Open access peer-reviewed chapter

Mesenchymal Stem Cell Therapy of Male Infertility

Written By

Amin Tamadon, Ulanbek Zhan-byrbekuly, Ilyas Kairgaliyev and Arezoo Khoradmehr

Submitted: 25 March 2019 Reviewed: 02 July 2019 Published: 27 September 2019

DOI: 10.5772/intechopen.88343

From the Edited Volume

Male Reproductive Health

Edited by Wei Wu, Francesco Ziglioli and Umberto Maestroni

Chapter metrics overview

1,208 Chapter Downloads

View Full Metrics

Abstract

Nowadays mesenchymal stem cell (MSC) therapy offers a broad spectrum of treatment of different conditions, including male infertility. Lots of studies suggest that injection of MSC promotes differentiation of germ cells and/or stimulates gonadal tissue development. Currently, there are plenty of MSC therapies of azoospermia which have been studied on animal models and demonstrated good results. Most of the studies were conducted using MSC derived from adipose tissue, the bone marrow, and umbilical cord. Despite the fact that this type of treatment in humans is not established as a first choice option, the use of these techniques gives us hope for a gradual introduction into our daily practice.

Keywords

  • infertility
  • azoospermia
  • stem cell therapy
  • mesenchymal stem cell

1. Introduction

Infertility is a disease of the reproductive system defined by the failure to achieve a clinical pregnancy after 12 months or more of regular unprotected sexual intercourse [1]. This disorder affects one in six couples [2]. Different factors including genetics, environmental factors, and anatomical defects have been illustrated to play a role in the fertility ability of individuals [3, 4].

After identifying the high capability of stem cells to produce different cell types, a number of scientists have proposed the use of stem cells and cell-based therapies as a possible new therapeutic choice for male infertility [5]. Stem cells are undifferentiated cell types that have two main characteristics, self-renewal with the production of identical daughter cells, and the ability to differentiate into more specialized cell types. The two main non-manipulated stem cell classes are embryonic (ESCs) and adult stem cells (ASCs) [5]. The next type of stem cells is induced pluripotent stem cells (iPSCs) which are genetically manipulated somatic cells [6]. The experimental platform for understanding the development of germ cells could be provided by the in vitro generation of male germ cells from stem cells [7, 8].

The ESCs, iPSCs, and spermatogonial stem cells (SSCs) are among the most investigated stem cells for the production of male germ cells in in vitro conditions [9, 10]. Application of these cell types has some limitations. ESCs present with some ethical problems and their sources are limited. iPSCs have both oncological and genetic instabilities. SSCs have low content in the testis, and their isolation, identification, and culturing are difficult in vitro.

On the other hand, mesenchymal stem cells (MSCs) do not have such problems in applications. MSCs are a group of ASCs which are available in most tissues. These cells were separated from the bone marrow [11], adipose tissue [12, 13, 14], hair follicle [15], endometrium [16, 17], dental pulp [18], nose [19], umbilical cord [20], and menstrual blood [21]. The MSCs in bone marrow stromal comprise a restricted area; but it can be easily proliferated [22]. These cells have the potential to proliferate and differentiate into other cells such as osteoblast [23, 24], adipocyte [25], chondroblasts [26], and neuron-like cells [27, 28] which can be a good candidate for treatment of male infertility. MSCs contains heterogeneous population of cells and contain pluripotent stem cells, namely multilineage-differentiating stress-enduring cells, which is the same as ESCs, which has the ability to differentiate into all cells from three germ layers spontaneously [29, 30].

Advertisement

2. Treatment of male infertility by MSC transplantation

Several animal studies have been conducted to investigate the effect of MSC transplantation on azoospermia. The effect of bone marrow-derived MSCs (BM-MSCs) and adipose tissue-derived MSCs (AD-MSCs) in induced azoospermia rodents was explored [11, 12, 24, 26, 31, 32]. After busulfan injection for azoospermia induction [33], the rats were injected with the MSCs into rete testes. After 2 months, testes treated with MSCs appeared morphologically normal. Spermatogenesis was detected, not in every but in some tubules of cell-treated testes. The trans-differentiation of MSCs into spermatogenetic cells in the appropriate microenvironment has been shown in some studies [32]. To demonstrate the entire recovery of spermatogenesis, rats, which were under cell treatment, were mated, and consequently next generations were obtained. The GFP expression was identified in the MSCs derived from the bone marrow and adipose tissue and in the sperm of offsprings as well [32].

