DOI QR코드

DOI QR Code

Intraovarian vascular enhancement via stromal injection of platelet-derived growth factors: Exploring subsequent oocyte chromosomal status and in vitro fertilization outcomes

  • Wood, Samuel H. (Gen 5 Fertility Center) ;
  • Sills, E. Scott (Gen 5 Fertility Center)
  • Received : 2019.11.01
  • Accepted : 2019.12.24
  • Published : 2020.06.30

Abstract

The inverse correlation between maternal age and pregnancy rate represents a major challenge for reproductive endocrinology. The high embryo ploidy error rate in failed in vitro fertilization (IVF) cycles reflects genetic misfires accumulated by older oocytes over time. Despite the application of different follicular recruitment protocols during IVF, gonadotropin modifications are generally futile in addressing such damage. Even when additional oocytes are retrieved, quality is frequently poor. Older oocytes with serious cytoplasmic and/or chromosomal errors are often harvested from poorly perfused follicles, and ovarian vascularity and follicular oxygenation impact embryonic chromosomal competency. Because stimulation regimens exert their effects briefly and immediately before ovulation, gonadotropins alone are an ineffective antidote to long-term hypoxic pathology. In contrast, the tissue repair properties (and particularly the angiogenic effects) of platelet-rich plasma (PRP) are well known, with applications in other clinical contexts. Injection of conventional PRP and/or its components (e.g., isolated platelet-derived growth factors as a cell-free substrate) into ovarian tissue prior to IVF has been reported to improve reproductive outcomes. Any derivative neovascularity may modulate oocyte competence by increasing cellular oxygenation and/or lowering concentrations of intraovarian reactive oxygen species. We propose a mechanism to support intrastromal angiogenesis, improved follicular perfusion, and, crucially, embryo ploidy rescue. This last effect may be explained by mRNA upregulation coordinated by PRP-associated molecular signaling, as in other tissue systems. Additionally, we outline an intraovarian injection technique for platelet-derived growth factors and present this method to help minimize reliance on donor oocytes and conventional hormone replacement therapy.

