Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-ttngx Total loading time: 0 Render date: 2024-05-12T17:06:04.528Z Has data issue: false hasContentIssue false

4 - Sperm RNA and Its Use as a Clinical Marker

Published online by Cambridge University Press:  25 May 2017

Christopher J. De Jonge
Affiliation:
University of Minnesota
Christopher L. R. Barratt
Affiliation:
University of Dundee
Ryuzo Yanagimachi
Affiliation:
University of Hawaii, Manoa
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
The Sperm Cell
Production, Maturation, Fertilization, Regeneration
, pp. 59 - 72
Publisher: Cambridge University Press
Print publication year: 2017

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Inhorn, MC, Patrizio, P. Infertility around the globe: New thinking on gender, reproductive technologies and global movements in the 21st century. Hum Reprod Update 2015; 21: 411–26.Google Scholar
Petraglia, F, Serour, GI, Chapron, C. The changing prevalence of infertility. Int J Gynaecol Obstet 2013; 123 Suppl 2: S48.Google Scholar
Roy, A, Lin, YN, Matzuk, MM. Genetics of idiopathic male infertility: The power of a cross-species approach. In: Carrell, D (Ed.), The Genetics of Male Infertility. Humana, 2007.Google Scholar
WHO. WHO laboratory manual for examination and processing of human semen (fifth ed.). World Health Organitzation, 2010.Google Scholar
Lefievre, L et al. Counting sperm does not add up any more: Time for a new equation? Reproduction 2007; 133: 675–84.Google Scholar
Lewis, SE. Is sperm evaluation useful in predicting human fertility? Reproduction 2007; 134: 3140.Google Scholar
Anton, E, Krawet, SA. Spermatozoa as biomarkers for the assessment of human male infertility and genotoxicity. Syst Biol Reprod Med 2012; 58: 4150.Google Scholar
Johnson, GD et al. The sperm nucleus: Chromatin, RNA, and the nuclear matrix. Reproduction 2011; 141: 2136.Google Scholar
Martins, RP, Krawetz, SA. Nuclear organization of the protamine locus. Soc Reprod Fertil Suppl 2007; 64: 112.Google Scholar
Oliva, R. Protamines and male infertility. Hum Reprod Update, 2006; 12: 417–35.Google Scholar
Ostermeier, GC et al. Reproductive biology: Delivering spermatozoan RNA to the oocyte. Nature 2004; 429: 154.Google Scholar
Hamatani, T. Human spermatozoal RNAs. Fertil Steril 2012; 97: 275–81.Google Scholar
Boerke, A, Dieleman, SJ, Gadell, BM. A possible role for sperm RNA in early embryo development. Theriogenology 2007; 68 Suppl 1: S147–55.Google Scholar
Jodar, M et al. The presence, role and clinical use of spermatozoal RNAs. Hum Reprod Update 2013; 19: 604–24.Google Scholar
Sendler, E et al. Stability, delivery and functions of human sperm RNAs at fertilization. Nucl Acids Res 2013; 41: 4,104–17.Google Scholar
Goodrich, RJ, Anton, E, Krawetz, SA. Isolating mRNA and small noncoding RNAs from human sperm. Methods Mol Biol 2013; 927: 385–96.Google Scholar
Sendler, E et al. Stability, delivery and functions of human sperm RNAs at fertilization. Nucl Acids Res 2013; 41: 4104–17.Google Scholar
Johnson, GD et al. Cleavage of rRNA ensures translational cessation in sperm at fertilization. Mol Hum Reprod 2011; 17: 721–6.Google Scholar
Jodar, M et al. Absence of sperm RNA elements correlates with idiopathic male infertility. Sci Transl Med 2015; 7: 295re6.Google Scholar
He, Z et al. Small RNA molecules in the regulation of spermatogenesis. Reproduction 2009; 137: 901–11.Google Scholar
Matzuk, MM, Lamb, DJ. The biology of infertility: Research advances and clinical challenges. Nat Med 2008; 14: 1,197213.Google Scholar
Ostermeier, GC et al. A suite of novel human spermatozoal RNAs. J Androl 2005; 26: 70–4.Google Scholar
Griffiths-Jones, S. The microRNA Registry. Nucl Acids Res 2004 32: D109–11.Google Scholar
Friedman, RC et al. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 2009; 19: 92105.Google Scholar
Zamore, PD, Haley, B. Ribo-gnome: The big world of small RNAs. Science 2005; 309: 1,519–24.Google Scholar
McIver, SC et al. miRNA and mammalian male germ cells. Hum Reprod Update 2012; 1: 4459.Google Scholar
