Open access peer-reviewed chapter

MicroRNAs and Male Infertility

Written By

Mohsin Munawar, Irfana Liaqat and Shaukat Ali

Submitted: 24 May 2022 Reviewed: 26 July 2022 Published: 14 October 2022

DOI: 10.5772/intechopen.106757

From the Edited Volume

Recent Advances in Noncoding RNAs

Edited by Lütfi Tutar

Chapter metrics overview

181 Chapter Downloads

View Full Metrics

Abstract

Spermatozoan production is tightly controlled by the multistep process of spermatogenesis and spermiogenesis. Physiological and molecular disruption in spermatogenesis can lead to various reproductive disorders including male infertility. Male infertility is associated with various etiologies, but mechanism is not determined yet. MicroRNAs (miRNAs) are almost 22 nucleotides long, non-protein coding RNA that play an essential role in posttranscriptional regulations in various biological processes including spermatogenesis. The current review is aimed to summarize the recent literature on the role of miRNAs in male infertility and spermatogenesis and their potential in diagnosis, prognosis, and therapy of the disease. miRNAs have shown tremendous potential to be used as diagnostic and prognostic marker and therapeutic target in diseases related to male infertility. Experimental evidence reveals that aberrant expression of miRNAs affects different cell types and different stages of spermatogenesis, which ultimately leads to male infertility. To exploit the full potential of miRNAs, characterization of unidentified miRNAs is required to understand the miRNA-mediated regulatory mechanism related to male infertility.

Keywords

  • miRNAs
  • male infertility
  • spermatogenesis
  • spermiogenesis
  • non-coding RNA
  • therapeutic agents

1. Introduction

Infertility is the inability to conceive spontaneously in 1 year by a sexually active, non-contracepting couple. Primary infertility, the complete inability to conceive, ranges from 2 to 5%, while secondary infertility, indicating cessation of further fertility, globally has a prevalence rate of 20%. Approximately 15% (48.5 million) of couples cannot conceive worldwide [1].

Among infertile men, the etiology of 30−40% of cases remains unknown, so considered idiopathic male infertility [2]. The possible causes of male infertility are physiological anomalies, immunological factors, some genetic abnormalities [3], and some environmental toxicants, which can disturb the reproductive health of male by disturbing steroidogenesis, spermatogenesis, and histopathological structures [4]. Among all these possible causes of male infertility, several types of small non-coding RNAs, including microRNAs (miRNAs), are expressed in the male germ line and impair the mammalian spermatogenesis [5, 6].

Protein coding genes represent only 1.5%of the genome, which can be increased by up to 2% if untranslated regions (UTRs) are also included. The remaining 98% genome, the non-protein-coding region, has been considered a “black box” until the characterization of non-coding RNAs (ncRNAs) was done using novel nucleotide sequencing technologies. ncRNAs are present in cellular compartments, participating in multiple biological functions, including the male reproductive system, whereas some are identified in extracellular fluids, correctly named as circulating ncRNAs, where they can be detected in exosomes, bound on lipoprotein and free circulating molecules [7].

Ambros and his colleagues 1993 discovered the miRNA and then revealed the ncRNAs as gene regulators in eukaryotes [8, 9]. Lin-4 and let-7 in Caenorhabditis elegans were first discovered ncRNAs, later classified as miRNAs. Thus far, almost 38579 entries have been recorded, and among these, up to 1982 miRNAs are only from the human genome [10]. miRNAs code is either located in the intergenic region or introns of genes of the whole genome. In contrast, nearly half of total miRNAs are arranged nearby (within 50kb of another miRNA code) to form clusters. These clusters range from 2 to 46 miRNAs; C19MC is the major known cluster of miRNAs located on chromosome 19 in primates [11]. It is predicted that over 30% of mammalian and 60% of human genes are regulated by miRNAs [12, 13].

miRNAs are small RNA molecules, derived from the genome-encoded-precursor loop, consisting of almost 19–23 nucleotides. They are conserved in species and so regulate the expression of the mRNAs by recognizing the targeted mRNA through base-pair binding to the 3′UTR [14, 15].