Keywords

References

  1. Keefe DL. Telomeres, reproductive aging, and genomic instability during early development. Reprod Sci 2016;23:1612-5. https://doi.org/10.1177/1933719116676397
  2. Van Blerkom J. Epigenetic influences on oocyte developmental competence: perifollicular vascularity and intrafollicular oxygen. J Assist Reprod Genet 1998;15:226-34. https://doi.org/10.1023/A:1022523906655
  3. Borini A, Maccolini A, Tallarini A, Bonu MA, Sciajno R, Flamigni C. Perifollicular vascularity and its relationship with oocyte maturity and IVF outcome. Ann N Y Acad Sci 2001;943:64-7. https://doi.org/10.1111/j.1749-6632.2001.tb03791.x
  4. Fernandez-Moure JS, Van Eps JL, Cabrera FJ, Barbosa Z, Medrano Del Rosal G, Weiner BK, et al. Platelet-rich plasma: a biomimetic approach to enhancement of surgical wound healing. J Surg Res 2017;207:33-44. https://doi.org/10.1016/j.jss.2016.08.063
  5. Dhillon RS, Schwarz EM, Maloney MD. Platelet-rich plasma therapy: future or trend? Arthritis Res Ther 2012;14:219. https://doi.org/10.1186/ar3914
  6. Pantos K, Nitsos N, Kokkali G, Vaxevanoglou T, Markomichali C, Pantou A, et al. Ovarian rejuvenation and folliculogenesis reactivation in peri-menopausal women after autologous plateletrich plasma treatment. ESHRE 32nd annual meeting; 2016 Jul 3-6; Helsinki, Finland. Hum Reprod 2016;Suppl 1:i301.
  7. Gkini MA, Kouskoukis AE, Tripsianis G, Rigopoulos D, Kouskoukis K. Study of platelet-rich plasma injections in the treatment of androgenetic alopecia through an one-year period. J Cutan Aesthet Surg 2014;7:213-9. https://doi.org/10.4103/0974-2077.150743
  8. Vahabi S, Yadegari Z, Mohammad-Rahimi H. Comparison of the effect of activated or non-activated PRP in various concentrations on osteoblast and fibroblast cell line proliferation. Cell Tissue Bank 2017;18:347-53. https://doi.org/10.1007/s10561-017-9640-7
  9. Kim KH, Oh DS, Jeong JH, Shin BS, Joo BS, Lee KS. Follicular blood flow is a better predictor of the outcome of in vitro fertilization-embryo transfer than follicular fluid vascular endothelial growth factor and nitric oxide concentrations. Fertil Steril 2004;82:586-92. https://doi.org/10.1016/j.fertnstert.2004.02.120
  10. Lakhani K, Leonard A, Seifalian AM, Hardiman P. Microvascular dysfunction in women with polycystic ovary syndrome. Hum Reprod 2005;20:3219-24. https://doi.org/10.1093/humrep/dei199
  11. Chattopadhayay R, Ganesh A, Samanta J, Jana SK, Chakravarty BN, Chaudhury K. Effect of follicular fluid oxidative stress on meiotic spindle formation in infertile women with polycystic ovarian syndrome. Gynecol Obstet Invest 2010;69:197-202. https://doi.org/10.1159/000270900
  12. Tao Y, Liu XJ. Deficiency of ovarian ornithine decarboxylase contributes to aging-related egg aneuploidy in mice. Aging Cell 2013;12:42-9. https://doi.org/10.1111/acel.12016
  13. Menezo YJ, Silvestris E, Dale B, Elder K. Oxidative stress and alterations in DNA methylation: two sides of the same coin in reproduction. Reprod Biomed Online 2016;33:668-83. https://doi.org/10.1016/j.rbmo.2016.09.006
  14. Zhang M, Miao Y, Chen Q, Cai M, Dong W, Dai X, et al. BaP exposure causes oocyte meiotic arrest and fertilization failure to weaken female fertility. FASEB J 2018;32:342-52. https://doi.org/10.1096/fj.201700514R
  15. Jana SK, K NB, Chattopadhyay R, Chakravarty B, Chaudhury K. Upper control limit of reactive oxygen species in follicular fluid beyond which viable embryo formation is not favorable. Reprod Toxicol 2010;29:447-51. https://doi.org/10.1016/j.reprotox.2010.04.002
  16. Yadav AK, Yadav PK, Chaudhary GR, Tiwari M, Gupta A, Sharma A, et al. Autophagy in hypoxic ovary. Cell Mol Life Sci 2019;76:3311-22. https://doi.org/10.1007/s00018-019-03122-4
  17. Mihalas BP, Redgrove KA, McLaughlin EA, Nixon B. Molecular mechanisms responsible for increased vulnerability of the ageing oocyte to oxidative damage. Oxid Med Cell Longev 2017;2017:4015874.
  18. Callejo J, Salvador C, Gonzalez-Nunez S, Almeida L, Rodriguez L, Marques L, et al. Live birth in a woman without ovaries after autograft of frozen-thawed ovarian tissue combined with growth factors. J Ovarian Res 2013;6:33. https://doi.org/10.1186/1757-2215-6-33
  19. Bakacak M, Bostanci MS, Inanc F, Yaylali A, Serin S, Attar R, et al. Protective effect of platelet rich plasma on experimental ischemia/reperfusion injury in rat ovary. Gynecol Obstet Invest 2016;81:225-31. https://doi.org/10.1159/000440617
