jurisicova A, Casper RF. Neutral mitochondrial heteroplasmy alters physiological function in mice. Biol Reprod 2007; 77 (3) 569-576
  • 63 Sharpley MS, Marciniak C, Eckel-Mahan K , et al. Heteroplasmy of mouse mtDNA is genetically unstable and results in altered behavior and cognition. Cell 2012; 151 (2) 333-343
  • 64 Maximow A. The lymphocyte as a stem cell common to different blood elements in embryonic development and during the post-fetal life of mammals. Folia Haematol (Frankf) 1909; 8: 125-134
  • 65 Mimeault M, Batra SK. Recent progress on tissue-resident adult stem cell biology and their therapeutic implications. Stem Cell Rev 2008; 4 (1) 27-49
  • 66 Orford KW, Scadden DT. Deconstructing stem cell self-renewal: genetic insights into cell-cycle regulation. Nat Rev Genet 2008; 9 (2) 115-128
  • 67 Jung Y, Brack AS. Cellular mechanisms of somatic stem cell aging. Curr Top Dev Biol 2014; 107: 405-438
  • 68 Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science 2010; 327 (5965) 542-545
  • 69 Boyle M, Wong C, Rocha M, Jones DL. Decline in self-renewal factors contributes to aging of the stem cell niche in the Drosophila testis. Cell Stem Cell 2007; 1 (4) 470-478
  • 70 Pan L, Chen S, Weng C , et al. Stem cell aging is controlled both intrinsically and extrinsically in the Drosophila ovary. Cell Stem Cell 2007; 1 (4) 458-469
  • 71 Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell 2008; 132 (4) 598-611
  • 72 Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 2005; 433 (7027) 760-764
  • 73 Brack AS, Conboy MJ, Roy S , et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 2007; 317 (5839) 807-810
  • 74 Villeda SA, Luo J, Mosher KI , et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 2011; 477 (7362) 90-94
  • 75 Ruckh JM, Zhao JW, Shadrach JL , et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 2012; 10 (1) 96-103
  • 76 Ryu BY, Orwig KE, Oatley JM, Avarbock MR, Brinster RL. Effects of aging and niche microenvironment on spermatogonial stem cell self-renewal. Stem Cells 2006; 24 (6) 1505-1511
  • 77 Brinster RL, Zimmermann JW. Spermatogenesis following male germ-cell transplantation. Proc Natl Acad Sci USA 1994; 91 (24) 11298-11302
  • 78 Brinster RL, Avarbock MR. Germline transmission of donor haplotype following spermatogonial transplantation. Proc Natl Acad Sci USA 1994; 91 (24) 11303-11307
  • 79 Woods DC, Tilly JL. Germline stem cells in adult mammalian ovaries. In: Sanders S, ed., Ten Critical Topics in Reproductive Medicine. Washington, DC: Science/AAAS; 2013: 10-12
  • 80 Silvestris E, D'Oronzo S, Cafforio P, D'Amato G, Loverro G. Perspective in infertility: the ovarian stem cells. J Ovarian Res 2015; 8: 55
  • 81 Woods DC, Tilly JL. The next (re)generation of human ovarian biology and female fertility: is current science tomorrow's practice?. Fertil Steril 2012; 98 (1) 3-10
  • 82 Yesodi V, Yaron Y, Lessing JB, Amit A, Ben-Yosef D. The mitochondrial DNA mutation (ΔmtDNA5286) in human oocytes: correlation with age and IVF outcome. J Assist Reprod Genet 2002; 19 (2) 60-66
  • 83 Keefe DL, Niven-Fairchild T, Powell S, Buradagunta S. Mitochondrial deoxyribonucleic acid deletions in oocytes and reproductive aging in women. Fertil Steril 1995; 64 (3) 577-583
  • 84 Chan CC, Liu VW, Lau EY, Yeung WS, Ng EH, Ho PC. Mitochondrial DNA content and 4977 bp deletion in unfertilized oocytes. Mol Hum Reprod 2005; 11 (12) 843-846
  • 85 Monnot S, Samuels DC, Hesters L , et al. Mutation dependence of the mitochondrial DNA copy number in the first stages of human embryogenesis. Hum Mol Genet 2013; 22 (9) 1867-1872
  • 86 Fakih MH, El Shmoury M, Szeptycki J , et al. The AUGMENTSM treatment: physician reported outcomes of the initial global patient experience. JFIV Reprod Med Genet 2015; 3: 154
  • 87 Park A. The incredible, surprising, controversial new way to make a baby. Time 2015; 185 (18) 42-45
  • 88 Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet 1978; 2 (8085) 366
  • 89 Steptoe PC, Edwards RG, Walters DE. Observations on 767 clinical pregnancies and 500 births after human in-vitro fertilization. Hum Reprod 1986; 1 (2) 89-94
  • 90 Telfer EE, McLaughlin M, Ding C, Thong KJ. A two-step serum-free culture system supports development of human oocytes from primordial follicles in the presence of activin. Hum Reprod 2008; 23 (5) 1151-1158
  • 91 Telfer EE, McLaughlin M. In vitro development of ovarian follicles. Semin Reprod Med 2011; 29 (1) 15-23
  • 92 Telfer EE, Zelinski MB. Ovarian follicle culture: advances and challenges for human and nonhuman primates. Fertil Steril 2013; 99 (6) 1523-1533
  • 93 Cha KY, Chian RC. Maturation in vitro of immature human oocytes for clinical use. Hum Reprod Update 1998; 4 (2) 103-120
  • 94 Chang EM, Song HS, Lee DR, Lee WS, Yoon TK. In vitro maturation of human oocytes: Its role in infertility treatment and new possibilities. Clin Exp Reprod Med 2014; 41 (2) 41-46
  • 95 Shea LD, Woodruff TK, Shikanov A. Bioengineering the ovarian follicle microenvironment. Annu Rev Biomed Eng 2014; 16: 29-52
  • 96 Niikura Y, Niikura T, Tilly JL. Aged mouse ovaries possess rare premeiotic germ cells that can generate oocytes following transplantation into a young host environment. Aging (Albany, NY Online) 2009; 1 (12) 971-978
  • 97 Xiong J, Lu Z, Wu M , et al. Intraovarian transplantation of female germline stem cells rescues ovarian function in chemotherapy-injured ovaries. PLoS One 2015; 10 (10) e0139824
  • 98 Woods DC, Tilly JL. Reply to human and mouse ovaries lack DDX4-expressing functional oogonial stem cells. Nat Med 2015; 21 (10) 1118-1121
  • 99 Oktay K, Baltaci V, Sonmezer M , et al. Oogonial precursor cell derived autologous mitochondria injection (AMI) to improve outcomes in women with multiple IVF failures due to low oocyte quality: a clinical translation. Reprod Sci 2015; 22 (12) 1612-1617