Advertisement

2. miRNA nomenclature

MicroRNAs (prefix; “miR" for mature sequence whereas precursor hairpins are labeled as "mir") are named by using the number in sequential order, such as miR-1, miR-2. Despite the discovery of identical miRNAs in different organisms, the same number is assigned. Lettered suffix such as miR-1a and miR-1b is assigned to the sequences whose mature miRNAs sequences differ at one of two positions. Numbered suffix (e.g., miR-1-1 and miR-1-2) is used to label the identical miRNAs that have originated from distinct hairpin loci. The -3p and -5p suffixes refer to the arm from which the mature miRNA sequence originates. Detailed guidelines for naming the miRNAs can be seen in [16, 17].

Advertisement

3. MicroRNAs: biogenesis

miRNAs have a vital role as epigenetic regulators. The enzyme (Dicer protein) used for the biosynthesis of miRNA plays a very crucial role for the developmental processes [18]. During miRNAs biogenesis, following steps will be followed sequentially as elaborated in Figure 1:

  1. Primarily, a long imperfect ~80 nt hairpin-like structure primary microRNA (pri-miRNA) is transcribed by RNA polymerase (RNA pol) II from intergenic non-coding region or intragenic region intron (untranslated region of protein-coding genes).

  2. Secondarily, pri-miRNA undergoes two steps enzymes (Drosha and Dicer from RNase III superfamily) catalyzed reaction.

    1. Pri-miRNA (~80-nt hairpin structure) is excised to form an intermediate precursor (pre-) miRNA (~65–70 nt) processed by Nuclear Drosha-DGCR8(DiGeorge syndrome critical region 8), then transported to cytoplasm through export factor Exportin-5.

    2. Pri-miRNA is processed by called Dicer-TRBP (trans-activating region binding protein), a second RNase III endonuclease enzyme, to remove the terminal loop of pre-miRNA resulting in ~20 bp miRNA: miRNA* duplex.

  3. The Dicer-TRBP processed duplex is loaded onto Ago-2 (Argonaute) protein to form an miRNA induced silencing complex (miRISC). miRISC contains mature miRNA that regulates the gene expression, while another miRNA* will be released and degraded.

  4. Finally activated region (miRNA) of miRISC binds with 3′ UTR of targeted mRNA, ultimately leading to translational repression, deadenylation, or degradation of targeted mRNA [19].

Figure 1.

Mechanism of miRNA’s biosynthesis.

Advertisement

4. miRNAs: regulators of biological processes

miRNAs play a vital role in the regulation of various biological phenomena including cellular differentiation, cell growth, programmed cell death, and embryonic development [20, 21, 22], whereas up- or downregulation may lead to serious abnormality. For instance, cardiovascular disease, cancer, diabetes, renal abnormality, schizophrenia, muscular dystrophy, and reproductive disorder especially related to sperm formation and its function have been linked with biosynthesis impairment, mutation, or improper regulation of miRNA [23, 24, 25, 26, 27, 28, 29]. miRNA is not solely responsible for the whole regulatory mechanism, but their activities are also dependent on other proteins or complexes depending upon the processor stage [30].

Mature human spermatozoa retain various miRNAs; moreover, variable expression of spermatozoal RNAs has been associated with male infertility [31]. miRNAs, present in primordial germ cells, spermatogonia, primary and secondary spermatocytes, spermatids, and mature spermatozoa, play an epigenetic role in the spermatogenesis and early development as well (Figure 2) [32].

Figure 2.

miRNAs expressed during different stages of spermatogenesis in mammals.

Advertisement

5. miRNA as potential biomarker in male infertility

In order to confirm the male infertility, various methods have been adopted, but the most recent way is the use of biomarker. Biomarkers can be used to diagnose the disease before the appearance of physiological symptoms. miRNA is known as the most reliable biomarker due to various known properties.

