Potential Pathogenic Bacteria in Seminal Microbiota of Patients with Different Types of Dysspermatism

Huijun Yang, Jiaming Zhang, Zhiwei Xue, Changying Zhao, Lijun Lei, Yan Wen, Yunling Dong, Junjie Yang, Lei Zhang, Huijun Yang, Jiaming Zhang, Zhiwei Xue, Changying Zhao, Lijun Lei, Yan Wen, Yunling Dong, Junjie Yang, Lei Zhang

Abstract

Human microbiota play an important role in the health of their human hosts. Recent studies have demonstrated that microbiota exist in seminal plasma. The current study aims to elucidate whether seminal microbiota exist in patients with different types of dysspermatism and whether bacterial biomarkers can be identified for them. A total of 159 study participants were recruited, including 22 patients with oligoasthenospermia, 58 patients with asthenospermia, 8 patients with azoospermia, 13 patients with oligospermia, and 58 matched healthy controls. Seminal microbiota composition was analyzed using 16S rRNA gene-based sequencing. The results showed that the composition of seminal microbiota of patients with dysspermatism differed from those of healthy controls. Comparison of the microbiota composition in semen samples from patients with different types of dysspermatism showed that microbiota in patients with asthenospermia and oligoasthenospermia were distinct from healthy controls in beta diversity (P < 0.05). Characteristic biomarkers, including Ureaplasma, Bacteroides, Anaerococcus, Finegoldia, Lactobacillus and Acinetobacter lwoffii, were identified based on LEfSe analysis. Inferred functional analysis based on seminal microbiome data further indicated the presence of potential pathogenic biomarkers in patients with asthenospermia and oligoasthenospermia. These results provided profiles of seminal microbiota exhibited in different types of dysspermatism, thus providing new insights into their pathogenesis.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Relative abundance of taxa among five groups. Comparison of OTUs and relative taxa abundance among asthenospermia, oligospermia, oligoasthenospermia, azoospermia, and healthy controls. (A) At phylum level; (B) at genus level.
Figure 2
Figure 2
Comparison of alpha diversity and relative abundance at phylum level based on the OTUs profile. Box plots depict differences in microbiome diversity based on (A) Shannon index and (B) ACE index between patients with dysspermatism and healthy controls. (C,D) show Shannon index and ACE index among patients with asthenospermia, oligospermia, oligoasthenospermia, and azoospermia, and healthy controls. The p value was calculated using the Wilcoxon rank-sum test.
Figure 3
Figure 3
PCoA analysis of microbiota between patients with dysspermatism and healthy controls. (A) Unweighted unifrac PCoA; (B) Weighted unifrac PCoA. PCoA analysis of the microbiota among patients with asthenospermia, oligospermia, oligoasthenospermia, and azoospermia, and healthy controls; (C) Unweighted unifrac PCoA; (D) Weighted unifrac PCoA.
Figure 4
Figure 4
The most differentially abundant taxa between patients with dysspermatism and healthy controls (LDA score above 3) generated using LEfSe analysis.
Figure 5
Figure 5
LEfSe analysis between patients asthenospermia and healthy controls. (A,B) Comparison of the most differentially abundant taxa between patients with asthenospermia and healthy controls (LDA score above 3) generated using LEfSe analysis. We selected 4 biomarkers to predict the probability of patients with asthenospermia. (CF) These biomarkers are Propionibacterium, Pelomonas, Lactobacillus, and Propionibacterium acnes. The ROC curves as well as the AUC (Area Under the Curve) values were calculated using SPSS.
Figure 6
Figure 6
LEfSe analysis between patients with oligoasthenospermia and healthy controls. (A,B) Comparison of the most differentially abundant taxa between patients with oligoasthenospermia and healthy controls (LDA score above 3), were generated using LEfSe analysis. (CH) These biomarkers are Lactobacillus, Acinetobacter, Propionibacterium, Pelomonas, Prevotella copri, and Propionibacterium acnes. The ROC curves as well as the AUC (Area Under the Curve) value was calculated using SPSS.
Figure 7
Figure 7
Predicted metagenome function based on KEGG pathway analysis. Extended error bar plot showed significantly different KEGG pathways between (A) patients with asthenospermia and healthy controls; (B) between patients with oligoasthenospermia and healthy controls.