Khazaie, Y, Nasr Esfahani, MH. MicroRNA and male infertility: A potential for diagnosis. Int J Fertil Steril 2014; 8: 113–8.Google Scholar
Ha, M, Kim, VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 2014; 15: 509–24.Google Scholar
Saini, HK Griffiths-Jones, S, Enright, AJ. Genomic analysis of human microRNA transcripts. Proc Natl Acad Sci USA 2007; 104: 17,719–24.Google Scholar
Fang, Z et al. The sequence structures of human microRNA molecules and their implications. PLoS One 2013; 8: e54215.Google Scholar
Goh, WS et al. piRNA-directed cleavage of meiotic transcripts regulates spermatogenesis. Genes Dev 2015; 29: 1,032–44.Google Scholar
Sai Lakshmi, S Agrawal, S. piRNABank: A Web resource on classified and clustered Piwi-interacting RNAs. Nucl Acids Res 2008; 36: D173–7.Google Scholar
Siomi, MC et al. PIWI-interacting small RNAs: The vanguard of genome defence. Nat Rev Mol Cell Biol 2011; 12: 246–58.Google Scholar
Lin, H. piRNAs in the germ line. Science 2007; 316: 397.Google Scholar
Reuter, M et al. Miwi catalysis is required for piRNA amplification-independent LINE1 transposon silencing. Nature 2011; 480: 264–7.CrossRefGoogle ScholarPubMed
Peng, JC, Lin, H. Beyond transposons: The epigenetic and somatic functions of the Piwi-piRNA mechanism. Curr Opin Cell Biol 2013; 25: 190–4.Google Scholar
Zhang, P et al. MIWI and piRNA-mediated cleavage of messenger RNAs in mouse testes. Cell Res 2015; 25: 193207.Google Scholar
Zhao, S et al. piRNA-triggered MIWI ubiquitination and removal by APC/C in late spermatogenesis. Dev Cell 2013; 24: 1325.Google Scholar
Sytnikova, Y, Lau, NC. Does the APC/C mark MIWI and piRNAs for a final farewell? Dev Cell 2013; 24: 119–20.Google Scholar
Aravin, A et al. A novel class of small RNAs bind to MILI protein in mouse testes. Nature 2006; 442: 203–7.Google Scholar
Deng, W, Lin, H. miwi, a murine homolog of piwi, encodes a cytoplasmic protein essential for spermatogenesis. Dev Cell 2002; 2: 819–30.Google Scholar
Girard, A et al. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 2006; 442: 199202.Google Scholar
Krawetz, SA et al. A survey of small RNAs in human sperm. Hum Reprod 2011 26: 3,401–12.Google Scholar
Pantano, L et al. The small RNA content of human sperm reveals pseudogene-derived piRNAs complementary to protein-coding genes. RNA 2015; 21: 1085–95.Google Scholar
Gou, LT et al. Pachytene piRNAs instruct massive mRNA elimination during late spermiogenesis. Cell Res 2014; 24: 680700.Google Scholar
Gou, LT et al. Pachytene piRNAs instruct massive mRNA elimination during late spermiogenesis. Cell Res 2015; 25: 266.Google Scholar
Bouhallier, F et al. Role of miR-34c microRNA in the late steps of spermatogenesis. RNA 2010; 16: 720–31.Google Scholar
Liu, WM et al. Sperm-borne microRNA-34c is required for the first cleavage division in mouse. Proc Natl Acad Sci USA 2012; 109: 490–4.Google Scholar
Yuan, S et al. mir-34b/c and mir-449a/b/c are required for spermatogenesis, but not for the first cleavage division in mice. Biol Open 2015; 4: 212–23.Google Scholar
Lalancette, C et al. Identification of human sperm transcripts as candidate markers of male fertility. J Mol Med 2009; 87: 735–48.Google Scholar
Esteves, SC, Miyaoka, R, Agarwal, A. An update on the clinical assessment of the infertile male [corrected]. Clinics (São Paulo) 2011; 66: 691700.Google Scholar
Bansal, SK et al. Differential genes expression between fertile and infertile spermatozoa revealed by transcriptome analysis. PLoS One 2015; 10: e0127007.Google Scholar
Jodar, M et al. Differential RNAs in the sperm cells of asthenozoospermic patients. Hum Reprod 2012; 27: 1,431–8.Google Scholar
Montjean, D et al. Sperm transcriptome profiling in oligozoospermia. J Assist Reprod Genet, 2012. 29(1): 310.Google Scholar
Platts, AE et al. Disease progression and solid tumor survival: A transcriptome decoherence model. Mol Cell Probes 2010; 24: 5360.Google Scholar