Numerous in vivo research surveys have been conducted to assess the spermatogenesis induction potential of mesenchymal stem cells in mouse and rat animal models. In the mentioned study groups, bone marrow-derived mesenchymal stem cells have been used for the induction of spermatogenesis. There are some disputes, in mice model, regarding advisability of bone marrow-derived mesenchymal stem cell transplantation in azoospermic mice, for example, there was a report that bone marrow-derived mesenchymal stem cells could not differentiate into sperm [34], but other studies confirmed the generation of germ cells in vivo in BM-MSC-transplanted mice [35, 36]. At the same time, in azoospermic rat model, BM-MSC allotransplantation amplified endogenous fertility recovery in both testicular torsion model and busulfan-induced model of azoospermia induction [26, 37, 38, 39, 40]. The potential of BM-MSCs to differentiate or trans-differentiate into multi-lineage cells, secrete paracrine factors to recruit the resident stem cells to participate in tissue regeneration, or fuse with the local cells in the affected region has been demonstrated [39].

The next group used AT-MSCs for induction of spermatogenesis. Intra-tubal injection of AT-MSCs in rat model of busulfan-treated azoospermia led to recovery of fertility [12, 32]. The last group of studies induces spermatogenesis using xenotransplantation of human umbilical cord MSCs in immunodeficient mice seminiferous tubule [41] or a combination of in vitro differentiation of induced pluripotent stem cells from mice and humans into germ cells, and also their transplantation was performed to obtain advanced differentiated spermatozoa [42]. Interestingly, the capability of human umbilical cord MSCs (UC-MSCs) for differentiation into germ cells in the lumen of seminiferous tubules of immunocompetent mice has been shown [41, 43]. Furthermore, the therapeutic effects of BM-MSCs against toxic effects of lead (Pb) in the male gonads of rats have been shown [44].

MSC transplantation may induce reconstitution of the tubular microenvironment in azoospermic hamster which helps the remaining inactivated germinal cells to proliferate in the host seminiferous tubules. Sertoli cells play a major role in cooperation with seminiferous tubules, providing cyclic and dynamic regulation of spermatogenesis. In recent time, it has been demonstrated that in in vitro co-culture system, there is an availability of Sertoli cells to mediate differentiation of male germ cell-like cells, which were derived from human umbilical cord mesenchymal stem cells [45]. Moreover, Sertoli cells are considered as immune tolerant cells [46], and they can cause protection and survival of the allotransplanted donor AT-MSCs against inflammatory or immune reaction. In contrast, the hypoimmunogenic character of mesenchymal stem cells makes them appropriate for allogenic transplantation [47]. Nevertheless, mesenchymal stem cells generate immunosuppression or immunosurveillance upon transplantation [48]. It is interesting to mention that, related to the treatment of azoospermia, IV allogenic bone marrow-derived mesenchymal stem cell transfusion encourages the production of antisperm antibody modulated by immune system, after testis rupture in mice [49], which illustrates the other therapeutic potential of mesenchymal stem cells in the treatment of infertility. In fact, the possible mechanisms of azoospermia healing by mesenchymal stem cells are unclear; three main mechanisms could be responsible for the recovering of testicular function during the tissue regeneration process period by mesenchymal stem cells. The first option of mesenchymal stem cells differentiation into the spermatozoa via appropriate induction conditions as it has been demonstrated in rats models [32]. Another mechanism which is not confirmed yet is that secretion of growth factors by mesenchymal stem cells stimulates the restoration of spermatogenesis in the inactivated spermatogonia stem cells or Sertoli cells. And the last one is that mesenchymal stem cells merged with the endogenous spermatogonia stem cells recover the spermatogenesis, which is also needed to be further studied deeply.

The necessity for consideration of another point in cell therapy of azoospermia was illustrated by histomorphometric analysis of the treated seminiferous tubules [31]. The increase of the area of the seminiferous tubules before transplantation caused the decline of the number of tubes per unit area in azoospermia. This pathological condition may be created due to decrease of cellular layers which caused the reduction of the tubal structure and collapsing of several tubules under the pressure of intratubular hydrostatic pressure of hamster seminiferous tubules. The growth of the amount of the spaces in testis may result in rise of the other tubes’ diameter and also decrease in intratubular hydrostatic pressure. It is necessary to mention that the role of this pressure in the mechanism of spermatogenesis has not been clarified, but it could be the one reason for the increase of diameter of cellular layer in mesenchymal stem cell-treated tubes. Next, histomorphometric analysis with the increase of the number of tubules per unit area in mesenchymal stem cells treated tubes has been obtained [31]. This alteration may be caused by the decline of the volumes of intertubular spaces or by the busulfan therapy complications. Moreover, the increase of the whole area of tubes during azoospermia induction could reduce the ability of contraction of contractile myofibroblast cells, in which the decrease of intratubular hydrostatic pressure in peritubular layer probably resulted in the reduction of spermatozoa concentration in epididymis after mesenchymal stem cell therapy.

