Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2054 2022-12-22 17:52:37 |
2 format correct + 16 word(s) 2070 2022-12-23 04:49:28 | |
3 format correct + 7 word(s) 2077 2022-12-23 04:54:16 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Rodríguez-Eguren, A.;  Gómez-Álvarez, M.;  Francés-Herrero, E.;  Romeu, M.;  Ferrero, H.;  Seli, E.;  Cervelló, I. Application of Umbilical Cord Stem Cells in Ovary. Encyclopedia. Available online: https://encyclopedia.pub/entry/39113 (accessed on 03 May 2024).
Rodríguez-Eguren A,  Gómez-Álvarez M,  Francés-Herrero E,  Romeu M,  Ferrero H,  Seli E, et al. Application of Umbilical Cord Stem Cells in Ovary. Encyclopedia. Available at: https://encyclopedia.pub/entry/39113. Accessed May 03, 2024.
Rodríguez-Eguren, Adolfo, María Gómez-Álvarez, Emilio Francés-Herrero, Mónica Romeu, Hortensia Ferrero, Emre Seli, Irene Cervelló. "Application of Umbilical Cord Stem Cells in Ovary" Encyclopedia, https://encyclopedia.pub/entry/39113 (accessed May 03, 2024).
Rodríguez-Eguren, A.,  Gómez-Álvarez, M.,  Francés-Herrero, E.,  Romeu, M.,  Ferrero, H.,  Seli, E., & Cervelló, I. (2022, December 22). Application of Umbilical Cord Stem Cells in Ovary. In Encyclopedia. https://encyclopedia.pub/entry/39113
Rodríguez-Eguren, Adolfo, et al. "Application of Umbilical Cord Stem Cells in Ovary." Encyclopedia. Web. 22 December, 2022.
Application of Umbilical Cord Stem Cells in Ovary
Edit

Stem cell (SC) therapies have been investigated as alternative treatment strategies. Human umbilical cord (hUC) mesenchymal stem cells (hUC-MSC), along with their secreted paracrine factors, extracts, and biomolecules, have emerged as promising therapeutic alternatives in regenerative medicine, due to their remarkable potential to promote anti-inflammatory and regenerative processes more efficiently than other autologous treatments. Infertility is a disease of the male or female reproductive system, defined by the World Health Organization as the failure to achieve a pregnancy, after at least 12 months of regular unprotected sexual intercourse. Female infertility can be caused by various disorders of the reproductive system, including premature ovarian insufficiency (POI), polycystic ovary syndrome (PCOS), endometriosis, Asherman’s syndrome (AS), or endometrial atrophy (EA), among others.

ovary umbilical cord stem cell

1. Cellular Therapies Based on Human umbilical cord (hUC) mesenchymal stem cells: Current Applications, Administration, and Fertility Restoration