McLachlan, RI. Approach to the patient with oligozoospermia. J Clin Endocrinol Metab 2013; 98: 873–80.Google Scholar
Guo, X et al. Differential expression of VASA gene in ejaculated spermatozoa from normozoospermic men and patients with oligozoospermia. Asian J Androl 2007; 9: 339–44.Google Scholar
Yatsenko, AN et al. UBE2B mRNA alterations are associated with severe oligozoospermia in infertile men. Mol Hum Reprod 2013; 19: 388394.Google Scholar
Ferlin, A et al. Heat shock protein and heat shock factor expression in sperm: Relation to oligozoospermia and varicocele. J Urol 2010; 183: 1,248–52.Google Scholar
Geijsen, N et al. Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 2004; 427: 148–54.Google Scholar
Baarends, WM, Grootegoed, JA. Chromatin dynamics in the male meiotic prophase. Cytogenet Genome Res 2003; 103: 225–34.Google Scholar
de Jong, AM et al. Effect of alcohol intake and cigarette smoking on sperm parameters and pregnancy. Andrologia 2014; 46: 112–7.Google Scholar
Eslamian, G et al. Intake of food groups and idiopathic asthenozoospermia: A case-control study. Hum Reprod 2012; 27: 3,328–36.Google Scholar
Marchini, M et al. Etiology of severe asthenozoospermia and fertility prognosis. A screening of 5216 semen analyses. Andrologia 1991; 23: 115–20.Google Scholar
Kempisty, B et al. Evaluation of protamines 1 and 2 transcript contents in spermatozoa from asthenozoospermic men. Folia Histochem Cytobiol 2007; 45: S109–13.Google Scholar
Jedrzejczak, P et al. Quantitative assessment of transition proteins 1, 2 spermatid-specific linker histone H1-like protein transcripts in spermatozoa from normozoospermic and asthenozoospermic men. Arch Androl 2007; 53: 199205.Google Scholar
Miyagawa, Y et al. Single-nucleotide polymorphisms and mutation analyses of the TNP1 and TNP2 genes of fertile and infertile human male populations. J Androl 2005; 26: 779–86.Google Scholar
Liu, B et al. Analysis and difference of voltage-dependent anion channel mRNA in ejaculated spermatozoa from normozoospermic fertile donors and infertile patients with idiopathic asthenozoospermia. J Assist Reprod Genet 2010; 27: 719–24.Google Scholar
Shen, S et al. Low-expressed testis-specific calcium-binding protein CBP86-IV (CABYR) is observed in idiopathic asthenozoospermia. World J Urol 2015; 33: p633–8.Google Scholar
Chen, K et al. Low NRF2 mRNA expression in spermatozoa from men with low sperm motility. Tohoku J Exp Med 2012; 228: 259–66.Google Scholar
Imamovic Kumalic, S, Pinter, B. Review of clinical trials on effects of oral antioxidants on basic semen and other parameters in idiopathic oligoasthenoteratozoospermia. Biomed Res Int 2014; 2014: 426951.Google Scholar
Kruger, TF et al. A quick, reliable staining technique for human sperm morphology. Arch Androl 1987; 18: 275–7.Google Scholar
Gaur, DS, Talekar, MS, Pathak, VP. Alcohol intake and cigarette smoking: Impact of two major lifestyle factors on male fertility. Indian J Pathol Microbiol 2010; 53: 3540.Google Scholar
Ragni, G et al. Evaluation of semen and pituitary gonadotropin function in men with untreated Hodgkin's disease. Fertil Steril 1985; 43: 927–30.Google Scholar
Volta, U, Villanacci, V. Celiac disease: Diagnostic criteria in progress. Cell Mol Immunol 2011; 8: 96102.Google Scholar
Platts, AE et al. Success and failure in human spermatogenesis as revealed by teratozoospermic RNAs. Hum Mol Genet 2007; 16: 763–73.Google Scholar
Kong, M, Diaz, ES, Morales, P. Participation of the human sperm proteasome in the capacitation process and its regulation by protein kinase A and tyrosine kinase. Biol Reprod 2009; 80: 1,026–35.Google Scholar
Cedenho, AP et al. Oligozoospermia and heat-shock protein expression in ejaculated spermatozoa. Hum Reprod 2006; 21: 1,791–4.Google Scholar
Li, C et al. Absence of nerve growth factor and comparison of tyrosine kinase receptor A levels in mature spermatozoa from oligoasthenozoospermic, asthenozoospermic and fertile men. Clin Chim Acta 2010; 411: 1,482–6.Google Scholar
