Skip to main content

VSELs may obviate cryobanking of gonadal tissue in cancer patients for fertility preservation

Abstract

Background

Infertility is an undesirable side effect and gonadal tissue banking is advocated in young cancer patients who are unable to preserve embryos or gametes prior to oncotherapy to achieve biological parenthood later on. Banking gonadal tissue is challenging and protocols to mature gametes in vitro are not yet clinically established. Transplanting ovarian cortical tissue at hetero-or orthotopic sites in women and bone marrow transplantation (BMT) in both men and women has resulted in spontaneous recovery of fertility, pregnancy and live births. Various studies in humans and mice suggest that genetic origin of offspring after BMT is similar to transplanted patient and not the donor. Thus the source of oocytes/sperm which result in spontaneous pregnancies still remains contentious.

Findings

Very small embryonic-like stem cells (VSELs) have been reported in adult human testis and ovary, in azoospermic testicular biopsies from survivors of childhood cancer and also in women with premature ovarian failure and menopause. VSELs survive chemotherapy because of their quiescent nature and can be detected in chemoablated mice gonads at protein and mRNA level and also by flow cytometry. Surviving VSELs spontaneously differentiate into oocyte-like structures and sperm when inhibitory factors are overcome in vitro. Transplantation of mesenchymal cells (isolated from different sources) has led to regeneration of chemoablated mouse gonads and also live births. Spermatogenesis is also restored from endogenous stem cells on inter-tubular transplantation of Sertoli cells in chemoablated mouse testis.

Conclusions

Endogenous VSELs (which survive oncotherapy) can possibly regenerate non-functional gonads in cancer survivors when exposed to a healthy niche in vitro or in vivo (by way of transplanting mesenchymal cells which secrete trophic factors required for endogenous VSELs to differentiate into gametes). Presence of VSELs can also explain spontaneous pregnancies after BMT and cortical tissue transplantation (at heterotopic or orthotopic sites). This understanding once verified and accepted by the scientific community could obviate the need to remove whole ovary or testicular biopsy for cryopreservation prior to oncotherapy.

Background

With advanced treatment options available today, more than 80 % of young cancer patients survive however infertility is often an unwanted side effect. The only option available to them is to cryopreserve the sperm, oocytes or embryos and use later for assisted reproduction to achieve biological parenthood. They could also be counseled to adopt a child. Another option in young children where sperm/oocytes/embryos may not be available is to cryopreserve testicular biopsy or ovarian cortical tissue. One whole ovary is removed surgically and cortical tissue slices are cryopreserved. Later on, when the cancer survivor plans a family, the cryopreserved tissue can be used as a source of gametes for assisted reproduction. Various other strategies which could avoid harming the gonads during oncotherapy like shielding the gonads during radiotherapy and GnRH antagonist are also used as a part of ‘onco-medicine’. The current understanding, clinical practice and American Society of Clinical Oncology (ASCO) guidelines in the field for fertility preservation in cancer patients are available for ready reference [1, 2].

Procedures to obtain gametes from ovarian/testicular tissue or stem cells are not yet clinically established. Spontaneous parenthood followed by live births has been noticed after bone marrow transplantation (BMT) and also on transplanting cortical tissue at hetero- or orthotopic sites. The exact source of sperm or oocyte which results in these spontaneous pregnancies is still not clear. A genetic similarity exists between the patient (recipient) and offspring after BMT refuting earlier claims that transplanted bone marrow could be a source of gametes. Similarly transplanting cortical tissue both subcutaneously or on the surface or vicinity of the non-functional ovary has resulted in live births. What was the source of these oocytes? It could be the transplanted cortical tissue slices or the non-functional gonad regenerated somehow and produced oocytes! Research in authors’ laboratory over last 6–7 years suggests that non-functional gonads possibly regenerate after BMT and cortical tissue transplantation. This is the main focus of the current review which will hopefully result in a major paradigm shift in the field of oncofertility in times to come.

Stem cells (VSELs) and progenitors (OSCs, SSCs) in ovary and testis

More than a decade ago, Professor Jonathan Tilly from Harvard University, USA challenged the existing dogma that a female is born with a fixed number of eggs at birth; rather he suggested that a continuous replenishment of follicles occurs throughout life [3]. Several groups were drawn to this field of research and evidence was generated that stem cells are housed in adult mammalian ovary surface epithelium (human, sheep, marmoset, rabbit, and mouse) as well as in women with premature ovarian failure (POF) and menopause [4–8]. Presence of stem cells in adult ovary is still not well accepted [9]; however Woods and Tilly [10] have published protocols to isolate ovarian stem cells (OSCs) from human ovaries and equated them to spermatogonial stem cells in testis [11]. Zhang et al. [12] recently described their inability to sort human OSCs using DDX4 as a cell surface marker by compiling together independent work done in four different laboratories to which Woods and Tilly have responded [13]. Silvestris et al. [14] successfully isolated DDX4 positive OSCs and showed further that the DDX4 positive OSCs are also positive for OCT-4. We routinely isolate VSELs and OSCs by mechanical scraping of ovary surface epithelium or by enzymatic method and use flow cytometry only as a tool to further characterize the stem cells [15].