References

    1. Spadoni I, et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science. 2015;350:830–834. doi: 10.1126/science.aad0135.
    1. Cavarretta I, et al. The Microbiome of the Prostate Tumor Microenvironment. Eur. Urol. 2017;72:625–631. doi: 10.1016/j.eururo.2017.03.029.
    1. Liu S, et al. Effect of Acrylamide on Oocyte Nuclear Maturation and Cumulus Cells Apoptosis in Mouse In Vitro. Plos One. 2015;10:e0135818. doi: 10.1371/journal.pone.0135818.
    1. Bhattacharyya M, Ghosh T, Shankar S, Tomar N. The conserved phylogeny of blood microbiome. Mol. Phylogenet Evol. 2017;109:404–408. doi: 10.1016/j.ympev.2017.02.001.
    1. Altmae S, Franasiak JM, Mandar R. The seminal microbiome in health and disease. Nat. Rev. Urol. 2019;16:703–721. doi: 10.1038/s41585-019-0250-y.
    1. Merino G, et al. Bacterial infection and semen characteristics in infertile men. Arch. Androl. 1995;35:43–47. doi: 10.3109/01485019508987852.
    1. La Vignera S, Vicari E, Condorelli RA, D’Agata R, Calogero AE. Male accessory gland infection and sperm parameters (review) Int. J. Androl. 2011;34:e330–347. doi: 10.1111/j.1365-2605.2011.01200.x.
    1. Ahmadi MH, Mirsalehian A, Gilani MAS, Bahador A, Talebi M. Improvement of semen parameters after antibiotic therapy in asymptomatic infertile men infected with Mycoplasma genitalium. Infection. 2018;46:31–38. doi: 10.1007/s15010-017-1075-3.
    1. Kaur K, Prabha V. Spermagglutinating Escherichia coli and its role in infertility: in vivo study. Microb. Pathog. 2014;69-70:33–38. doi: 10.1016/j.micpath.2014.03.010.
    1. Esmailkhani A, et al. Assessing the prevalence of Staphylococcus aureus in infertile male patients in Tabriz, northwest Iran. Int. J. Reprod. Biomed. 2018;16:469–474.
    1. Berjis K, Ghiasi M, Sangy S. Study of seminal infection among an infertile male population in Qom, Iran, and its effect on sperm quality. Iran. J. Microbiol. 2018;10:111–116.
    1. Ahmadi MH, Mirsalehian A, Sadighi Gilani MA, Bahador A, Talebi M. Asymptomatic Infection With Mycoplasma hominis Negatively Affects Semen Parameters and Leads to Male Infertility as Confirmed by Improved Semen Parameters After Antibiotic Treatment. Urology. 2017;100:97–102. doi: 10.1016/j.urology.2016.11.018.
    1. Hou D, et al. Microbiota of the seminal fluid from healthy and infertile men. Fertil. Steril. 2013;100:1261–1269. doi: 10.1016/j.fertnstert.2013.07.1991.
    1. Weng SL, et al. Bacterial communities in semen from men of infertile couples: metagenomic sequencing reveals relationships of seminal microbiota to semen quality. Plos One. 2014;9:e110152. doi: 10.1371/journal.pone.0110152.
    1. Alfano M, et al. Testicular microbiome in azoospermic men-first evidence of the impact of an altered microenvironment. Hum. Reprod. 2018;33:1212–1217. doi: 10.1093/humrep/dey116.
    1. Javurek AB, et al. Discovery of a Novel Seminal Fluid Microbiome and Influence of Estrogen Receptor Alpha Genetic Status. Sci. Rep. 2016;6:23027. doi: 10.1038/srep23027.
    1. Javurek AB, et al. Consumption of a high-fat diet alters the seminal fluid and gut microbiomes in male mice. Reprod. Fertil. Dev. 2017;29:1602–1612. doi: 10.1071/RD16119.