Zheng, L et al. Expression of brain-derived neurotrophic factor in mature spermatozoa from fertile and infertile men. Clin Chim Acta 2011; 412: p44–7.Google Scholar
Abu-Halima, M et al. Altered microRNA expression profiles of human spermatozoa in patients with different spermatogenic impairments. Fertil Steril 2013; 99: 1,249–55 e16.Google Scholar
Abu-Halima, M et al. A panel of five microRNAs as potential biomarkers for the diagnosis and assessment of male infertility. Fertil Steril 2014; 102: 989–97 e1.Google Scholar
Salas-Huetos, A et al. New insights into the expression profile and function of micro-ribonucleic acid in human spermatozoa. Fertil Steril 2014; 102: 213–22 e4.Google Scholar
Salas-Huetos, A et al. Spermatozoa from patients with seminal alterations exhibit a differential miRNA profile. Fertil Steril 2015; doi: 10.1016/j.fertnstert.2015.06.015.Google Scholar
Duran, HE et al. Intrauterine insemination: A systematic review on determinants of success. Hum Reprod Update 2002; 8: 373–84.Google Scholar
Practice Committees of the American Society for Reproductive Medicine and Society for Assisted Reproductive Technology. Intracytoplasmic sperm injection (ICSI) for non-male factor infertility: A committee opinion. Fertil Steril 2012; 98: 1,395–9.Google Scholar
Pashayan, N, Lyratzopoulos, G, Mathur, R. Cost-effectiveness of primary offer of IVF vs. primary offer of IUI followed by IVF (for IUI failures) in couples with unexplained or mild male factor subfertility. BMC Health Serv Res 2006; 6: 80.Google Scholar
Piskol, R, Ramaswami, G, Li, JB. Reliable identification of genomic variants from RNA-seq data. Am J Hum Genet 2013; 93: 641–51.Google Scholar
Selvaraju, S, Jodar, M, Krawetz, S. The influence of environmental contaminants and lifestyle on testicular damage and male fertility. Humana Press, 2014: 119.Google Scholar
Carone, BR et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 2010; 143: 1,084–96.Google Scholar
Ng, S et al. Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature 2010; 467: 963U103.Google Scholar
Lombo, M et al. Transgenerational inheritance of heart disorders caused by paternal bisphenol A exposure. Environ Pollut 2015; 206: 667–78.Google Scholar
Dias, BG, Ressler, KJ. Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat Neurosci 2014; 17: 8996.Google Scholar
Cossetti, C et al. Soma-to-germline transmission of RNA in mice xenografted with human tumour cells: Possible transport by exosomes. PLoS One 2014; 9: e101629.Google Scholar
Gapp, K et al. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat Neurosci 2014; 17: 667–9.Google Scholar
Rando, OJ. Daddy issues: Paternal effects on phenotype. Cell 2012; 151: 702–8.Google Scholar
Fullston, T et al. Paternal obesity initiates metabolic disturbances in two generations of mice with incomplete penetrance to the F2 generation and alters the transcriptional profile of testis and sperm microRNA content. FASEB J 2013; 27: 4,226–43.Google Scholar
McPherson, NO et al. Preconception diet or exercise intervention in obese fathers normalizes sperm microRNA profile and metabolic syndrome in female offspring. Am J Physiol Endocrinol Metab 2015; 308: E805–21.Google Scholar
Vojtech, L et al. Exosomes in human semen carry a distinctive repertoire of small non-coding RNAs with potential regulatory functions. Nucleic Acids Res 2014; 42: 7,290304.Google Scholar
Bromfield, JJ. Seminal fluid and reproduction: Much more than previously thought. J Assist Reprod Genet 2014; 31: 627–36.Google Scholar
Jodar, M, Sendler, E, Krawetz, SA. The protein and transcript profiles of human semen. Cell Tissue Res 2015; 363( ): 8596.Google Scholar
Pansa, A et al. ESX1 mRNA expression in seminal fluid is an indicator of residual spermatogenesis in non-obstructive azoospermic men. Hum Reprod 2014; 29: 2,620–7.Google Scholar
Practice Committee of the American Society for Reproductive Medicine. Effectiveness and treatment for unexplained infertility. Fertil Steril 2006; 86: S111–4.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×