5.1 Properties of biomarker

A biomarker molecule should be stable, target-specific, and sensitive to detection, noninvasive, and from an easily accessible source. In the field of medical science, protein molecules, which are easily found in serum plasma, have been used as a biomarker for the detection of pathological conditions and their prognosis. However, as a biomarker, protein molecules have some limitations such as reduced diagnostic value due to low sensitivity and specificity of their detection. On the other hand, miRNAs present in the extracellular or intracellular environment appear much earlier and detected at minute concentration, thus allowing early diagnosis. Expressions of miRNAs are often biological-stage-specific [33, 34]. Moreover, miRNA is observed as the homogeneous population of molecules, but protein molecules may become heterogeneous due to different posttranslational modifications.

miRNAs are more stable than mRNA but lesser stable than DNA. Extracellular miRNAs exist in microvesicles, exosomes, and lipoproteins or form a complex with Ago-2 protein, so protected from endogenous RNAse activity, which suggests their reliable use as a noninvasive biomarker for spermatogenic impairment [35, 36, 37, 38]. Some miRNAs are stable at room temperature, sustain a repeated freeze-thaw cycle, and got stability under harsh conditions even found in degraded RNA preparations [39, 40, 41]. In a study, exosomal miRNAs were found to be stable at 4°C for 2 weeks, at −80°C for 2 months, and at −20°C for 5 year [42]. So their relative stability enhances their usage as biomarker molecules. miRNA (both extracellular and intracellular) can be isolated by using noninvasive procedures and detected with techniques in common routines such as RT-qPCR and miRNA microarray, which is more advantageous than conventional and invasive (e.g., prostate and testicular biopsy) diagnostic procedures [43, 44, 45, 46, 47]. Despite all these characteristics of miRNAs as a biomarker, the use of protein as a biomarker may not be completely replaced, but no doubt miRNAs can be labeled as a good candidate for complementary diagnosis tools.

Circulating (serum) miRNAs were firstly used as biomarkers for the detection of diffuse large B-cell lymphoma [48]. In another study, the downregulation of miR-34c, miR-122, and miR-181a was observed in the serum of patients suffering from oligoasthenospermia [49]. Wang and his colleagues reported a decreased level of various miRNAs in azoospermic patients when compared with control [44]. Many other such published literature suggested that miRNAs are suitable biomarkers for diagnostic purposes [50, 51].

In mammals, sperm deliver genome and epigenome to the oocyte, which includes protein factors, methylation of DNA, and non-coding RNAs [52]. During mammalian spermatogenesis, mRNAs undergo posttranscriptional and posttranslational regulation in differentiating germ cells [53], so the regulatory role of miRNAs during the process of spermatogenesis can be observed clearly.

Many miRNAs as families or individual candidates are found to be very crucial in many systems of humans including the reproductive system. The role of some miRNA's family and individual miRNA candidates is summarized.

5.2 miRNA families

Let-7 family is a large family encoded by 12 different loci [54]. A well-known regulatory role of the let-7 family is gamete differentiation and embryonic implantation in humans [55, 56]. Different members of this family were identified in the testis, spermatozoa, and seminal plasma with differential expression [57, 58, 59].

miR-30-5p family (five members miR30a/b/c/d/e-5p), encoded by six loci located on chromosomes 1, 6, and 8 in humans. All miRNAs are observed in male reproductive tissue cells [60]. These miRNAs were expressed differentially in spermatozoa and seminal plasma of asthenozoospermic, oligozoospermic, and normozoospermic infertile men [39].

miR-345p/449-5p family with five miRNAs (miR-34a-5p, miR-34c-5p, miR-449a-5p, miR-449b-5p miR-449c-5p) has shown a very important regulatory role in mitosis [61]. These are highly expressed in male reproductive cells but variably expressed in pathologically infertile men especially nonobstructive azoospermic and obstructive azoospermic patients [62]. Their role was also investigated in the process of fertilization, cleavage division, and pre-implantation. These miRNAs are highly expressed in the sperm, as miR-34c-5p is the most abundant human sperm miRNA [52].