Within the female reproductive tract, the ovary is the organ most commonly treated with human umbilical cord (hUC) cells, especially MSCs. The main reports of the use of human umbilical cord (hUC) mesenchymal stem cells (hUC-MSCs) and UC blood derivatives were included in Table 1. In 2013, Wang et al. [1] pioneered the use of hUC-MSCs to treat POI in mice, restoring ovarian function, serum estrogen levels, adequate follicle development, and notably reducing cell apoptosis.
Similar to how other cell therapies have been infused in rodents [2], hUC-MSCs have been administered via tail vein injection, to foster their natural migration towards the injured site [3][4][5][6][7][8][9][10][11]. This strategy not only improved ovarian morphology, primordial follicle development, and hormone production, but also restored estrous cyclicity [3][5][7][10], implying ovarian performance was recovered. More importantly, systemic treatment with hUC-MSCs restored fertility in all cases [5][9][11]. Furthermore, hUC-MSC therapy provoked ovarian secretion of cytokines such as HGF, VEGF, and insulin growth factor 1 (IGF1), suggesting ovarian regeneration was mediated by paracrine mechanisms, and these factors could improve the ovarian function and delay ovarian senescence [4]. Using metabolomics, Zhao et al. [5] showed that hUC-MSCs activated the phosphoinositide 3-kinase (PI3K) pathway by stimulating synthesis of free amino acids, improved lipid metabolism, and decreased concentration of monosaccharides [5]. Meanwhile, Lu’s group [6] also focused on the protective effect of the hUC-MSCs on the theca-interstitial cells. In vitro, they observed an interesting reduction in autophagy levels in the theca-interstitial cells, induced by the decrease in oxidative stress and regulation of the AMP-activated protein kinase (AMPK)/Mammalian target of rapamycin (mTOR) signaling pathway. Augmented tropomyosin receptor kinase A (TrkA) and nerve growth factor (NGF) following hUC-MSC treatment was also reported by Zheng et al. [3], corroborating the involvement of the NGF/TrkA signaling pathway in ovarian regeneration. Moreover, the efficacy of multiple intravenous injections in a chemotherapy-induced POI mouse model was analyzed by Lv et al. [11]. After introducing cells, the ovarian morphology, follicle count, and fertility improved, regardless of whether mice received a single or triple administration. However, higher levels of serum anti-Müllerian hormone (AMH) and local Ki67 expression suggested that multiple doses of hUC-MSCs have a superior therapeutic effect than a single hUC-MSC bolus.
Alternatively, local intraovarian injections were also reported [12][13][14], and were capable of restoring follicle development and fertility in rodents. Interestingly, Shi et al. [13] analyzed the toxicity associated with injecting increasing doses of hUC-MSCs. The maximum tolerated dose was 106 cells/ovary, with severe toxicity and lethality observed after exceeding this limit. Meanwhile, Pan et al. [14] found that hUC-MSC and amniotic MSC treatment, comparably restored damaged ovarian morphology, functionality, elasticity, and toughness. Finally, intraperitoneal treatment with hUC-MSCs showed similar results in terms of follicle populations, hormone regulation, and fertility restoration [15].
Despite these promising findings, administration routes for hUC-MSCs remain controversial. Three independent groups compared the efficacy of intravenous or local administration for POI models [16][17][18]. In these studies, both routes reduced cell apoptosis, augmented the proportion of growing follicles, and restored the ovarian function (as determined by regulated hormonal levels and recuperated estrous cyclicity). On the other hand, Song’s group [17] did not find any differences, Zhang et al. [18] reasoned there was a better restoration of the ovarian function after intravenous treatment, and Zhu et al. [16] found that local injection was more efficient. In terms of ovarian aging models, Zhang et al. [19] observed the positive effects of hUC-MSCs regardless of the injection route (intravenous or intraovarian), with increased follicle development, restored fertility, and reduced apoptosis of granulosa cells and reactive oxygen species (ROS) production. Finally, in humans, ovarian injection of hUC-MSCs rescued ovarian function, increased follicle development, and ultimately, led to live births [20].
Recently, Lu et al. [21] elegantly studied the impact of hUC-MSCs throughout the ovary and the endometrium. The SCs were introduced via tail injection, two weeks after inducing a POI condition. They reported improved ovarian morphology and folliculogenesis, as well as restored serum hormone levels. Regarding the endometrium, an increased number of glands and enhanced neoangiogenesis were observed, in addition to a noticeable overexpression of Homeobox A10 (HOXA10, an essential regulator of endometrial decidualization [22]), altogether revealing improved morphology and functionality. Furthermore, the type 1/2 T helper (Th1/Th2) cell ratio, and expression of NK cells, significantly decreased in the endometrium, suggesting treatment significantly regulated the ovarian function and endometrial receptivity. Likewise, Aygün et al. [23] loaded hUC-MSCs in hyaluronic acid scaffolds, and transplanted intraperitoneally in an abdominal adhesion rat model, and demonstrated the synergistic effect of the SCs on restoring hormone levels, improving follicle development and endometrial angiogenesis (after reducing the adhesions macroscopically).
While hyaluronic acid is the most popular scaffold used to develop alternative treatments in murine models of ovarian pathologies [24][25], other synthetic scaffolds have been mixed with biological products to enhance their pharmacodynamics [26]. Notably, only Sun’s group [27] has translated the use of collagen scaffolds from mice [28] to humans. Nevertheless, in mice, both hyaluronic acid and collagen scaffolds sustained the effects of the hUC-MSCs, improving ovarian morphology, cell proliferation, angiogenesis, and ultimately, restoring fertility [24][25][28]. Likewise, women treated with the hUC-MSC-collagen complex achieved a complete restoration of fertility, with primordial follicle activation (determined by the expression of phosphorylation of Forkhead box O3a (FOXO3a) and Forkhead box O1 (FOXO 1)), increased proportion of growing follicles, and consequent high serum estradiol concentration [27].