Interesting data has been generated showing that the stem cells in adult ovary can spontaneously differentiate into oocyte-like structures in vitro [7, 16, 17] and on injection in human cortical biopsies lead to primordial follicle assembly [18]. A careful examination of the OSE cells smears showed the presence of small (2–5μm), round cells which expressed pluripotent markers SSEA-4, Oct-4, Nanog, Sox-2, and c-kit [5, 6, 19]. Using a polyclonal OCT-4 antibody, Parte et al. [7] for the first time documented the presence of two distinct populations of stem cells in OSE smears including very small stem cells (earlier described by Viran-Klun’s group as well) expressing nuclear OCT-4 and cell surface SSEA-4 termed very small embryonic-like stem cells (VSELs) and slightly bigger stem cells termed ovarian germ stem cells (OGSCs) with cytoplasmic OCT-4. Based on size difference between VSELs (2–5 μm) and OGSCs (>8–10 μm), we speculate that Tilly’s group detected the OGSCs (OSCs) and missed out on the VSELs in adult ovary because of very small size and low abundance of VSELs. Protocols to isolate and characterize both (VSELs and OGSCs) the stem cell populations in adult mammalian ovaries were recently published [15, 20]. Follicle stimulating hormone (FSH) exerts direct action on the ovarian VSELs (express FSH receptors) to undergo self-renewal, clonal expansion to form germ cell nests and further oocyte differentiation [21]. We also discussed that menopause occurs despite high circulatory FSH due to a compromised niche that is unable to support stem cells differentiation into follicles [22]. Stem cells from menopausal ovary spontaneously differentiate into oocyte-like structures when the inhibitory factors are overcome in vitro [5, 7]. Similarly stem cells from aged mouse ovaries differentiate and give rise to oocytes on being transplanted into a young somatic environment [23]. Similar VSELs were earlier reported by our group in adult human testis as a sub-group among spermatogonial stem cells (SSCs) on the basis of size and nuclear versus cytoplasmic staining of OCT-4. This was established through extensive characterization by immunolocalization using 3 different OCT-4 antibodies, qRT-PCR studies, in- situ hybridization and Western analysis [24]. VSELs have also been extensively characterized in adult mouse testis [25].

To conclude this section, both ovary and testis harbor pluripotent VSELs along with the specific progenitors which include OSCs in the ovary and SSCs in the testis.

VSELs are the quiescent stem cell population in the gonads and survive oncotherapy

The VSELs were first reported by Ratajczak’s group [26] in various adult mouse organs including testis and they postulate that pluripotent primordial germ cells (PGCs) during their migration along the dorsal mesentery towards the gonadal ridge to form the germ cells, migrate and settle in various adult organs throughout life [27]. The work became controversial recently when a leading stem cell biologist was unable to detect VSELs in mouse bone marrow [28], but the underlying reasons for their failure were technical as discussed by Ratajczak’s group [29, 30]. We have recently confirmed and extensively characterized VSELs in human cord blood [31]. It is probably because of the very small size, low abundance and minimal cytoplasm that VSELs have remained obscure till now. When cord blood/bone marrow is subjected to Ficoll-Hypaque centrifugation – the VSELs settle down with red blood cells and have been invariably discarded unknowingly in the past [32]. In the gonads, it is relatively easier to conceptualize that a small number of PGCs survive in adult gonads as VSELs and this has also been suggested by other group [33]. Ratajczak’s group have shown that VSELs are relatively quiescent and when mice are subjected to total body irradiation, bone marrow gets depleted of hematopoietic stem cells whereas the VSELs survive and show increased uptake of BrdU [34]. Shin et al. [35] reported that quiescent state of VSELs is because of unique DNA methylation pattern of developmentally crucial imprinted-genes showing hypomethylation/erasure of imprints in paternally methylated genes and hypermethylation of imprints in maternally methylated ones. As a result VSELs express increased levels of H19and Cdkn1c and lowered levels of Igf2 and Rasgrf1accounting for their quiescence.

Available literature suggests that all renewing body organs including skin, hair follicle, gut epithelium, hematopoietic system harbor two populations of stem cells which include quiescent and actively dividing stem cells [36, 37]. Based on these published literature, we decided to study VSELs in mouse ovary and testis to gauge the effect of busulphan and cyclophosphamide treatment on them [8, 25]. Besides immuno-localization and qRT-PCR analysis to show presence of pluripotent VSELs, we were able to quantitate them by flow cytometry as <6 μm sized cells which are LIN-/CD45-/SCA-1+. Results shown in Table 1 demonstrate that VSELs exist in normal gonads, survive chemotherapy and undergo self-renewal in response to PMSG treatment.

Table 1 Flow cytometry results on VSELs (LIN-/CD45-/SCA-1+) expressed as % of total cells

OCT-4 positive stem cells were also detected by flow cytometry in OSE cells from sheep ovaries (VSELs 1.26 ± 0.19 % of 2–4μm and OGSCs 6.86 ± 0.5 % of 4–9μm). Parte et al. [38] showed that a large number of stem cells are shed on cell culture inserts during both marmoset and human cortical tissue culture and these stem cells undergo spontaneous differentiation into oocyte-like structures in vitro. Presence of germ cell nests, Balbiani body-like structures and cytoplasmic streaming extensively described during fetal ovary development, are indeed well recapitulated during in vitro oogenesis in adult OSE cultures along with characteristic expression of stem/germ cell/oocyte markers [13]. Similar VSELs have also been reported in testicular biopsy collected from azoospermic cancer survivors [39] and also in POF ovaries [5, 6].