  20. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 2008;453:314-21. https://doi.org/10.1038/nature07039
  21. Lacci KM, Dardik A. Platelet-rich plasma: support for its use in wound healing. Yale J Biol Med 2010;83:1-9.
  22. Sfakianoudis K, Simopoulou M, Nitsos N, Rapani A, Pantou A, Vaxevanoglou T, et al. A case series on platelet-rich plasma revolutionary management of poor responder patients. Gynecol Obstet Invest 2019;84:99-106. https://doi.org/10.1159/000491697
  23. Sills ES, Rickers NS, Li X, Palermo GD. First data on in vitro fertilization and blastocyst formation after intraovarian injection of calcium gluconate-activated autologous platelet rich plasma. Gynecol Endocrinol 2018;34:756-60. https://doi.org/10.1080/09513590.2018.1445219
  24. Lubkowska A, Dolegowska B, Banfi G. Growth factor content in PRP and their applicability in medicine. J Biol Regul Homeost Agents 2012;26(2 Suppl 1):3S-22S.
  25. Martinez CE, Smith PC, Palma Alvarado VA. The influence of platelet-derived products on angiogenesis and tissue repair: a concise update. Front Physiol 2015;6:290. https://doi.org/10.3389/fphys.2015.00290
  26. Sills ES, Rickers NS, Svid CS, Rickers JM, Wood SH. Normalized ploidy following 20 consecutive blastocysts with chromosomal error: healthy 46, XY pregnancy with IVF after intraovarian injection of autologous enriched platelet-derived growth factors. Int J Mol Cell Med 2019;8:84-90.
  27. Imai S, Kumagai K, Yamaguchi Y, Miyatake K, Saito T. Platelet-rich plasma promotes migration, proliferation, and the gene expression of scleraxis and vascular endothelial growth factor in paratenon-derived cells in vitro. Sports Health 2019;11:142-8. https://doi.org/10.1177/1941738118807479
  28. Zhang Y, Morgan BJ, Smith R, Fellows CR, Thornton C, Snow M, et al. Platelet-rich plasma induces post-natal maturation of immature articular cartilage and correlates with LOXL1 activation. Sci Rep 2017;7:3699. https://doi.org/10.1038/s41598-017-02297-9
  29. Shen H, Cheng H, Chen H, Zhang J. Identification of key genes induced by platelet-rich plasma in human dermal papilla cells using bioinformatics methods. Mol Med Rep 2017;15:81-8. https://doi.org/10.3892/mmr.2016.5988
  30. Sills ES, Takeuchi T, Tucker MJ, Palermo GD. Genetic and epigenetic modifications associated with human ooplasm donation and mitochondrial heteroplasmy: considerations for interpreting studies of heritability and reproductive outcome. Med Hypotheses 2004;62:612-7. https://doi.org/10.1016/j.mehy.2003.10.008
  31. Jing Y, Li L, Li YY, Ouyang YC, Sun QY, Zhang CL, et al. Embryo quality, and not chromosome nondiploidy, affects mitochondrial DNA content in mouse blastocysts. J Cell Physiol 2019;234:10481-8. https://doi.org/10.1002/jcp.27713
  32. Lledo B, Ortiz JA, Morales R, Garcia-Hernandez E, Ten J, Bernabeu A, et al. Comprehensive mitochondrial DNA analysis and IVF outcome. Hum Reprod Open 2018;2018:hoy023. https://doi.org/10.1093/hropen/hoy023
  33. Whitcomb BW, Purdue-Smithe AC, Szegda KL, Boutot ME, Hankinson SE, Manson JE, et al. Cigarette smoking and risk of early natural menopause. Am J Epidemiol 2018;187:696-704. https://doi.org/10.1093/aje/kwx292
  34. Sills ES, Li X, Rickers NS, Wood SH, Palermo GD. Metabolic and neurobehavioral response following intraovarian administration of autologous activated platelet rich plasma: first qualitative data. Neuro Endocrinol Lett 2019;39:427-33.
  35. Oliveira PH, Boura JS, Abecasis MM, Gimble JM, da Silva CL, Cabral JM. Impact of hypoxia and long-term cultivation on the genomic stability and mitochondrial performance of ex vivo expanded human stem/stromal cells. Stem Cell Res 2012;9:225-36. https://doi.org/10.1016/j.scr.2012.07.001
  36. Aiken CE, Tarry-Adkins JL, Spiroski AM, Nuzzo AM, Ashmore TJ, Rolfo A, et al. Chronic gestational hypoxia accelerates ovarian aging and lowers ovarian reserve in next-generation adult rats. FASEB J 2019;33:7758-66. https://doi.org/10.1096/fj.201802772r
  37. Pampanini V, Jahnukainen K, Sahlin L, Germani D, Puglianiello A, Cianfarani S, et al. Impact of uteroplacental insufficiency on ovarian follicular pool in the rat. Reprod Biol Endocrinol 2019;17:10. https://doi.org/10.1186/s12958-019-0453-3

Cited by

  1. Ovarian Rejuvenation Using Autologous Platelet-Rich Plasma vol.2, pp.1, 2020, https://doi.org/10.3390/endocrines2010002
  2. The Scientific and Cultural Journey to Ovarian Rejuvenation: Background, Barriers, and Beyond the Biological Clock vol.8, pp.6, 2020, https://doi.org/10.3390/medicines8060029