2. Emerging Alternatives: Acellular Therapies

2.1. Extracellular Vesicles

When hUC-MSCs are isolated and applied for regenerative treatments, the bioactive executors of hUC-MSCs effects (including EVs) remain present in the cell secretome [29]. Based on this premise, independent groups have employed the medium collected from hUC-MSC culture as a reparative treatment for POI models [24][30], and found it increased AMH levels and folliculogenesis.
Specifically, several studies have described that hUC-MSCs secrete EVs, such as exosomes and microvesicles, to deliver biomolecules (i.e., lipids, carbohydrates, nucleic acids, and proteins) that facilitate cellular and host cell reprogramming [31][32]. Consistent with the findings of two other independent groups [33][34], Liu et al. [35] reported that administering EVs, derived from the hUC, through the tail vein efficiently improved fertility of mice with POI, by improving ovarian and follicle morphology, balancing hormone levels, returning estrous cyclicity, and reducing apoptosis.
The intraperitoneal transplantation of exosomes also proved beneficial in mice, by returning folliculogenesis and hormones to nearly normal levels [36]. This improvement in reproductive outcomes was associated with the regulation of the Hippo pathway, which is critical for regulating follicle activation and survival, and thus, ovarian function [37]. Furthermore, Ding et al. [38] demonstrated, in a murine model, that exosomal miRNA-17-5p was strongly associated with improvements in ovarian function. Indeed, the inhibition of this miRNA via a knockdown approach in hUC-MSCs diminished the regenerative effects of the exosomes.

2.2. Growth Factors

The hUC, and its cells, actively secrete GFs that can potentially be used for regenerative applications, since they stimulate cell proliferation and differentiation [39]. Specifically, the hUC-MSCs are reservoirs that, in response to specific regenerative signals in their microenvironment, release molecules to promote tissue repair. In fact, Wang et al. [40] reported that intraperitoneal treatment with GM-CSFs (which are present in the hUC-MSC secretome and immunomodulate hematopoietic cells [40]) promoted follicle development (as validated through the expression of folliculogenesis-related biomarkers) [41]. Similarly, other GFs, such as HGF (enriched in the hUC-MSC secretome) [25], or EGF (loaded into collagen-based scaffolds as Matrigel®) [42] have also been associated with positive fertility outcomes.

2.3. Plasma and Platelet-Rich Plasma

In terms of ovarian regeneration, Buigues et al. [43] compared the effect of hUC plasma (rich in many GFs) to that of mobilizing G-CSF treatment and demonstrated that both therapies increased the populations of growing follicles, proliferation, and angiogenesis, in addition to restoring fertility of mice with gonadotoxic damage. Furthermore, Wang et al. [44] observed the ample benefits of hUC-PRP treatment in a murine model of POI, including preserved ovarian morphology, regulated hormonal levels and estrous cyclicity, increased angiogenesis, and reduced apoptosis, with respect to the untreated groups.