Testicular function restoration in cancer survivors

Spontaneous paternity and live birth following bone marrow conditioning and transplantation has surprised everyone. Ignatove et al. [40] reviewed 8 such cases and reported two more themselves, where although the patients were azoospermic, testicular sperm helped attain biological parenthood. Dupont et al. [41] recently reported spontaneous conception after autologus BMT. A 20 years old man suffering from acute promyelocytic leukemia, even though was repeatedly diagnosed azoospermic with zero sperm count fathered healthy children spontaneously [41]. The underlying reason is not yet clear but it is likely that there may be a stem cell link for fertility restoration in men similar to that discussed by Oktay et al. [42] in women (please refer to next section).

Transplanting germ cells from cryopreserved testicular tissue still remains experimental and has not been extensively studied in men as yet. Hermann et al. [43] have reported initial success and carried out SSCs transplantation in 18 adult and 5 pre-pubertal busulphan treated Rhesus monkeys through ultrasound guided transfer via rete testis. Sperm collected from these monkeys were injected in 85 oocytes by ICSI, 81 fertilized and 4 cells to blastocyst stage embryos were obtained. 7/81 embryos were of donor origin. Struijk et al. [44] critically reviewed the progress made to restore fertility in survivors of childhood cancer by auto- transplanting SSCs and concluded that transplanted cells colonize but do not differentiate. Navaeian-Kalat et al. [45] reviewed available literature and suggested that trans-differentiation of mesenchymal cells is the only way to treat infertility. Several attempts have been reported to restore spermatogenesis in chemoablated testis by transplanting mesenchymal cells as shown in Table 2. Different doses of mesenchymal cells (and isolated from different sources) have been injected directly into the seminiferous tubules via rete testis or efferent ducts, intravenous injections or direct inter-tubular injections into the testis. It has been postulated that mesenchymal cells are ‘pluripotent’ and can trans-differentiate into gametes. But this is possibly not true [57] and our results suggest that mesenchymal cells only provide a niche (source of growth factors) to the VSELs that survive chemotherapy. Rather than injections into the rete testis or efferent ducts of the seminiferous tubules (challenging technically and not a practical solution), inter-tubular injections of Sertoli cells or mesenchymal cells as a simple OPD procedure may suffice to restore spermatogenesis – based on our studies in mice [25].

Table 2 Mesenchymal Cells to Restore Spermatogenesis

Ovarian function restoration in cancer survivors

A total of 37 births have occurred till now by transplanting cryopreserved cortical tissue in cancer survivors [58]. Recently, Demeestere et al. [59] report spontaneous pregnancy and live-birth in a 27 years old survivor from cryopreserved ovarian cortical tissue which was cryopreserved as she suffered from sickle cell anemia at the age of 5 years. Four thawed ovarian fragments were grafted on the residual left ovary, six were grafted in the right peritoneal bursa, and five were grafted subcutaneously. Normal menstruation along with normal FSH and undetectable AMH levels was maintained after transplantation and the woman became pregnant spontaneously after more than 2 years of transplantation of cortical tissue. They reported that cortical transplantation led to successful recovery of ovarian function. Since other reports have shown spontaneous recovery of atretic ovary even without direct ovarian transplantation, it is almost impossible to reason out source of oocytes that gave rise to the babies - whether it is cortical tissue or the ‘regenerated’ non-functional ovary. Four years ago, Oktay and his team reported 4 spontaneous pregnancies and 3 live births after subcutaneous transplantation of cryopreserved ovarian tissue [42] in a 28 years old survivor of Hodgkin’s disease. Origin of the pregnancy was proposed to be due to improvement of function of in situ ‘menopausal’ ovary by hormones secreted by transplanted cortical tissue [60]. Oktay et al. [61] discussed a possible stem cell link to spontaneous pregnancy. Besides destroying follicles, oncotherapy also destroys the somatic niche and transplantation of cortical tissue provides ‘regenerative signals’ to intact non-functional ovary enabling oocyte formation from stem cells (source could be bone marrow) and hence pregnancy and live births.

Authors propose that it could be the surviving VSELs in non-functional ovary that become functional and undergo neo-oogenesis and primordial follicle assembly and these follicles then undergo normal maturation and ovulation resulting in spontaneous pregnancy and live births. These views are strongly supported by a recent study by our group on chemoablated mice ovaries wherein Sriraman et al. [8] found the VSELs survive chemotherapy, are located in the OSE and are functional since they self-renew in response to PMSG treatment (Table 1). Culture of intact chemoablated ovary in presence of FSH resulted in appearance of proliferating germ cell clusters in situ that incorporated BrdU and expressed MVH and STRA8. When enzymatically isolated OSE in vitro were cultured, germ cell nests that co-expressed PCNA and OCT-4 were observed. Prolonged culture showed differentiated GDF-9 and MVH expressing oocyte-like structures in vitro. The study underwent a very strict review and we were advised to publish 4 images of germ cell nests (in the Supplement) to convince the readers as it is very significant finding that postnatal ovary even after chemoablation retains stem cells which can form germ cell nests. These results are in contrast to those published by Lei and Spradling [62] who earlier failed to detect any evidence for presence of stem cells in adult ovary and we had argued that absence of evidence does not necessarily imply evidence for absence [63]. It is interesting to point out that (i) Lee et al. [64] showed that although bone marrow transplantation could rescue long-term fertility in chemoablated mice- but all the offspring were of recipient germline (ii) Niikura et al. [23] reported that stem cells in aged ovary can form follicles when exposed to a young niche of the host (iii) Edessy et al. [65] transplanted autologus mesenchymal cells isolated from bone marrow into the POF ovary resulted in resumption of menstruation and secretory changes in the endometrium and (iv) Zhu et al. [66] reported beneficial effect of transplanting human cord blood mesenchymal cells in rat chemoablated ovary. Table 3 shows various studies demonstrating beneficial effect of transplanting mesenchymal cells in chemoablated ovary associated with live births.

