Log in

In vitro effect of granulocyte–macrophage colony-stimulating factor (GM-CSF) on the expression of genes related to sperm motility and energy metabolism and intracytoplasmic sperm injection outcomes in obstructive azoospermic patients

  • Original Article
  • Published:
Molecular Biology Reports Aims and scope Submit manuscript

Abstract

Background

The presence of granulocyte–macrophage colony-stimulating factor (GM-CSF) and its receptor in various testicular cells and spermatozoa suggests a potential role in enhancing spermatogonial and postmeiotic cell development. Moreover, GM-CSF activates the pivotal pathways implicated in sperm motility regulation and glucose metabolism. However, the impact of GM-CSF on testicular biopsies from patients with obstructive azoospermia (OA) remains unexplored. Therefore, this study aimed to investigate the in vitro effects of GM-CSF on the expression of genes related to glucose transporters and signaling pathways, sperm motility, and viability in testicular biopsies.

Methods and results

Following testicular sperm extraction from 20 patients diagnosed with OA, each sample was divided into two parts: the experimental samples were incubated with medium containing 2 ng/ml GM-CSF at 37 °C for 60 min, and the control samples were incubated with medium without GM-CSF. Subsequently, the oocytes retrieved from the partner were injected with sperm from the treatment and control groups. The sperm parameters (motility and viability), the expression levels of sperm motility-related genes (PIK3R1, PIK3CA, and AKT1), and the expression levels of sperm energy metabolism-related genes (GLUT1, GLUT3, and GLUT14) were assessed. Furthermore, the fertilization and day 3 embryo development rate and embryo quality were evaluated. Compared with those in the nontreated group, the motility parameters and the mRNA expression levels of PIK3R1, AKT1, and GLUT3 in testicular sperm supplemented with GM-CSF were significantly greater (p < 0.05). However, no significant differences in the mRNA expression of PIK3CA, GLUT1, or GLUT14 were detected. According to the ICSI results, compared with the control group, the GM-CSF treatment group exhibited significantly greater fertilization rates (p = 0.027), Day 3 embryo development rate (p = 0.001), and proportions of good-quality embryos (p = 0.002).

Conclusions

GM-CSF increased the expression of genes related to motility and the energy metabolism pathway and effectively promoted the motility of testis-extracted spermatozoa, consequently yielding positive clinical outcomes.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price includes VAT (France)

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

GM-CSF:

Granulocyte–macrophage colony-stimulating factor

ICSI:

Intracytoplasmic sperm injection

ART:

Assisted reproductive technology

TESE:

Testicular sperm extraction

PTF:

Pentoxifylline

GLUTs:

Glucose transporters

GLUT1:

Glucose transporter 1

PI3K/AKT:

Phosphoinositide-3-kinase/protein kinase B

OAT:

Oligoasthenoteratospermia

GnRH:

Gonadotropin-releasing hormone

hCG:

Human chorionic gonadotrophin

References

  1. Moein MR et al (2015) Evaluation of sperm retrieval rate with bilateral testicular sperm extraction in infertile patients with azoospermia. Iran J Reprod Med 13(11):711

    PubMed  PubMed Central  Google Scholar 

  2. Schill T et al (2003) Clinical and endocrine follow-up of patients after testicular sperm extraction. Fertil Steril 79(2):281–286

    Article  PubMed  Google Scholar 

  3. Nagy Z et al (1995) Andrology: the result of intracytoplasmic sperm injection is not related to any of the three basic sperm parameters. Hum Reprod 10(5):1123–1129

    Article  CAS  PubMed  Google Scholar 

  4. Ortega C et al (2011) Absolute asthenozoospermia and ICSI: what are the options? Hum Reprod Update 17(5):684–692

    Article  CAS  PubMed  Google Scholar 

  5. Stalf T et al (2005) Influence of motility and vitality in intracytoplasmic sperm injection with ejaculated and testicular sperm. Andrologia 37(4):125–130