    1. Mandar R, et al. Complementary seminovaginal microbiome in couples. Res. Microbiol. 2015;166:440–447. doi: 10.1016/j.resmic.2015.03.009.
    1. Baud D, et al. Sperm Microbiota and Its Impact on Semen Parameters. Front. Microbiol. 2019;10:234. doi: 10.3389/fmicb.2019.00234.
    1. Alahmar AT. The effects of oral antioxidants on the semen of men with idiopathic oligoasthenoteratozoospermia. Clin. Exp. Reprod. Med. 2018;45:57–66. doi: 10.5653/cerm.2018.45.2.57.
    1. Bonnici J, Fenech A, Muscat C, Calleja-Agius J. The role of seminal fluid in infertility. Minerva Ginecol. 2017;69:390–401. doi: 10.23736/S0026-4784.17.04049-7.
    1. Akinsal EC, Baydilli N, Dundar M, Ekmekcioglu O. The frequencies of Y chromosome microdeletions in infertile males. Turk. J. Urol. 2018;44:389–392. doi: 10.5152/tud.2018.73669.
    1. Li, W. et al. Biallelic mutations of CFAP251 cause sperm flagellar defects and human male infertility. J Hum Genet, 10.1038/s10038-018-0520-1 (2018).
    1. Adelman CA, Petrini JH. ZIP4H (TEX11) deficiency in the mouse impairs meiotic double strand break repair and the regulation of crossing over. Plos Genet. 2008;4:e1000042. doi: 10.1371/journal.pgen.1000042.
    1. Dohle GR, et al. EAU guidelines on male infertility. Eur. Urol. 2005;48:703–711. doi: 10.1016/j.eururo.2005.06.002.
    1. Pant N, et al. Lead and cadmium concentration in the seminal plasma of men in the general population: correlation with sperm quality. Reprod. Toxicol. 2003;17:447–450. doi: 10.1016/S0890-6238(03)00036-4.
    1. Emokpae MA, Chima HN. Effect of senescence on some apoptosis and oxidative stress markers in infertile normozospermic and oligospermic men: A cross-sectional study. Int. J. Reprod. Biomed. 2018;16:435–442. doi: 10.29252/ijrm.16.7.435.
    1. Du Plessis SS, Agarwal A, Halabi J, Tvrda E. Contemporary evidence on the physiological role of reactive oxygen species in human sperm function. J. Assist. Reprod. Genet. 2015;32:509–520. doi: 10.1007/s10815-014-0425-7.
    1. Zufferey F, et al. Steroid profiles in both blood serum and seminal plasma are not correlated and do not reflect sperm quality: Study on the male reproductive health of fifty young Swiss men. Clin. Biochem. 2018;62:39–46. doi: 10.1016/j.clinbiochem.2018.03.008.
    1. Ratnayake GM, Weerathunga PN, Ruwanpura LP, Wickramasinghe A, Katulanda P. Isolated follicle stimulated hormone deficiency in male: case report. BMC Res. Notes. 2018;11:24. doi: 10.1186/s13104-017-3109-4.
    1. Di Nisio, A. et al. Endocrine disruption of androgenic activity by perfluoroalkyl substances: clinical and experimental evidence. J Clin Endocrinol Metab, 10.1210/jc.2018-01855 (2018).
    1. Sukhn, C., Awwad, J., Ghantous, A. & Zaatari, G. Associations of semen quality with non-essential heavy metals in blood and seminal fluid: data from the Environment and Male Infertility (EMI) study in Lebanon. J Assist Reprod Genet, 10.1007/s10815-018-1236-z (2018).
    1. McDiarmid, M. A., Gucer, P., Centeno, J. A., Todorov, T. & Squibb, K. S. Semen Uranium Concentrations in Depleted Uranium Exposed Gulf War Veterans: Correlations with Other Body Fluid Matrices. Biol Trace Elem Res, 10.1007/s12011-018-1527-3 (2018).