References

  1. Wang, S.; Yu, L.; Sun, M.; Mu, S.; Wang, C.; Wang, D.; Yao, Y. The Therapeutic Potential of Umbilical Cord Mesenchymal Stem Cells in Mice Premature Ovarian Failure. Biomed Res. Int. 2013, 2013, 690491.
  2. Na, J.; Kim, G.J. Recent Trends in Stem Cell Therapy for Premature Ovarian Insufficiency and Its Therapeutic Potential: A Review. J. Ovarian Res. 2020, 13, 74.
  3. Zheng, Q.; Fu, X.; Jiang, J.; Zhang, N.; Zou, L.; Wang, W.; Ding, M.; Chen, H. Umbilical Cord Mesenchymal Stem Cell Transplantation Prevents Chemotherapy-Induced Ovarian Failure via the NGF/TrkA Pathway in Rats. Biomed Res. Int. 2019, 2019, 6539294.
  4. Li, J.; Mao, Q.X.; He, J.J.; She, H.Q.; Zhang, Z.; Yin, C.Y. Human Umbilical Cord Mesenchymal Stem Cells Improve the Reserve Function of Perimenopausal Ovary via a Paracrine Mechanism. Stem Cell Res. Ther. 2017, 8, 55.
  5. Zhao, Y.; Ma, J.; Yi, P.; Wu, J.; Zhao, F.; Tu, W.; Liu, W.; Li, T.; Deng, Y.; Hao, J.; et al. Human Umbilical Cord Mesenchymal Stem Cells Restore the Ovarian Metabolome and Rescue Premature Ovarian Insufficiency in Mice. Stem Cell Res. Ther. 2020, 11, 466.
  6. Lu, X.; Bao, H.; Cui, L.; Zhu, W.; Zhang, L.; Xu, Z.; Man, X.; Chu, Y.; Fu, Q.; Zhang, H. HUMSC Transplantation Restores Ovarian Function in POI Rats by Inhibiting Autophagy of Theca-Interstitial Cells via the AMPK/MTOR Signaling Pathway. Stem Cell Res. Ther. 2020, 11, 268.
  7. Shen, J.; Cao, D.; Sun, J.-L. Ability of Human Umbilical Cord Mesenchymal Stem Cells to Repair Chemotherapy-Induced Premature Ovarian Failure. World J. Stem Cells 2020, 12, 277–287.
  8. Jalalie, L.; Rezaie, M.J.; Jalili, A.; Rezaee, M.A.; Vahabzadeh, Z.; Rahmani, M.R.; Karimipoor, M.; Hakhamaneshi, M.S. Distribution of the CM-Dil-Labeled Human Umbilical Cord Vein Mesenchymal Stem Cells Migrated to the Cyclophosphamide-Injured Ovaries in C57BL/6 Mice. Iran Biomed J. 2019, 23, 200–208.
  9. Zhang, L.; Sun, Y.; Zhang, X.-X.; Liu, Y.-B.; Sun, H.-Y.; Wu, C.-T.; Xiao, F.-J.; Wang, L.-S. Comparison of CD146 +/− Mesenchymal Stem Cells in Improving Premature Ovarian Failure. Stem Cell Res. Ther. 2022, 13, 267.
  10. Wang, Z.; Wei, Q.; Wang, H.; Han, L.; Dai, H.; Qian, X.; Yu, H.; Yin, M.; Shi, F.; Qi, N. Mesenchymal Stem Cell Therapy Using Human Umbilical Cord in a Rat Model of Autoimmune-Induced Premature Ovarian Failure. Stem Cells Int. 2020, 2020, 3249495.
  11. Lv, X.; Guan, C.; Li, Y.; Su, X.; Zhang, L.; Wang, X.; Xia, H.F.; Ma, X. Effects of Single and Multiple Transplantations of Human Umbilical Cord Mesenchymal Stem Cells on the Recovery of Ovarian Function in the Treatment of Premature Ovarian Failure in Mice. J. Ovarian Res. 2021, 14, 119.