    Article  CAS  PubMed  Google Scholar 

  6. Angelopoulos T et al (1999) Enhancement or initiation of testicular sperm motility by in vitro culture of testicular tissue. Fertil Steril 71(2):240–243

    Article  CAS  PubMed  Google Scholar 

  7. Kovačič B, Vlaisavljević V, Reljič M (2006) Clinical use of pentoxifylline for activation of immotile testicular sperm before ICSI in patients with azoospermia. Journal of andrology 27(1):45–52

    Article  PubMed  Google Scholar 

  8. Aliabadi E et al (2012) Effects of l-carnitine and l-acetyl-carnitine on testicular sperm motility and chromatin quality. Iran J Reprod Med 10(2):77

    CAS  PubMed  PubMed Central  Google Scholar 

  9. McLay RN, Banks WA, Kastin AJ (1997) Granulocyte macrophage-colony stimulating factor crosses the blood-testis barrier in mice. Biol Reprod 57(4):822–826

    Article  CAS  PubMed  Google Scholar 

  10. Ziebe S et al (2013) A randomized clinical trial to evaluate the effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) in embryo culture medium for in vitro fertilization. Fertil Steril 99(6):1600.e2-1609.e2

    Article  Google Scholar 

  11. Zhao JM et al (2003) Clinical pathology and pathogenesis of severe acute respiratory syndrome. Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi 17(3):217–221

    PubMed  Google Scholar 

  12. Vilanova LT et al (2003) Expression of granulocyte–macrophage colony stimulating factor (GM-CSF) in male germ cells: GM-CSF enhances sperm motility. Theriogenology 60(6):1083–1095

    Article  CAS  PubMed  Google Scholar 

  13. Padilla L et al (2020) Granulocyte-macrophage colony stimulating factor (GM-CSF) is fully expressed in the genital tract, seminal plasma and spermatozoa of male pigs. Sci Rep 10(1):1–12

    Article  Google Scholar 

  14. Rodríguez-Gil J et al (2007) Expression of the GM-CSF receptor in ovine spermatozoa: GM-CSF effect on sperm viability and motility of sperm subpopulations after the freezing–thawing process. Theriogenology 67(8):1359–1370

    Article  PubMed  Google Scholar 

  15. Jorban A, Lunenfeld E, Huleihel M (2023) Granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced maturation of spermatogonial cells from prepubertal mice in vitro is enhanced by testosterone. Eur Cytokine Netw 34(4):54–62

    Article  PubMed  Google Scholar 

  16. Niemi M, Sharpe R, Brown W (1986) Macrophages in the interstitial tissue of the rat testis. Cell Tissue Res 243(2):337–344

    Article  CAS  PubMed  Google Scholar 

  17. Kern S et al (1995) Cytokine secretion by macrophages in the rat testis. Biol Reprod 53(6):1407–1416

    Article  CAS  PubMed  Google Scholar 

  18. Rubenstein M et al (1991) GM-CSF restoration of a differentiated (growth factor-regulated) phenotype in an anaplastic tumor. Urol Res 19(5):309–312

    Article  CAS  PubMed  Google Scholar 

  19. Ota K et al (2013) Expression of a2 vacuolar ATPase in spermatozoa is associated with semen quality and chemokine-cytokine profiles in infertile men. PLoS ONE 8(7):e70470

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hosseini E et al (2024) Granulocyte-macrophage colony-stimulating factor cytokine addition after the freeze-thawing process improves human sperm motility and vitality in asthenoteratozoospermia patients. Biopreserv Biobank 22(1):38–45

    Article  CAS  PubMed  Google Scholar 

  21. Datta S, Brunet A, Greenberg ME (1999) Cellular survival: a play in three Akts. Genes Dev 13:2905–2927

    Article  CAS  PubMed  Google Scholar 

  22. Zambrano A et al (2010) Cytokine stimulation promotes increased glucose uptake via translocation at the plasma membrane of GLUT1 in HEK293 cells. J Cell Biochem 110(6):1471–1480