    1. Camargo M, Intasqui P, Bertolla RP. Understanding the seminal plasma proteome and its role in male fertility. Basic. Clin. Androl. 2018;28:6. doi: 10.1186/s12610-018-0071-5.
    1. Monteiro C, et al. Characterization of microbiota in male infertility cases uncovers differences in seminal hyperviscosity and oligoasthenoteratozoospermia possibly correlated with increased prevalence of infectious bacteria. Am. J. Reprod. Immunol. 2018;79:e12838. doi: 10.1111/aji.12838.
    1. Qin J, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60. doi: 10.1038/nature11450.
    1. Younes JA, et al. Women and Their Microbes: The Unexpected Friendship. Trends Microbiol. 2018;26:16–32. doi: 10.1016/j.tim.2017.07.008.
    1. Lewkowicz P, Lewkowicz N, Sasiak A, Tchorzewski H. Lipopolysaccharide-activated CD4+CD25+ T regulatory cells inhibit neutrophil function and promote their apoptosis and death. J. Immunol. 2006;177:7155–7163. doi: 10.4049/jimmunol.177.10.7155.
    1. Fraczek M, Szumala-Kakol A, Dworacki G, Sanocka D, Kurpisz M. In vitro reconstruction of inflammatory reaction in human semen: effect on sperm DNA fragmentation. J. Reprod. Immunol. 2013;100:76–85. doi: 10.1016/j.jri.2013.09.005.
    1. Fraczek M, Szumala-Kakol A, Jedrzejczak P, Kamieniczna M, Kurpisz M. Bacteria trigger oxygen radical release and sperm lipid peroxidation in in vitro model of semen inflammation. Fertil. Steril. 2007;88:1076–1085. doi: 10.1016/j.fertnstert.2006.12.025.
    1. Fraczek M, et al. Fertilizing potential of ejaculated human spermatozoa during in vitro semen bacterial infection. Fertil. Steril. 2014;102:711–719 e711. doi: 10.1016/j.fertnstert.2014.06.002.
    1. Organization, W. H. WHO laboratory manual for the examination and processing of human semen. (2010).
    1. Liu, W. et al. Correlation of genetic results with testicular histology, hormones and sperm retrieval in nonobstructive azoospermia patients with testis biopsy. Andrologia49, 10.1111/and.12705 (2017).
    1. Jurewicz J, et al. Human Semen Quality, Sperm DNA Damage, and the Level of Reproductive Hormones in Relation to Urinary Concentrations of Parabens. J. Occup. Env. Med. 2017;59:1034–1040. doi: 10.1097/JOM.0000000000001106.
    1. Garolla A, et al. FSH treatment in infertile males candidate to assisted reproduction improved sperm DNA fragmentation and pregnancy rate. Endocrine. 2017;56:416–425. doi: 10.1007/s12020-016-1037-z.
    1. Wang X, et al. Gut microbiota dysbiosis is associated with Henoch-Schonlein Purpura in children. Int. immunopharmacology. 2018;58:1–8. doi: 10.1016/j.intimp.2018.03.003.
    1. Magoc T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics. 2011;27:2957–2963. doi: 10.1093/bioinformatics/btr507.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460. doi: 10.1093/bioinformatics/btq461.
    1. Cole JR, et al. The Ribosomal Database Project: improved alignments and new tools for rRNA analysis. Nucleic acids Res. 2009;37:D141–145. doi: 10.1093/nar/gkn879.

Source: PubMed

3
Subskrybuj