MiR-303-3p/370-3p/372-3p and 520-3p family each member of family miR-302-3p regulates over 450 target genes [63]. Members of this family were seen to be differentially expressed in the testis having histopathological impairments and in spermatozoa of patients with asthenozoospermia [62].

miR-99-5P/100-5P family is one of the most ancient families with three members (miR-99a/b/c) located in distinct chromosomal regions (ch 21, 19, and 11 respectively) in the human genome [64]. The member of this family shows differential expression in the testis of NOA patients [57].

miR-888 family (miR-888/ Mir-890, miR-891a/b, and miR-892a/b) is located on the X chromosome of primates only which has specific expression in epididymal tissues [65, 66]. Computational functional analyses predicted that the targets of the miR-888 family were related to morphogenesis of epididymis and sperm maturation [67].

5.3 Individual miRNA

hsa-miR-10b-5p, located on chromosome no 2(1q31.1), plays a pivotal role in gametogenesis as it is highly expressed in the testes. The expression level of hsa-miR-10B-5P, among others, was upregulated in round spermatids of NOA patients [57].

hsa-miR-27b-3p, present on chromosome 9 (9q22.32), was differentially expressed in the mature spermatozoa of infertile men. The expression of miR-27 has correlated with the expression of the CRISP2 (cysteine-rich secretory protein 2) gene, the highly expressed gene in the testis [59]. CGN1-CRISP2 complex has an important role in the development of the sperm tail (sperm motility) and predicts the regulatory role of miR27b-3p in spermatozoa development and motility potential [58].

miR-320a, located on chromosome 8, is highly expressed miRNA in epididymis and spermatozoa so has an important role in multiple reproductive processes [68]. Mmu-miR-320a (hsa-miR-320a) was exclusively expressed in murine Sertoli cells (SCs). The induction of apoptosis elicited miRNA expression was observed in meiotic spermatocytes and haploid spermatids. Forced expression of exogenous miR-320a in SCs may cause oligospermia and defection in sperm mobility, thus compromising the male fertility [69].

Advertisement

6. MicroRNAs: a key player in spermatogenesis

miRNAs play a vital role in the whole spermatogenic process, and their dysregulation ultimately leads to spermatogenic impairments [70]. To determine the role of miRNA, in male reproductive system, various models (mice) have been established by using the Cre/Lox system, which enables conditional manipulation of target genes, including knockout, insertion, replacement, activation, or modification of gene expression [44]. To understand the exact function of concerned miRNA in vivo, target deletion is the key step; however, in the absence of one miRNA, usually someone other has a compensatory role.

Double knockout of miR-34b/c and miR-449 in mice displayed severe disruption in spermatogenesis, dispensable fertilization, and pre-implantation development. Intracytoplasmic injection of miRNA-dKO sperm led to normal fertilization [52]. Deletion of miR34b/c and miR-449 in mice shows impaired spermatogenesis and spermatozoa maturation resulting in oligoasthenoteratozoospermia (OAT) [70]. Simultaneous inactivation of miR34b/c and miR449 in dKO mice leads to developmental defects and infertility [71, 72].

Salas-Huetos and his colleagues showed that 221 miRNAs were consistently detected in 10 healthy fertile men; among these most expressed, hsa-miR-191-5p was associated with sperm differentiation [73]. The first report on altered miRNA expression in patients with NOA showed that 154 miRNAs were differentially downregulated and 19 upregulated in NOA patients compared with fertile men [74]. Experimentally it is confirmed that specific genes are regulated by miRNAs involved in spermatogenesis. Decreased levels of estrogen-alpha (ERα) and a higher level of miR-100 and let-7b in oligospermia patients compared with fertile men indicated the regulatory role of miRNA over estrogen signaling [75, 76, 77]. Phosphatidylinositol-specific phospholipase C, X domain containing 3 (PLCXD3), expressed in spermatogenesis, was downregulated by miRNA34c-3p in severe oligozoospermic patients [78].