    Article  CAS  PubMed  Google Scholar 

  23. Sagare-Patil V et al (2013) Progesterone utilizes the PI3K-AKT pathway in human spermatozoa to regulate motility and hyperactivation but not acrosome reaction. Mol Cell Endocrinol 374(1–2):82–91

    Article  CAS  PubMed  Google Scholar 

  24. Dias TR et al (2014) Sperm glucose transport and metabolism in diabetic individuals. Mol Cell Endocrinol 396(1–2):37–45

    Article  CAS  PubMed  Google Scholar 

  25. Navale AM, Paranjape AN (2016) Glucose transporters: physiological and pathological roles. Biophys Rev 8(1):5–9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Alves MG et al (2013) Diabetes, insulin-mediated glucose metabolism and Sertoli/blood-testis barrier function. Tissue Barriers 1(2):e23992

    Article  PubMed  PubMed Central  Google Scholar 

  27. Ahmed AA et al (2020) Gum Arabic improves the reproductive capacity through upregulation of testicular glucose transporters (GLUTs) mRNA expression in Alloxan induced diabetic rat. Bioact Carbohydr Diet Fibre 22:100218

    Article  CAS  Google Scholar 

  28. Kishimoto A et al (2015) Immunohistochemical localization of GLUT3, MCT1, and MCT2 in the testes of mice and rats: the use of different energy sources in spermatogenesis. Biomed Res 36(4):225–234

    Article  CAS  PubMed  Google Scholar 

  29. Tanhaye Kalate Sabz F et al (2022) GM–CSF (granulocyte-macrophage colony-stimulating factor) treatment improves sperm parameters in men with oligoasthenoteratospermia via PI3K/AKT pathway. Andrologia 54(7):14427

    Article  Google Scholar 

  30. Nikmard F et al (2016) A comparative study on the results of agonist and antagonist protocols based on serum AMH levels in patients undergoing intracytoplasmic sperm injection. Int J Reprod BioMed 14(12):769

    CAS  PubMed  PubMed Central  Google Scholar 

  31. The Vienna consensus (2017) report of an expert meeting on the development of ART laboratory performance indicators. Reprod Biomed Online 35(5):494–510

    Article  Google Scholar 

  32. The Istanbul consensus workshop on embryo assessment (2011) proceedings of an expert meeting. Hum Reprod 26(6):1270–1283

    Article  Google Scholar 

  33. Edition F (2010) Examination and processing of human semen. World Health [Internet]

  34. Lacham-Kaplan O, Trounson A (1993) The effects of the sperm motility activators 2-deoxyadenosine and pentoxifylline used for sperm micro-injection on mouse and human embryo development. Hum Reprod 8(6):945–952

    Article  CAS  PubMed  Google Scholar 

  35. Shi J-Z et al (2010) Expressions of sperm-specific genes in carnitine-cultured testis sperm of obstructive azoospermia patients. Natl J Androl 16(6):504–509

    CAS  Google Scholar 

  36. Robertson SA et al (2001) Granulocyte-macrophage colony-stimulating factor promotes glucose transport and blastomere viability in murine preimplantation embryos. Biol Reprod 64(4):1206–1215

    Article  CAS  PubMed  Google Scholar 

  37. Peralta O et al (2016) Tissue localization of GM-CSF receptor in bovine ovarian follicles and its role on glucose uptake by mural granulosa cells. Anim Reprod Sci 170:157–169

    Article  CAS  PubMed  Google Scholar 

  38. Angulo C et al (1998) Hexose transporter expression and function in mammalian spermatozoa: cellular localization and transport of hexoses and vitamin C. J Cell Biochem 71(2):189–203

    Article  CAS  PubMed  Google Scholar 

  39. Burant C, Davidson N (1994) GLUT3 glucose transporter isoform in rat testis: localization, effect of diabetes mellitus, and comparison to human testis. Am J Physiol-Regul Integr Comp Physiol 267(6):R1488–R1495

    Article  CAS  Google Scholar 

  40. Costa J et al (2023) Mitochondria quality control and male fertility. Biology. https://doi.org/10.3390/biology12060827