Table 3 Mesenchymal Cells to Restore Ovarian Function

Just as in men, spontaneous pregnancy and birth of babies has been reported after BMT in women. Veitia et al. [76] observed spontaneous menstruation followed by pregnancy in a woman after 8 years of BMT, although she was expected to be infertile after suffering from Fanconi anemia and aggressive BMT conditioning regimen. Genetic analysis of the mother (patient), daughter and the donor revealed genetic relationship between the mother and daughter. As a result they ruled out that transplanted bone marrow was a source of oocytes. Similarly Nabhan et al. [77] also confirmed genetic relationship between the transplanted mother and daughter and excluded the possibility of germ cells transmission from the donor. We postulate that the transplanted bone marrow only helps to improve the niche (is not a source of germ cells) and endogenous VSELs differentiate into oocytes resulting in spontaneous birth of babies.

Conclusions

To conclude, we have made a humble attempt based on available literature, to reason out that both ovary and testis rendered infertile by cancer treatment retain a novel stem cell population of VSELs that survive because of their quiescent nature and have the potential to regenerate the non-functional gonads when a healthy niche is provided. Authors opine that thus there may not be any need to remove ovary or testicular biopsy in children/adults prior to cancer therapy for later transplantation and this will result in major paradigm shift in the field of oncofertility. The non-functional ovaries and testis in young cancer survivors could be regenerated by transplanting mesenchymal niche cells which will secrete trophic factors required for the surviving VSELs to differentiate into gametes. This will help them in normal development of secondary sexual characteristics, avoid hormone replacement therapy, normal menstrual cycles and they may plan parenthood whenever they desire. Our reasoning in this article has huge translational potential and will greatly help improve quality life of cancer survivors. We are open to anyone who is interested to visit our lab to observe and be convinced by the presence of VSELs in chemoablated testis and ovary.

End notes

1. Adult mammalian ovary and testis harbor two populations of stem cells including pluripotent VSELs which are relatively quiescent and undergo asymmetric divisions to self-renew and give rise to the progenitors which comprise spermatogonial stem cells (SSCs) in testis and ovarian stem cells (OSCs or OGSCs) in ovaries. This stem cells activity in the ovary surface epithelium and testicular basal seminiferous tubular epithelium maintains life-long homeostasis in the gonads.

2. Being quiescent, VSELs survive oncotherapy as the cancer treatment targets actively dividing cells. Several reports suggest that even the niche gets compromised by oncotherapy and thus cannot support (provide growth factors and cytokines) VSELs to differentiate and form gametes. Studies in mice and circumstantial evidence in humans suggest that when chemoablated testis and ovary are exposed to a healthy microenvironment, VSELs differentiate into healthy gametes and result in live pregnancies.

3. Differentiation of ES or iPS cells in vitro into gametes is not yet successful as they do not easily convert into PGCs whereas the VSELs (considered equivalent to PGCs) isolated from normal and chemoablated gonads spontaneously differentiate into sperm and oocyte-like structures [7, 8, 78]. The underlying reasons for better potential of VSELs compared to ES/iPS cells to produce ‘synthetic gametes’ were recently discussed [79].

4. There may be no need to cryopreserve testicular and ovarian tissue prior to cancer treatment. Urgent pilot clinical trials (transplantation of mesenchymal cells directly into chemoablated gonads) need to be undertaken to provide a strong basis to our viewpoint. Meantime, if testicular tissue is collected for cryopreservation, it may be ideal to expand Sertoli cells in culture and cryopreserve for future use in addition to the germ cells.

Abbreviations

BMT:

Bone marrow transplantation

ES cells:

Embryonic stem cells

iPS cells:

Induced pluripotent stem cells

MSCs:

Mesenchymal stem cells

OGSCs:

Ovarian germ stem cells

OSCs:

Oogonial stem cells

OSE:

Ovary surface epithelium

PGCs:

Primordial germ cells

POF:

Premature ovarian failure

SSCs:

Spermatogonial stem cells

VSELs:

Very small embryonic-like stem cells

References

  1. Sonigo C, Sermondade N, Benard J, Benoit A, Shore J, Sifer C, Grynberg M. The past, present and future of fertility preservation in cancer patients. Future Oncol. 2015. doi:10.2217/fon.15.152

  2. Loren AW, Mangu PB, Beck LN, Brennan L, Magdalinski AJ, Partridge AH, et al. Fertility preservation for patients with cancer: American society of clinical oncology clinical practice guideline update. J Clin Oncol. 2013;31:2500.

    Article  PubMed  Google Scholar 

  3. Johnson J, Canning J, Kaneko T, Pru JK, Tilly JL. Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature. 2004;428:145–50.