Conclusively, these results and some others (as shown in Table 1) confirm the regulatory role of miRNAs in male germ and somatic cells, and any change in their expression may lead to reproductive anomalies.

miRNAIsolation of miRNADysregulationCellular processesModel organismReferences
miR-19b and let-7aSeminal plasmaUp-regulatedFailure in spermatogenesisHuman
Oligozoospermia, non-obstructive and azoospermia
[79]
MiR-7-1-3p, miR-141 & miR-429Seminal plasmaUp-regulatedSpermatogenesis impairmentInfertile men[80]
hsa-miR-429SemenUp-regulatedBiomarker to access male infertilityHuman
Subfertile and non-obstructive azoospermia
[81]
hsa-miR-34b,
hsa-miR-34c-5p & hsa-miR-122
SemenUp-regulatedBiomarker to access male infertilityHuman
Subfertile and non-obstructive azoospermia
[81]
miR-34c-5p, miR-122, miR-146b-5p, miR-181a, miR-374b, miR-509-5p and miR-513-5pSeminal plasmaUp-regulated in asthenozoospermia
Down-regulated in azoospermia
Role in infertilityInfertile men
(Asthenozoospermia and
Azoospermia)
[39]
miR-27aSemenUpregulatedLow motility, abnormal MorphologyInfertile men
Asthenoteratozoospermia and normozoospermia
[59]
miR-17-92 cluster (miR-17, miR-18a, and miR-20a)TestesUpregulatedTesticular atrophy, decreased sperm productionMice[82]
miR-17-92 (Mirc1) and miR-106b-25 (Mirc3) clusterTestesKnock-outReduced testes and mild spermetogenic defectMice[83]
miR-146aRole in differentiation of spermatogonia.Mice[84]
miR-221/222Maintain the undifferentiated state of mammalian spermatogonia.[85]
miR-135aSpermatogonial stem cellsDownregulatedDownregulation of forkhead box protein O 1 (FOXO1) expression.Rats[86]
miR-449TestisMice[87]
MiR-518fSeminal plasmaDownregulatedEffected sperm quality and testicular environment exposure of Bisphenol AHuman[46]
miR- 23a/b- 3pSemenUpregulatedOligoasthenozoospermiaHuman[88]

Table 1.

miRNAs, the cellular processes affected by their dysregulation.

Advertisement

7. miRNAs: therapeutic advancements and infertility

In addition to the potential use of miRNAs as a biomarker, miRNAs have been considered therapeutic services against male infertility. miRNAs overexpression can be inhibited by anti-miR (laboratory designed molecule), or their downregulation might be supplemented with miRNAs mimics [89, 90]. A recent study showed that MRX34 (miRNA mimic of miR-34a) managed to suppress the tumor progression, and anti-miRs their inhibitory activity through binding to a miRNA, thus blocking it [91, 92, 93, 94].

In RNA-based therapeutics strategies, the challenge of degradation of the oligonucleotide by RNases in serum or cellular compartments is averted using two distinct approaches, either by altering oligonucleotide or through encapsulations of RNAs for protection (by adding phosphorothioate-like groups or developing delivery vehicles [95].

After several preclinical studies involving miRNA therapeutics, only a small number of miRNAs have moved into clinical development [96]. This is because of the many challenges, including;

  1. Difficulty in identifying the best miRNA candidates or miRNA targets for each disease type and the absence of efficient delivery systems in specific tissues. As described below, several novel delivery systems are currently being developed to develop miRNA therapeutics using a systematic approach.

  2. miRNA and its target identification by using bioinformatics tools, in silico interaction, and in vitro experimentation (cell culture, etc.)

    1. Selection of miRNA chemistry such as 2’-methyl, locked nucleic acid (LNC), or phosphothionate.

    2. Optimization of the delivery system by viral particle encapsulation or through liposomes.

    3. In vivo testing by using a model organism so that target localization, the efficiency of delivery, and toxicity level can be reported.

    4. Clinical trial for dose optimization, determination of the mode of drug delivery, and toxicity analysis.

  3. Another challenge is to provide stability, accuracy in in vivo targeting, avoid toxicity and thwarting off-target complex to a therapeutic miRNA [96].

  4. To achieve the efficient therapeutic functioning, correct in vivo dose concentration has not been determined yet [97].