  12. Mohamed, S.A.; Shalaby, S.; Brakta, S.; Elam, L.; Elsharoud, A.; Al-Hendy, A. Umbilical Cord Blood Mesenchymal Stem Cells as an Infertility Treatment for Chemotherapy Induced Premature Ovarian Insufficiency. Biomedicines 2019, 7, 7.
  13. Shi, L.; Zhang, Y.; Dong, X.; Pan, Y.; Ying, H.; Chen, J.; Yang, W.; Zhang, Y.; Fei, H.; Liu, X.; et al. Toxicity from a Single Injection of Human Umbilical Cord Mesenchymal Stem Cells into Rat Ovaries. Reprod. Toxicol. 2022, 110, 9–18.
  14. Pan, Y.; Zhang, L.; Zhang, X.; Hu, C.; Liu, R. Biological and Biomechanical Analysis of Two Types of Mesenchymal Stem Cells for Intervention in Chemotherapy-Induced Ovarian Dysfunction. Arch. Gynecol. Obstet. 2016, 295, 247–252.
  15. Cui, L.; Bao, H.; Liu, Z.; Man, X.; Liu, H.; Hou, Y.; Luo, Q.; Wang, S.; Fu, Q.; Zhang, H. HUMSCs Regulate the Differentiation of Ovarian Stromal Cells via TGF-Β1/Smad3 Signaling Pathway to Inhibit Ovarian Fibrosis to Repair Ovarian Function in POI Rats. Stem Cell Res. Ther. 2020, 11, 386.
  16. Zhu, S.F.; Hu, H.B.; Xu, H.Y.; Fu, X.F.; Peng, D.X.; Su, W.Y.; He, Y.L. Human Umbilical Cord Mesenchymal Stem Cell Transplantation Restores Damaged Ovaries. J. Cell Mol. Med. 2015, 19, 2108–2117.
  17. Li, H.; Song, D.; Zhong, Y.; Qian, C.; Zou, Q.; Ou, J.; Shi, Y.; Gao, L.; Wang, G.; Liu, Z.; et al. Human Umbilical Cord Mesenchymal Stem Cells Therapy in Cyclophosphamide-Induced Premature Ovarian Failure Rat Model. Biomed Res. Int. 2016, 2016, 2517514.
  18. Zhang, M.; Xie, T.; Dai, W.; Zhao, B.; Zheng, Y.; Hu, J.; Pan, R.; Wang, L. Umbilical Cord Mesenchymal Stem Cells Ameliorate Premature Ovarian Insufficiency in Rats. Evid.-Based Complementary Altern. Med. 2022, 2022, 9228456.
  19. Zhang, J.; Xiong, J.; Fang, L.; Lu, Z.; Wu, M.; Shi, L.; Qin, X.; Luo, A.; Wang, S. The Protective Effects of Human Umbilical Cord Mesenchymal Stem Cells on Damaged Ovarian Function: A Comparative Study. Biosci. Trends 2016, 10, 265–276.
  20. Yan, L.; Wu, Y.; Li, L.; Wu, J.; Zhao, F.; Gao, Z.; Liu, W.; Li, T.; Fan, Y.; Hao, J.; et al. Clinical Analysis of Human Umbilical Cord Mesenchymal Stem Cell Allotransplantation in Patients with Premature Ovarian Insufficiency. Cell Prolif. 2020, 53, e12938.
  21. Lu, X.; Cui, J.; Cui, L.; Luo, Q.; Cao, Q.; Yuan, W.; Zhang, H. The Effects of Human Umbilical Cord-Derived Mesenchymal Stem Cell Transplantation on Endometrial Receptivity Are Associated with Th1/Th2 Balance Change and UNK Cell Expression of Uterine in Autoimmune Premature Ovarian Failure Mice. Stem Cell Res. Ther. 2019, 10, 214.
  22. Ramathal, C.Y.; Bagchi, I.C.; Taylor, R.N.; Bagchi, M.K. Endometrial Decidualization: Of Mice and Men. Semin. Reprod. Med. 2010, 28, 17–26.