    Article  CAS  PubMed  Google Scholar 

  4. Bukovsky A, Caudle MR, Svetlikova M, Upadhyaya NB. Origin of germ cells and formation of new primary follicles in adult human ovaries. Reprod Biol Endocrinol. 2004;2:20.

    Article  PubMed Central  PubMed  Google Scholar 

  5. Virant-Klun I, Skutella T, Stimpfel M, Sinkovec J. Ovarian surface epithelium in patients with severe ovarian infertility: a potential source of cells expressing markers of pluripotent/multipotent stem cells. J Biomed Biotechnol. 2011;2011:381928.

    Article  PubMed Central  PubMed  Google Scholar 

  6. Virant-Klun I, Zech N, Rozman P, Vogler A, Cvjeticanin B, Klemenc P, et al. Putative stem cells with an embryonic character isolated from the ovarian surface epithelium of women with no naturally present follicles and oocytes. Differentiation. 2008;76:843–56.

    Article  CAS  PubMed  Google Scholar 

  7. Parte S, Bhartiya D, Telang J, Daithankar V, Salvi V, Zaveri K, et al. Detection, characterization, and spontaneous differentiation in vitro of very small embryonic-like putative stem cells in adult mammalian ovary. Stem Cells Dev. 2011;20:1451–64.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Sriraman K, Bhartiya D, Anand S, Bhutda S. Mouse ovarian very small embryonic-like stem cells resist chemotherapy and retain ability to initiate oocyte-specific differentiation. Reprod Sci. 2015;22:884–903.

    Article  CAS  PubMed  Google Scholar 

  9. Vassena R, Eguizabal C, Heindryckx B, Sermon K, Simon C, van Pelt AM, et al. Stem cells in reproductive medicine: ready for the patient? Hum Reprod. 2015;30:2014–21.

    Article  CAS  PubMed  Google Scholar 

  10. Woods DC, Tilly JL. Isolation, characterization and propagation of mitotically active germ cells from adult mouse and human ovaries. Nat Protoc. 2013;8:966–88.

    Article  PubMed  Google Scholar 

  11. Woods DC, Tilly JL. An evolutionary perspective on adult female germline stem cell function from flies to humans. Semin Reprod Med. 2013;31:24–32.

    Article  PubMed  Google Scholar 

  12. Zhang H, Panula S, Petropoulos S, Edsgärd D, Busayavalasa K, Liu L, et al. Adult human and mouse ovaries lack DDX4-expressing functional oogonial stem cells. Nat Med. 2015;21:1116–8.

    Article  PubMed  Google Scholar 

  13. Woods DC, Tilly JL. Reply to Adult human and mouse ovaries lack DDX4-expressing functional oogonial stem cells. Nat Med. 2015;21:1118–21.

    Article  PubMed  Google Scholar 

  14. 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.

    Article  PubMed Central  PubMed  Google Scholar 

  15. Parte S, Patel H, Sriraman K, Bhartiya D. Isolation and characterization of stem cells in the adult mammalian ovary. Methods Mol Biol. 2015;1235:203–29.

    Article  PubMed  Google Scholar 

  16. Bukovsky A, Svetlikova M, Caudle MR. Oogenesis in cultures derived from adult human ovaries. Reprod Biol Endocrinol. 2005;3:17.

    Article  PubMed Central  PubMed  Google Scholar 

  17. Virant-Klun I, Skutella T, Kubista M, Vogler A, Sinkovec J, Meden-Vrtovec H. Expression of pluripotency and oocyte-related genes in single putative stem cells from human adult ovarian surface epithelium cultured in vitro in the presence of follicular fluid. Biomed Res Int. 2013;2013:861460.

    PubMed Central  PubMed  Google Scholar 

  18. White YA, 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;18:413–21.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Virant Klun I, Rozman P, Cvjeticanin B, Vrtacnik Bokal E, Novakovic S, Rulicke TR, et al. Parthenogenetic embryo-like structures in the human ovarian surface epithelium cell culture in postmenopausal women with no naturally present follicles and oocytes. Stem Cells Dev. 2009;18:137–49.

    Article  CAS  PubMed  Google Scholar 

  20. Parte S, Bhartiya D, Patel H, Daithankar V, Chauhan A, Zaveri K, et al. Dynamics associated with spontaneous differentiation of ovarian stem cells in vitro. J Ovarian Res. 2014;7:25.

    Article  PubMed Central  PubMed  Google Scholar 

  21. Bhartiya D, Parte S, Patel H, Sriraman K, Zaveri K, and Hinduja I. Novel action of FSH on stem cells in adult mammalian ovary induces postnatal oogenesis and primordial follicle assembly. Stem Cells Int, Article ID 491476, in press.

  22. Bhartiya D, Singh J. FSH-FSHR3-stem cells in ovary surface epithelium: basis for adult ovarian biology, failure, aging, and cancer. Reproduction. 2015;149:R35–48.

    Article  PubMed  Google Scholar 

  23. 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). 2009;1:971–8.

    CAS  Google Scholar 

  24. Bhartiya D, Kasiviswanathan S, Unni SK, Pethe P, Dhabalia JV, Patwardhan S, et al. Newer insights into premeiotic development of germ cells in adult human testis using Oct-4 as a stem cell marker. J Histochem Cytochem. 2010;58:1093–106.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Anand S, Bhartiya D, Sriraman K, Patel H, Manjramkar DD. Very small embryonic-like stem cells survive and restore spermatogenesis after busulphan treatment in mouse testis. J Stem Cell Res Ther. 2014;4:216.