Advertisement

8. Conclusion

miRNAs play a vital role in developing an organism and regulating many biological processes in the living organism. This chapter summarized the role of miRNAs in male infertility. Many miRNAs are reported as regulators of many functions, including developing reproductive organs, spermatogenesis, spermiogenesis, sperm mobility, and fertilization. Despite using miRNAs as biomarkers, miRNAs are also investigated as more reliable therapeutic agents for various diseases such as cancer, diabetes, and infertility.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Robay A, Abbasi S, Akil A, El-Bardisi H, Arafa M, Crystal RG, et al. A systematic review on the genetics of male infertility in the era of next-generation sequencing. Arab Journal of Urology. 2018;16(1):53-64
  2. 2. Bracke A, Peeters K, Punjabi U, Hoogewijs D, Dewilde S. A search for molecular mechanisms underlying male idiopathic infertility. Reproductive Biomedicine Online. 2018;36(3):327-339
  3. 3. Nieschlag E, Behre HM. Andrology: Male Reproductive Health and Dysfunction. Germany (Berlin): Springer Science & Business Media; 2001
  4. 4. Manfo F, Nantia E, Mathur P. Effect of environmental contaminants on mammalian testis. Current Molecular Pharmacology. 2014;7(2):119-135
  5. 5. Jodar M, Anton E. Small RNAs present in sand their role in reproduction. In: Reproductomics. United States: Academic Press; 2018. pp. 109-123
  6. 6. Li S, Xu Z, Sheng J. tRNA-derived small RNA: A novel regulatory small non-coding RNA. Genes. 2018;9(5):246
  7. 7. Trzybulska D, Vergadi E, Tsatsanis C. miRNA and other non-coding RNAs as promising diagnostic markers. Ejifcc. 2018;29(3):221
  8. 8. Cai X, Hagedorn CH, Cullen BR. Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA. 2004;10(12):1957-1966
  9. 9. Selbach M, Schwanhäusser B, Thierfelder N, Fang Z, Khanin R, Rajewsky N. Widespread changes in protein synthesis induced by microRNAs. Nature. 2008;455(7209):58-63
  10. 10. Sanger miRBase v.22.1; www.mirbase.org. [Accessed: May 08, 2022]
  11. 11. Baskerville S, Bartel DP. Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes. RNA. 2005;11(3):241-247
  12. 12. Vishnoi A, Rani S. MiRNA biogenesis and regulation of diseases: An overview. MicroRNA Profiling. 2017;1:270-282
  13. 13. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Research. 2009;19(1):92-105
  14. 14. Berezikov E, Cuppen E, Plasterk RH. Approaches to microRNA discovery. Nature Genetics. 2006;38(6):S2-S7
  15. 15. Khawar MB, Mehmood R, Roohi N. MicroRNAs: Recent insights towards their role in male infertility and reproductive cancers. Bosnian Journal of Basic Medical Sciences. 2019;19(1):31
  16. 16. Ambros V, Bartel B, Bartel DP, Burge CB, Carrington JC, Chen X, et al. A uniform system for microRNA annotation. RNA. 2003;9(3):277-279
  17. 17. Budak H, Bulut R, Kantar M, Alptekin B. MicroRNA nomenclature and the need for a revised naming prescription. Briefings in Functional Genomics. 2016;15(1):65-71
  18. 18. Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, et al. Dicer is essential for mouse development. Nature Genetics. 2003;35(3):215-217
  19. 19. Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA biogenesis, function and decay. Nature Reviews. Genetics. 2010;11(9):597-610
  20. 20. Le Bot N. miRNAs and cell-cycle control in ESCs. Nature Cell Biology. 2012;14:658
  21. 21. Cheng AM, Byrom MW, Shelton J, Ford LP. Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Research. 2005;33(4):1290-1297