  23. Aygün, E.G.; Tümentemur, G. Effects of Stem Cells and Amniotic Fluid on Uterus and Ovaries on a Rat Model with Abdominal Adhesions: A Controlled Study. J. Turk. Ger. Gynecol. Assoc. 2022, 23, 154–166.
  24. Jiao, W.; Mi, X.; Yang, Y.; Liu, R.; Liu, Q.; Yan, T.; Chen, Z.J.; Qin, Y.; Zhao, S. Mesenchymal Stem Cells Combined with Autocrosslinked Hyaluronic Acid Improve Mouse Ovarian Function by Activating the PI3K-AKT Pathway in a Paracrine Manner. Stem Cell Res. Ther. 2022, 13, 49.
  25. Mi, X.; Jiao, W.; Yang, Y.; Qin, Y.; Chen, Z.J.; Zhao, S. HGF Secreted by Mesenchymal Stromal Cells Promotes Primordial Follicle Activation by Increasing the Activity of the PI3K-AKT Signaling Pathway. Stem Cell Rev. Rep. 2022, 18, 1834–1850.
  26. Francés-Herrero, E.; Lopez, R.; Hellström, M.; de Miguel-Gómez, L.; Herraiz, S.; Brännström, M.; Pellicer, A.; Cervelló, I. Bioengineering Trends in Female Reproduction: A Systematic Review. Hum. Reprod. Update 2022, 28, 798–837.
  27. Ding, L.; Yan, G.; Wang, B.; Xu, L.; Gu, Y.; Ru, T.; Cui, X.; Lei, L.; Liu, J.; Sheng, X.; et al. Transplantation of UC-MSCs on Collagen Scaffold Activates Follicles in Dormant Ovaries of POF Patients with Long History of Infertility. Sci. China Life Sci. 2018, 61, 1554–1565.
  28. Yang, Y.; Lei, L.; Wang, S.; Sheng, X.; Yan, G.; Xu, L.; Liu, J.; Liu, M.; Zhen, X.; Ding, L.; et al. Transplantation of Umbilical Cord–Derived Mesenchymal Stem Cells on a Collagen Scaffold Improves Ovarian Function in a Premature Ovarian Failure Model of Mice. In Vitro Cell Dev. Biol. Anim. 2019, 55, 302–311.
  29. Liu, F.; Hu, S.; Yang, H.; Li, Z.; Huang, K.; Su, T.; Wang, S. Hyaluronic Acid Hydrogel Integrated with Mesenchymal Stem Cell-Secretome to Treat Endometrial Injury in a Rat Model of Asherman’s Syndrome. Adv. Healthc. Mater. 2019, 8, 1900411.
  30. Hong, L.; Yan, L.; Xin, Z.; Hao, J.; Liu, W.; Wang, S.; Liao, S.; Wang, H.; Yang, X. Protective Effects of Human Umbilical Cord Mesenchymal Stem Cell-Derived Conditioned Medium on Ovarian Damage. J. Mol. Cell Biol. 2020, 12, 372–385.
  31. Jiao, Y.; Li, X.Y.; Liu, J. A New Approach to Cerebral Palsy Treatment: Discussion of the Effective Components of Umbilical Cord Blood and Its Mechanisms of. Cell Transplant. 2019, 28, 497.
  32. Yaghoubi, Y.; Movassaghpour, A.A.; Zamani, M.; Talebi, M.; Mehdizadeh, A.; Yousefi, M. Human Umbilical Cord Mesenchymal Stem Cells Derived-Exosomes in Diseases Treatment. Life Sci. 2019, 233, 116733.
  33. Yang, W.; Zhang, J.; Xu, B.; He, Y.; Liu, W.; Li, J.; Zhang, S.; Lin, X.; Su, D.; Wu, T.; et al. HucMSC-Derived Exosomes Mitigate the Age-Related Retardation of Fertility in Female Mice. Mol. Ther. 2020, 28, 1200–1213.
  34. Yang, Z.; Du, X.; Wang, C.; Zhang, J.; Liu, C.; Li, Y.; Jiang, H. Therapeutic Effects of Human Umbilical Cord Mesenchymal Stem Cell-Derived Microvesicles on Premature Ovarian Insufficiency in Mice. Stem Cell Res. Ther. 2019, 10, 250.