Advertisement

3. Conclusion

The obtained results of performed research trials on animal models provide a better and deeper overview of MSC therapy in male infertility conditions. The demonstrated results of used options in the treatment of these conditions revealed that methods using the MSCs derived from umbilical cord, adipose tissue, and the bone marrow appear more appropriate to recover the fertility due to better results. Although a case report is available for treatment of azoospermia in man [50], applying those methods into the human practice seems to be investigated before introducing this method into clinic.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Zegers-Hochschild F, Adamson GD, de Mouzon J, Ishihara O, Mansour R, Nygren K, et al. The International Committee for Monitoring Assisted Reproductive Technology (ICMART) and the World Health Organization (WHO) revised glossary on ART terminology, 2009. Human Reproduction. 2009;24(11):2683-2687
  2. 2. Boivin J, Bunting L, Collins JA, Nygren KG. International estimates of infertility prevalence and treatment-seeking: Potential need and demand for infertility medical care. Human Reproduction. 2007;22(6):1506-1512
  3. 3. Miyamoto T, Minase G, Okabe K, Ueda H, Sengoku K. Male infertility and its genetic causes. Journal of Obstetrics and Gynaecology Research. 2015;41(10):1501-1505
  4. 4. Miyamoto T, Tsujimura A, Miyagawa Y, Koh E, Namiki M, Sengoku K. Male infertility and its causes in human. Advances in Urology. 2012;2012:384520
  5. 5. Volarevic V, Bojic S, Nurkovic J, Volarevic A, Ljujic B, Arsenijevic N, et al. Stem cells as new agents for the treatment of infertility: Current and future perspectives and challenges. BioMed Research International. 2014;2014:507234
  6. 6. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663-676
  7. 7. Fang F, Li Z, Zhao Q , Li H, Xiong C. Human induced pluripotent stem cells and male infertility: An overview of current progress and perspectives. Human Reproduction. 2018;33(2):188-195
  8. 8. Valli H, Phillips BT, Shetty G, Byrne JA, Clark AT, Meistrich ML, et al. Germline stem cells: Toward the regeneration of spermatogenesis. Fertility and Sterility. 2014;101(1):3-13
  9. 9. Hou J, Yang S, Yang H, Liu Y, Liu Y, Hai Y, et al. Generation of male differentiated germ cells from various types of stem cells. Reproduction. 2014;147(6):R179-RR88
  10. 10. Nagamatsu G, Hayashi K. Stem cells, in vitro gametogenesis and male fertility. Reproduction. 2017;154(6):F79-F91
  11. 11. Hajihoseini M, Vahdati A, Ebrahim Hosseini S, Mehrabani D, Tamadon A. Induction of spermatogenesis after stem cell therapy of azoospermic guinea pigs. Veterinarski Arhiv. 2017;87(3):333-350
  12. 12. 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. Journal of Human Reproductive Sciences. 2015;8(2):103-110
  13. 13. Mehrabani D, Rabiee M, Tamadon A, Zare S, Jahromi IR, Dianatpour M, et al. The growth kinetic, differentiation properties, karyotyping, and characterization of adipose tissue-derived stem cells in hamster. Comparative Clinical Pathology. 2016;25(5):1017-1022
  14. 14. Shaterzadeh-Yazdi H, Mehrabani D, Khodakaram-Tafti A, Dianatpour M, Zare SH, Tamadon A, et al. Osteogenic potential of subcutaneous adipose-derived stem cells in a rabbit model. Online Journal of Veterinary Research. 2015;19(6):436-445
  15. 15. Liu JY, Peng HF, Gopinath S, Tian J, Andreadis ST. Derivation of functional smooth muscle cells from multipotent human hair follicle mesenchymal stem cells. Tissue Engineering Parts A. 2010;16(8):2553-2564
  16. 16. Mehrabani D, Rahmanifar F, Mellinejad M, Tamadon A, Dianatpour M, Zare S, et al. Isolation, culture, characterization, and adipogenic differentiation of heifer endometrial mesenchymal stem cells. Comparative Clinical Pathology. 2015;24(5):1159-1164