  22. 22. Dennis LM. MicroRNAs in Early Embryonic Development: Dissecting the Role of miR-290 through miR-295 in the Mouse. United States: Massachusetts Institute of Technology; 2008
  23. 23. Latronico MV, Condorelli G. MicroRNAs and cardiac pathology. Nature Reviews. Cardiology. 2009;6(6):419-429
  24. 24. Esquela-Kerscher A, Slack FJ. Oncomirs—microRNAs with a role in cancer. Nature Reviews. Cancer. 2006;6(4):259-269
  25. 25. Hansen T, Olsen L, Lindow M, Jakobsen KD, Ullum H, Jonsson E, et al. Brain expressed microRNAs implicated in schizophrenia etiology. PLoS One. 2007;2(9):e873
  26. 26. Li JY, Yong TY, Michael MZ, Gleadle JM. Review: The role of microRNAs in kidney disease. Nephrology (Carlton). 2010;15(6):599-608
  27. 27. Sonkoly E, Wei T, Janson PC, Saaf A, Lundeberg L, Tengvall-Linder M, et al. MicroRNAs: Novel regulators involved in the pathogenesis of psoriasis. PloS One. 2007;2(7):e610
  28. 28. Eisenberg I, Eran A, Nishino I, Moggio M, Lamperti C, Amato AA, et al. Distinctive patterns of microRNA expression in primary muscular disorders. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(43):17016-17021
  29. 29. Kantharidis P, Wang B, Carew RM, Lan HY. Diabetes complications: The microRNA perspective. Diabetes. 2011;60(7):1832-1837
  30. 30. Kedde M, Agami R. Interplay between microRNAs and RNA-binding proteins determines developmental processes. Cell Cycle. 2008;7(7):899-903
  31. 31. Sendler E, Johnson GD, Mao S, Goodrich RJ, Diamond MP, Hauser R, et al. Stability, delivery and functions of human sperm RNAs at fertilization. Nucleic Acids Research. 2013;41(7):4104-4117
  32. 32. Pratt SL, Calcatera SM. Expression of microRNA in male reproductive tissues and their role in male fertility. Reproduction, Fertility, and Development. 2016;29(1):24-31
  33. 33. Etheridge A, Lee I, Hood L, Galas D, Wang K. Extracellular microRNA: A new source of biomarkers. Mutation Research, Fundamental and Molecular Mechanisms of Mutagenesis. 2011;717(1-2):85-90
  34. 34. Pal MK, Jaiswar SP, Dwivedi VN, Tripathi AK, Dwivedi A, Sankhwar P. MicroRNA: A new and promising potential biomarker for diagnosis and prognosis of ovarian cancer. Cancer Biology & Medicine. 2015;12(4):328
  35. 35. Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, et al. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One. 2008;3(11):e3694
  36. 36. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 2007;9(6):654-659
  37. 37. Sohel MH. Extracellular/circulating microRNAs: Release mechanisms, functions and challenges. Achievements in the Life Sciences. 2016;10(2):175-186
  38. 38. Turchinovich A, Burwinkel B. Distinct AGO1 and AGO2 associated miRNA profiles in human cells and blood plasma. RNA Biology. 2012;9(8):1066-1075
  39. 39. Wang C, Yang C, Chen X, Yao B, Yang C, Zhu C, et al. Altered profile of seminal plasma microRNAs in the molecular diagnosis of male infertility. Clinical Chemistry. 2011;57(12):1722-1731
  40. 40. Glinge C, Clauss S, Boddum K, Jabbari R, Jabbari J, Risgaard B, et al. Stability of circulating blood-based microRNAs–pre-analytic methodological considerations. PLoS One. 2017;12(2):e0167969
  41. 41. Jung M, Schaefer A, Steiner I, Kempkensteffen C, Stephan C, Erbersdobler A, et al. Robust microRNA stability in degraded RNA preparations from human tissue and cell samples. Clinical Chemistry. 2010;56(6):998-1006
  42. 42. Ge Q , Zhou Y, Lu J, Bai Y, Xie X, Lu Z. miRNA in plasma exosome is stable under different storage conditions. Molecules. 2014;19(2):1568-1575
  43. 43. Björkgren I, Petra S. The role of Dicer1 in the male reproductive tract. Asian journal of andrology 5. 2015;17(5):737