  35. Liu, C.; Yin, H.; Jiang, H.; Du, X.; Wang, C.; Liu, Y.; Li, Y.; Yang, Z. Extracellular Vesicles Derived from Mesenchymal Stem Cells Recover Fertility of Premature Ovarian Insufficiency Mice and the Effects on Their Offspring. Cell Transplant. 2020, 29, 1–11.
  36. Li, Z.; Zhang, M.; Zheng, J.; Tian, Y.; Zhang, H.; Tan, Y.; Li, Q.; Zhang, J.; Huang, X. Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Improve Ovarian Function and Proliferation of Premature Ovarian Insufficiency by Regulating the Hippo Signaling Pathway. Front. Endocrinol. (Lausanne) 2021, 12, 996.
  37. Gershon, E.; Dekel, N. Newly Identified Regulators of Ovarian Folliculogenesis and Ovulation. Int. J. Mol. Sci. 2020, 21, 4565.
  38. Ding, C.; Zhu, L.; Shen, H.; Lu, J.; Zou, Q.; Huang, C.; Li, H.; Huang, B. Exosomal MiRNA-17-5p Derived from Human Umbilical Cord Mesenchymal Stem Cells Improves Ovarian Function in Premature Ovarian Insufficiency by Regulating SIRT7. Stem Cells 2020, 38, 1137–1148.
  39. Velarde, F.; Castañeda, V.; Morales, E.; Ortega, M.; Ocaña, E.; Álvarez-Barreto, J.; Grunauer, M.; Eguiguren, L.; Caicedo, A. Use of Human Umbilical Cord and Its Byproducts in Tissue Regeneration. Front. Bioeng. Biotechnol. 2020, 8, 117.
  40. Braunstein, S.; Kaplan, G.; Gottlieb, A.B.; Schwartz, M.; Walshs, G.; Abalos, R.M.; Fajardo, T.T.; Guido, L.S.; Krueger, J.G. GM-CSF Activates Regenerative Epidermal Growth and Stimulates Keratinocyte Proliferation in Human Skin in Vivo. J. Investig. Dermatol. 1994, 103, 601–604.
  41. Wang, H.; Wen, Y.; Polan, M.L.; Boostanfar, R.; Feinman, M.; Behr, B. Exogenous Granulocyte–Macrophage Colony-Stimulating Factor Promotes Follicular Development in the Newborn Rat in Vivo. Hum. Reprod. 2005, 20, 2749–2756.
  42. Zhang, J.; Yan, L.; Wang, Y.; Zhang, S.; Xu, X.; Dai, Y.; Zhao, S.; Li, Z.; Zhang, Y.; Xia, G.; et al. In Vivo and in Vitro Activation of Dormant Primordial Follicles by EGF Treatment in Mouse and Human. Clin. Transl. Med. 2020, 10, e182.
  43. Buigues, A.; Marchante, M.; de Miguel-Gómez, L.; Martinez, J.; Cervelló, I.; Pellicer, A.; Herraiz, S. Stem Cell-Secreted Factor Therapy Regenerates the Ovarian Niche and Rescues Follicles. Am. J. Obstet. Gynecol. 2021, 225, 65.e1–65.e14.
  44. Wang, J.; Zhao, Y.; Zheng, F.; Ma, N.; Qin, R.; Qin, W.; Liu, B.; Qin, A. Activated Human Umbilical Cord Blood Platelet-Rich Plasma Enhances the Beneficial Effects of Human Umbilical Cord Mesenchymal Stem Cells in Chemotherapy-Induced POF Rats. Stem Cells Int. 2021, 2021, 8293699.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 337
Revisions: 3 times (View History)
Update Date: 23 Dec 2022
1000/1000