    Google Scholar 

  26. Kucia M, Reca R, Campbell FR, Zuba-Surma E, Majka M, Ratajczak J, et al. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow. Leukemia. 2006;20:857–69.

    Article  CAS  PubMed  Google Scholar 

  27. Ratajczak MZ, Marycz K, Poniewierska-Baran A, Fiedorowicz K, Zbucka-Kretowska M, Moniuszko M. Very small embryonic-like stem cells as a novel developmental concept and the hierarchy of the stem cell compartment. Adv Med Sci. 2014;59:273–80.

    Article  PubMed  Google Scholar 

  28. Miyanishi M, Mori Y, Seita J, Chen JY, Karten S, Chan CKF, et al. Do pluripotent stem cells exist in adult mice as very small embryonic stem cells? Stem Cell Reports. 2013;1:198–208.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Ratajczak MZ, Zuba-Surma E, Wojakowski W, Suszynska M, Mierzejewska K, Liu R, et al. Very small embryonic-like stem cells (VSELs) represent a real challenge in stem cell biology: recent pros and cons in the midst of a lively debate. Leukemia. 2014;28:473–84.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Shin DM, Suszynska M, Mierzejewska K, Ratajczak J, Ratajczak MZ. Very small embryonic-like stem-cell optimization of isolation protocols: an update of molecular signatures and a review of current in vivo applications. Exp Mol Med. 2013;45:e56.

    Article  PubMed Central  PubMed  Google Scholar 

  31. Shaikh A, Nagvenkar P, Pethe P, Hinduja I, Bhartiya D. Molecular and phenotypic characterization of CD133 and SSEA4 enriched very small embryonic-like stem cells in human cord blood. Leukemia. 2015. doi:10.1038/leu.2015.100.

    PubMed  Google Scholar 

  32. Bhartiya D, Shaikh A, Nagvenkar P, Kasiviswanathan S, Pethe P, Pawani H, et al. Very small embryonic-like stem cells with maximum regenerative potential get discarded during cord blood banking and bone marrow processing for autologous stem cell therapy. Stem Cells Dev. 2012;21:1–6.

    Article  CAS  PubMed  Google Scholar 

  33. De Felici M. Germ stem cells in the mammalian adult ovary: considerations by a fan of the primordial germ cells. Mol Hum Reprod. 2010;16:632–6.

    Article  PubMed  Google Scholar 

  34. Ratajczak J, Wysoczynski M, Zuba-Surma E, Wan W, Kucia M, Yoder MC, et al. Adult murine bone marrow-derived very small embryonic-like stem cells differentiate into the hematopoietic lineage after co-culture over OP9 stromal cells. Exp Hematol. 2011;39:225–37.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. Shin DM, Zuba-Surma EK, Wu W, Ratajczak J, Wysoczynski M, Ratajczak MZ, et al. Novel epigenetic mechanisms that control pluripotency and quiescence of adult bone marrow-derived Oct4(+) very small embryonic-like stem cells. Leukemia. 2009;23:2042–51.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science. 2010;327(5965):542–5. doi:10.1126/science.1180794.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  37. De Rosa L, De Luca M. Cell biology: Dormant and restless skin stem cells. Nature. 2012;489(7415):215–7.

    Article  PubMed  Google Scholar 

  38. Parte S, Bhartiya D, Manjramkar DD, Chauhan A, Joshi A. Stimulation of ovarian stem cells by follicle stimulating hormone and basic fibroblast growth factor during cortical tissue culture. J Ovarian Res. 2013;6:20.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Kurkure PA, Prasad M, Dhamankar V, Bakshi G. Very small embryonic-like stem cells (VSELs) detected in azoospermic testicular biopsies of adult survivors of childhood cancer. Reprod Biol Endocrinol. 2015; doi:10.1186/s12958-015-0121-1.

  40. Ignatov AP, Eisenberg MS, Turek PJ. Paternity after directed collection of testicular sperm for in vitro fertilization after BMT for hematological malignancies. Bone Marrow Transplant. 2010;45:1474–6.

    Article  CAS  PubMed  Google Scholar 

  41. Dupont C, Bally C, Eustache F, Sermondade N, Benzacken B, Fenaux P, et al. Spontaneous conception after autologous hematopoietic stem cell transplantation: a case report. Asian J Androl. 2013;15:155–6.

    Article  PubMed Central  PubMed  Google Scholar 

  42. Oktay K, Türkçüoğlu I, Rodriguez Wallberg KA. Four spontaneous pregnancies and three live births following subcutaneous transplantation of frozen banked ovarian tissue: what is the explanation? Fertil Steri. 2011;95(804):e7–e10.

    Google Scholar 

  43. Hermann BP, Sukhwani M, Winkler F, Pascarella JN, Peters KA, Sheng Y, et al. Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm. Cell Stem Cell. 2012;11:715–26.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  44. Struijk RB, Mulder CL, van der Veen F, van Pelt AM, Repping S. Restoring fertility in sterile childhood cancer survivors by auto transplanting spermatogonial stem cells: are we there yet? Biomed Res Int. 2013;2013:903142.

    Article  PubMed Central  PubMed  Google Scholar 

  45. Navaeian Kalat E, Jalali M, Fathi Z. Treating infertility in IVF clinics: Therapeutic potential of stem cells. J Genes Cells. 2015;1:22–8.