  17. 17. Tamadon A, Mehrabani D, Zarezadeh Y, Rahmanifar F, Dianatpour M, Zare S. Caprine endometrial mesenchymal stromal stem cell: Multi-lineage potential, characterization and growth kinetics in breeding and anestrous stages. Veterinary Medicine International. 2017;2017:5052801
  18. 18. Mehrabani D, Mahdiyar P, Torabi K, Robati R, Zare S, Dianatpour M, et al. Growth kinetics and characterization of human dental pulp stem cells: Comparison between third molar and first premolar teeth. Journal of Clinical and Experimental Dentistry. 2017;9(2):e172-e1e7
  19. 19. Delorme B, Nivet E, Gaillard J, Häupl T, Ringe J, Devèze A, et al. The human nose harbors a niche of olfactory ectomesenchymal stem cells displaying neurogenic and osteogenic properties. Stem Cells and Development. 2009;19(6):853-866
  20. 20. Khodabandeh Z, Vojdani Z, Talaei-Khozani T, Jaberipour M, Hosseini A, Bahmanpour S. Comparison of the expression of hepatic genes by human Wharton’s jelly mesenchymal stem cells cultured in 2D and 3D collagen culture systems. Iranian Journal of Medical Sciences. 2016;41(1):28-36
  21. 21. Mehrabani D, Bahrami Nazarabadi R, Dianatpour M, Vahdati A, Tamadon A, Kasraeian M, et al. Growth kinetics, characterization and plasticity of human menstrual blood stem cells. Iranian Journal of Medical Sciences. 2015;41(2):132-139
  22. 22. Mehrabani D, Khodakaram-Tafti A, Asadi-Yousefabad SL, Dianatpour M, Zare S, Tamadon A, et al. Effect of age and passage on canine bone marrow derived mesenchymal stem cells. Online Journal of Veterinary Research. 2015;19(10):663-671
  23. 23. Khajehahmadi Z, Mehrabani D, Ashraf MJ, Rahmanifar F, Tanideh N, Tamadon A, et al. Healing effect of conditioned media from bone marrow-derived stem cells in thioacetamide-induced liver fibrosis of rat. Journal of Medical Sciences. 2016;16(1-2):7-15
  24. 24. Tamadon A, Mehrabani D, Rahmanifar F, Raayat Jahromi A, Panahi M, Zare S, et al. Induction of spermatogenesis by bone marrow-derived mesenchymal stem cells in busulfan-induced azoospermia in hamster. International Journal of Stem Cells. 2015;8:134-145
  25. 25. Aliborzi G, Vahdati A, Mehrabani D, Ebrahim Hosseini S, Tamadon A. Isolation, characterization and growth kinetic comparison of bone marrow and adipose tissue mesenchymal stem cells of Guinea pig. International Journal of Stem Cells. 2015;9(1):115-123
  26. 26. Rahmanifar F, Tamadon A, Mehrabani D, Zare S, Abasi S, Keshavarz S, et al. Histomorphometric evaluation of treatment of rat azoospermic seminiferous tubules by allotransplantation of bone marrow-derived mesenchymal stem cells. Iranian Journal of Basic Medical Sciences. 2016;19(6):653
  27. 27. Razeghian Jahromi I, Mehrabani D, Mohammadi A, Dianatpour M, Tamadon A, Zare S, et al. The effect of fetal rat brain extract on morphology of bone marrow-derived mesenchymal stem cells. Comparative Clinical Pathology. 2016;25(2):343-349
  28. 28. Razeghian Jahromi I, Mehrabani D, Mohammadi A, Ghahremani Seno MM, Dianatpour M, Zare S, et al. Emergence of signs of neural cells after exposure of bone marrow-derived mesenchymal stem cells to fetal brain extract. Iranian Journal of Basic Medical Sciences. 2017;20:301-307
  29. 29. Dezawa M. Muse cells provide the pluripotency of mesenchymal stem cells: Direct contribution of muse cells to tissue regeneration. Cell Transplantation. 2016;25(5):849-861
  30. 30. Kuroda Y, Wakao S, Kitada M, Murakami T, Nojima M, Dezawa M. Isolation, culture and evaluation of multilineage-differentiating stress-enduring (Muse) cells. Nature Protocols. 2013;8(7):1391
  31. 31. Karimaghai N, Tamadon A, Rahmanifar F, Mehrabani D, Jahromi AR, Zare S, et al. Spermatogenesis after transplantation of adipose tissue-derived mesenchymal stem cells in busulfan-induced azoospermic hamster. Iranian Journal of Basic Medical Sciences. 2018;21(7):660