    Article  PubMed  PubMed Central  Google Scholar 

  41. Barbagallo F et al (2020) Evaluation of sperm mitochondrial function: a key organelle for sperm motility. J Clin Med. https://doi.org/10.3390/jcm9020363

    Article  PubMed  PubMed Central  Google Scholar 

  42. Wessendarp M et al (2022) Role of GM-CSF in regulating metabolism and mitochondrial functions critical to macrophage proliferation. Mitochondrion 62:85–101

    Article  CAS  PubMed  Google Scholar 

  43. Wang Y-Y et al (2021) Protein kinases regulate hyperactivated motility of human sperm. Chin Med J 134(20):2412–2414

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang J et al (2017) Leucine mediates autophagosome-lysosome fusion and improves sperm motility by activating the PI3K/Akt pathway. Oncotarget 8(67):111807

    Article  PubMed  PubMed Central  Google Scholar 

  45. Pujianto DA, Curry BJ, Aitken RJ (2010) Prolactin exerts a prosurvival effect on human spermatozoa via mechanisms that involve the stimulation of Akt phosphorylation and suppression of caspase activation and capacitation. Endocrinology 151(3):1269–1279

    Article  CAS  PubMed  Google Scholar 

  46. Koppers AJ et al (2011) Phosphoinositide 3-kinase signalling pathway involvement in a truncated apoptotic cascade associated with motility loss and oxidative DNA damage in human spermatozoa. Biochem J 436(3):687–698

    Article  CAS  PubMed  Google Scholar 

  47. Aitken RJ et al (2021) Evidence that extrapancreatic insulin production is involved in the mediation of sperm survival. Mol Cell Endocrinol 526:111193

    Article  CAS  PubMed  Google Scholar 

  48. Peña FJ et al (2022) An integrated overview on the regulation of sperm metabolism (glycolysis-Krebs cycle-oxidative phosphorylation). Anim Reprod Sci 246:106805

    Article  PubMed  Google Scholar 

  49. Imoedemhe DA et al (1992) The effect of caffeine on the ability of spermatozoa to fertilize mature human oocytes. J Assist Reprod Genet 9(2):155–160

    Article  CAS  PubMed  Google Scholar 

  50. Takeda N et al (2009) Clinical outcome of sorting and recovery of ultrasmall amounts of live sperm stimulated with pentoxifylline followed by intracytoplasmic sperm injection. J Mamm Ova Res 26(2):79–85

    Article  Google Scholar 

Download references

Acknowledgements

 The authors would like to thank the Clinical Research Development Unit of Ayatollah Mousavi Hospital, Zanjan University of Medical Sciences, Zanjan, Iran for their cooperation and assistance throughout the period of study.

Funding

This study was financially supported by a grant from the Zanjan University of Medical Sciences (Grant number: A-11-1314-1).

Author information

Authors and Affiliations

Authors

Contributions

FTKS: Original draft preparation. EH., FSA., ZZ., and MM: Conceptualization and Methodology. EH., and FSA: Supervision. MA., and FM: patient recruitment. FTKS., and RK: Performing laboratory work, collecting the data and analysis. All the authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Elham Hosseini or Fatemeh Sadat Amjadi.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Ethics approval and consent to participate

The experimental protocol was approved by the ethical committee of Zanjan University of Medical Sciences, Zanjan, Iran, (IR.ZUMS.REC.1398.357). All participants signed informed consent to participate.

Consent for publication

Not applicable.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tanhaye Kalate Sabz, F., Hosseini, E., Amjadi, F.S. et al. In vitro effect of granulocyte–macrophage colony-stimulating factor (GM-CSF) on the expression of genes related to sperm motility and energy metabolism and intracytoplasmic sperm injection outcomes in obstructive azoospermic patients. Mol Biol Rep 51, 727 (2024). https://doi.org/10.1007/s11033-024-09676-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11033-024-09676-2

Keywords

Navigation