    Article  Google Scholar 

  46. Hsiao CH, Ji AT, Chang CC, Cheng CJ, Lee LM, Ho JH. Local injection of mesenchymal stem cells protects testicular torsion-induced germ cell injury. Stem Cell Res Ther. 2015;6:113.

    Article  PubMed Central  PubMed  Google Scholar 

  47. Mehrabani D, Hassanshahi MA, Tamadon A, Zare S, Keshavarz S, Rahmanifar F, et al. Adipose tissue-derived mesenchymal stem cells repair germinal cells of seminiferous tubules of busulfan-induced azoospermic rats. J Hum Reprod Sci. 2015;8:103–10.

    Article  PubMed Central  PubMed  Google Scholar 

  48. Abd El Dayem SM, Arafa MM, Foda FMA, Helal MAM, Zahra FAA, Haggag NZ. Role of bone marrow mesenchymal stem cells (MSCs) on restoration of fertility in male rats after exposure to endocrine disrupter. Int J Pharmacy Rev Res. 2015;5:158–69.

    Google Scholar 

  49. Zahkook SAM, Atwa A, Shahat MM, Mansour AM, Bakry S. Mesenchymal stem cells restore fertility in induced azoospermic rats following chemotherapy administration. J Reproduc Infertil. 2014;5(2):50–7.

    Google Scholar 

  50. Zhang D, Liu X, Peng J, He D, Lin T, Zhu J, et al. Potential spermatogenesis recovery with bone marrow mesenchymal stem cells in an azoospermic rat model. Int J Mol Sci. 2014;15:13151–65.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Cakici C, Buyrukcu B, Duruksu G, Haliloglu AH, Aksoy A, Isık A, et al. Recovery of fertility in azoospermia rats after injection of adipose-tissue-derived mesenchymal stem cells: The sperm generation. Bio Med Res Int. 2013;529589:18. (2013), Article ID.

    Google Scholar 

  52. Monsefi M, Fereydouni B, Rohani L, Talaei T. Mesenchymal stem cells repair germinal cells of seminiferous tubules of sterile rats. Iran J Reprod Med. 2013;11:537–44.

    PubMed Central  PubMed  Google Scholar 

  53. Sabbaghi MA, Bahrami AR, Feizzade B, Kalantar SM, Matin MM, Kalantari M, et al. Trial evaluation of bone marrow derived mesenchymal stem cells (MSCs) transplantation in revival of spermatogenesis in testicular torsion. Middle East Fertil Soc J. 2012;17:243–9.

    Article  Google Scholar 

  54. Aziz AMT, Mostafa T, Atta H, Asaad S, Fouad HH, Mohsen G, Rashed L, Sabry D, Abbas M. In vitro and in vivo lineage conversion of bone marrow stem cells into germ cells in experimental azoospermia in rat. DOI: http://dx.doi.org/ 10.4081/scs.2011.e15

  55. Lassalle B, Mouthon MA, Riou L, Barroca V, Coureuil M, Boussin F, et al. Bone marrow-derived stem cells do not reconstitute spermatogenesis in vivo. Stem Cells. 2008;26(5):1385–6.

    Article  PubMed  Google Scholar 

  56. Lue YH, Erkkila K, Liu PY, Ma K, Wang C, Hikim AS, et al. Fate of bone marrow stem cells transplanted into the testis potential implication for men with testicular failure. Am J Pathol. 2007;170:899–908.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Bhartiya D. Are mesenchymal cells indeed pluripotent stem cells or just stromal cells? OCT-4 and VSELs biology has led to better understanding. Stem Cells Int. 2013;2013:547501.

    PubMed Central  PubMed  Google Scholar 

  58. Donnez J, Dolmans MM, Pellicer A, Diaz-Garcia C, Ernst E, Macklon KT, et al. Fertility preservation for age-related fertility decline. Lancet. 2015;385:506–7.

    Article  PubMed  Google Scholar 

  59. Demeestere I, Simon P, Dedeken L, Moffa F, Tsépélidis S, Brachet C, Delbaere A, Devreker F, Ferster A. Live birth after autograft of ovarian tissue cryopreserved during childhood. Hum Reprod. 2015.[Epub ahead of print]

  60. Chen X. An explanation for the true origin of spontaneous pregnancies after subcutaneous ovarian transplantation. Fertil Steril. 2011;95:e50.

    Article  PubMed  Google Scholar 

  61. Oktay K, Goswami S, Darzynkiewicz Z. Manipulating ovarian aging: a new frontier in fertility preservation. Aging (Albany NY). 2011;3:19–21.

    Google Scholar 

  62. Lei L, Spradling AC. Female mice lack adult germ-line stem cells but sustain oogenesis using stable primordial follicles. Proc Natl Acad Sci U S A. 2013;110:8585–90.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Bhartiya D, Sriraman K, Parte S, Patel H. Ovarian stem cells: Absence of evidence is not evidence of absence. J Ovarian Res. 2013;6:65.

    Article  PubMed Central  PubMed  Google Scholar 

  64. Lee HJ, Selesniemi K, Niikura Y, Niikura T, Klein R, Dombkowski DM, et al. Bone marrow transplantation generates immature oocytes and rescues long-term fertilityin a preclinical mouse model of chemotherapy-induced premature ovarian failure. J ClinOncol. 2007;25:3198–204.