  32. 32. 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. BioMed Research International. 2013;2013:529589
  33. 33. Panahi M, Keshavarz S, Rahmanifar F, Tamadon A, Mehrabani D, Karimaghai N, et al. Busulfan induced azoospermia: Stereological evaluation of testes in rat. Veterinary Research Forum. 2015;6(4):273
  34. 34. Van Saen D, Goossens E, De Block G, Tournaye H. Bone marrow stem cells transplanted to the testis of sterile mice do not differentiate into spermatogonial stem cells and have no protective effect on fertility. Fertility and Sterility. 2009;91(4):1549-1552
  35. 35. Yang S, Bo J, Hu H, Guo X, Tian R, Sun C, et al. Derivation of male germ cells from induced pluripotent stem cells in vitro and in reconstituted seminiferous tubules. Cell Proliferation. 2012;45(2):91-100
  36. 36. 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. The American Journal of Pathology. 2007;170(3):899-908
  37. 37. Monsefi M, Fereydouni B, Rohani L, Talaei T. Mesenchymal stem cells repair germinal cells of seminiferous tubules of sterile rats. Iranian Journal of Reproductive Medicine. 2013;11(7):537
  38. 38. Zahkook SA, Atwa A, Shahat M, Mansour AM, Bakry S. Mesenchymal stem cells restore fertility in induced azoospermic rats following chemotherapy administration. Journal of Reproduction and Infertility. 2014;5(2):50-57
  39. 39. 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. International Journal of Molecular Sciences. 2014;15(8):13151-13165
  40. 40. 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 Fertility Society Journal. 2012;17(4):243-249
  41. 41. Chen H, Tang QL, Wu XY, Xie LC, Lin LM, Ho GY, et al. Differentiation of human umbilical cord mesenchymal stem cells into germ-like cells in mouse seminiferous tubules. Molecular Medicine Reports. 2015;12(1):819-828
  42. 42. Zhu Y, Hu H-L, Li P, Yang S, Zhang W, Ding H, et al. Generation of male germ cells from induced pluripotent stem cells (iPS cells): An in vitro and in vivo study. Asian Journal of Andrology. 2012;14(4):574-579
  43. 43. Allah SHA, Pasha HF, Abdelrahman AA, Mazen NF. Molecular effect of human umbilical cord blood CD34-positive and CD34-negative stem cells and their conjugate in azoospermic mice. Molecular and Cellular Biochemistry. 2017;428(12):179-191
  44. 44. Hassan AI, Alam SS. Evaluation of mesenchymal stem cells in treatment of infertility in male rats. Stem Cell Research & Therapy. 2014;5(6):131
  45. 45. Xie L, Lin L, Tang Q , Li W, Huang T, Huo X, et al. Sertoli cell-mediated differentiation of male germ cell-like cells from human umbilical cord Wharton’s jelly-derived mesenchymal stem cells in an in vitro co-culture system. European Journal of Medical Research. 2015;20(1):9
  46. 46. Mital P, Kaur G, Dufour JM. Immunoprotective sertoli cells: Making allogeneic and xenogeneic transplantation feasible. Reproduction. 2010;139(3):495-504
  47. 47. Ryan JM, Barry FP, Murphy JM, Mahon BP. Mesenchymal stem cells avoid allogeneic rejection. Journal of Inflammation. 2005;2:8
  48. 48. Bibber B, Sinha G, Lobba ARM, Greco SJ, Rameshwar P. A review of stem cell translation and potential confounds by cancer stem cells. Stem Cells International. 2013;2013:8, Article ID: 241048
  49. 49. Aghamir SMK, Salavati A, Yousefie R, Tootian Z, Ghazaleh N, Jamali M, et al. Does bone marrow-derived mesenchymal stem cell transfusion prevent antisperm antibody production after traumatic testis rupture? Urology. 2014;84(1):82-86
  50. 50. Cassim MI, Mohamed T. A novel therapy for the treatment of malefactor infertility due to non-obstructive azoospermia: A case report. Crescent Journal of Medical and Biological Sciences. 2019;6(1):129-131

Written By

Amin Tamadon, Ulanbek Zhan-byrbekuly, Ilyas Kairgaliyev and Arezoo Khoradmehr

Submitted: 25 March 2019 Reviewed: 02 July 2019 Published: 27 September 2019