    Article  CAS  Google Scholar 

  65. Edessy M, Hala N, Hosni Y, Wafa S, Shady BK, Kamel M. Stem cells transplantation in premature ovarian failure. World J Med Sci. 2014;10:12–6.

    CAS  Google Scholar 

  66. Zhu SF, Hu HB, Xu HY, Fu XF, Peng DX, Su WY, He YL. Human umbilical cord mesenchymal stem cell transplantation restores damaged ovaries. J Cell Mol Med. 2015. doi:10.2217/fon.15.152

  67. Lai D, Wang F, Yao X, Zhang Q, Wu X, Xiang C. Human endometrial mesenchymal stem cells restore ovarian function through improving the renewal of germline stem cells in a mouse model of premature ovarian failure. J Transl Med. 2015;13:155.

    Article  PubMed Central  PubMed  Google Scholar 

  68. Zhu SF, Hu HB, Xu HY, Fu XF, Peng DX, Su WY, et al. Human umbilical cord mesenchymal stem cell transplantation restores damaged ovaries. J Cell Mol Med. 2015;19(9):2108–17.

    CAS  PubMed Central  PubMed  Google Scholar 

  69. Liu J, Zhang H, Zhang Y, Li N, Wen Y, Cao F, et al. Homing and restorative effects of bone marrow-derived mesenchymal stem cells on cisplatin injured ovaries in rats. Mol Cells. 2014;37(12):865–72.

    Article  PubMed Central  PubMed  Google Scholar 

  70. Liu T, Huang Y, Zhang J, Qin W, Chi H, Chen J, et al. Transplantation of human menstrual blood stem cells to treat premature ovarian failure in mouse model. Stem Cells Dev. 2014;23(13):1548–57.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  71. Terraciano P, Garcez T, Ayres L, Durli I, Baggio M, Kuhl CP, et al. Cell therapy for chemically induced ovarian failure in mice. Stem Cells Int. 2014;2014:720753.

    Article  PubMed Central  PubMed  Google Scholar 

  72. Kilic S, Pinarli F, Ozogul C, Tasdemir N, Naz Sarac G, Delibasi T. Protection from cyclophosphamide-induced ovarian damage with bone marrow-derived mesenchymal stem cells during puberty. Gynecol Endocrinol. 2014;30:135–40.

    Article  CAS  PubMed  Google Scholar 

  73. Abd-Allah SH, Shalaby SM, Pasha HF, El-Shal AS, Raafat N, Shabrawy SM, et al. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy. 2013;15:64–75.

    Article  CAS  PubMed  Google Scholar 

  74. Guo JQ, Gao X, Lin ZJ, Wu WZ, Huang LH, Dong HY, et al. BMSCs reduce rat granulosa cell apoptosis induced by cisplatin and perimenopause. BMC Cell Biol. 2013;14:18.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  75. Ghadami M, El-Demerdash E, Zhang D, Salama SA, Binhazim AA, Archibong AE, et al. Bone marrow transplantation restores follicular maturation and steroid hormones production in a mouse model for primary ovarian failure. PLoS One. 2012;7:e32462.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  76. Fu X, He Y, Xie C, Liu W. Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy. 2008;10(4):353–63.

    Article  CAS  PubMed  Google Scholar 

  77. Veitia RA, Gluckman E, Fellous M, Soulier J. Recovery of female fertility after chemotherapy, irradiation, and bone marrow allograft: further evidence against massive oocyte regeneration by bone marrow-derived germline stem cells. Stem Cells. 2007;25:1334–5.

    Article  PubMed  Google Scholar 

  78. Nabhan SK, Bitencourt MA, Duval M, Abecasis M, Dufour C, Boudjedir K, et al. Fertility recovery and pregnancy after allogeneic hematopoietic stem cell transplantation in Fanconi anemia patients. Haematologica. 2010;95:1783–7.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  79. Anand S, Patel H, Bhartiya D. Chemoablated mouse seminiferous tubular cells enriched for very small embryonic-like stem cells undergo spontaneous spermatogenesis in vitro. Reprod Biol Endocrinol. 2015;13:33.

    Article  PubMed Central  PubMed  Google Scholar 

  80. Bhartiya D, Hinduja I, Patel H, Bhilawadikar R. Making gametes from pluripotent stem cells--a promising role for very small embryonic-like stem cells. Reprod Biol Endocrinol. 2014;12:114.

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

Authors are thankful to Indian Council of Medical Research and Department of Health Research, Government of India, New Delhi for financial support for studies to arrive at this understanding. We also acknowledge all our colleagues working in the field whose work may be relevant but has not been quoted. We also sincerely acknowledge Dr Kalpana Sriraman contribution for her work on chemoablated mouse ovaries. NIRRH Accession number OTH/278/07-2015.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Deepa Bhartiya.

Additional information

Competing interests

Authors declare no conflict of interest.

Authors’ contributions

DB designed studies, reviewed available literature and compiled the article. SA did the experiments on chemoablated mouse testis whereas SP studied stem cells in adult mammalian ovary. Both SA and SP provided significant contributions to prepare the review. Final version was read and approved by all the authors.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bhartiya, D., Anand, S. & Parte, S. VSELs may obviate cryobanking of gonadal tissue in cancer patients for fertility preservation. J Ovarian Res 8, 75 (2015). https://doi.org/10.1186/s13048-015-0199-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13048-015-0199-2

Keywords