  44. 44. Björkgren I, Gylling H, Turunen H, Huhtaniemi I, Strauss L, Poutanen M, et al. Imbalanced lipid homeostasis in the conditional Dicer1 knockout mouse epididymis causes instability of the sperm membrane. The FASEB Journal. 2015;29(2):433-442
  45. 45. Li H, Huang S, Guo C, Guan H, Xiong C. Cell-free seminal mRNA and microRNA exist in different forms. PLoS One. 2012;7(4):e34566
  46. 46. Barbu MG, Thompson DC, Suciu N, Voinea SC, Cretoiu D, Predescu DV. The roles of micrornas in male infertility. International Journal of Molecular Sciences. 2021;22(6):2910
  47. 47. Khazaie Y, Esfahani MH. MicroRNA and male infertility: A potential for diagnosis. International Journal of Fertility & Sterility. 2014;8(2):113
  48. 48. Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. British Journal of Haematology. 2008;141(5):672-675
  49. 49. Tian H, Li Z, Peng D, Bai X, Liang W. Expression difference of miR-10b and miR-135b between the fertile and infertile semen samples (p). Forensic Science International: Genetics Supplement Series. 2017;6:e257-e259
  50. 50. Abu-Halima M, Backes C, Leidinger P, Keller A, Lubbad AM, Hammadeh M, et al. MicroRNA expression profiles in human testicular tissues of infertile men with different histopathologic patterns. Fertility and Sterility. 2014;101(1):78-86
  51. 51. Trzybulska D, Bobjer J, Giwercman A, Tsatsanis C. Serum microRNAs in male subfertility—Biomarkers and a potential pathogenetic link to metabolic syndrome. Journal of Assisted Reproduction and Genetics. 2017;34(10):1277-1282
  52. 52. Yuan S, Tang C, Zhang Y, Wu J, Bao J, Zheng H, 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(2):212-223
  53. 53. Braun RE. Post-transcriptional control of gene expression during spermatogenesis. Seminars in Cell & Developmental Biology. 1998;9(4):483-489
  54. 54. Roush S, Slack FJ. The let-7 family of microRNAs. Trends in Cell Biology. 2008;18(10):505-516
  55. 55. McIver SC, Roman SD, Nixon B, McLaughlin EA. miRNA and mammalian male germ cells. Human Reproduction Update. 2012;18(1):44-59
  56. 56. Paul A, Sadek ST, Mahesan AM. The role of microRNAs in human embryo implantation: A review. Journal of Assisted Reproduction and Genetics. 2019;36(2):179-187
  57. 57. Yao C, Yuan Q , Niu M, Fu H, Zhou F, Zhang W, et al. Distinct expression profiles and novel targets of MicroRNAs in human spermatogonia, pachytene spermatocytes, and round spermatids between OA patients and NOA patients. Molecular Therapy--Nucleic Acids. 2017;9:182-194
  58. 58. Muñoz X, Mata A, Bassas L, Larriba S. Altered miRNA signature of developing germ-cells in infertile patients relates to the severity of spermatogenic failure and persists in spermatozoa. Scientific Reports. 2015;5(1):1-2
  59. 59. Zhou JH, Zhou QZ, Yang JK, Lyu XM, Bian J, Guo WB, et al. MicroRNA-27a-mediated repression of cysteine-rich secretory protein 2 translation in asthenoteratozoospermic patients. Asian Journal of Andrology. 2017;19(5):591
  60. 60. Ye L, Evans J, Gargett CE. Lim1/LIM1 is expressed in developing and adult mouse and human endometrium. Histochemistry and Cell Biology. 2012;137(4):527-536
  61. 61. Fededa JP, Esk C, Mierzwa B, Stanyte R, Yuan S, Zheng H, et al. Micro RNA-34/449 controls mitotic spindle orientation during mammalian cortex development. The EMBO journal. 2016;35(22):2386-2398
  62. 62. Noveski P, Popovska-Jankovic K, Kubelka-Sabit K, Filipovski V, Lazarevski S, Plaseski T, et al. Micro RNA expression profiles in testicular biopsies of patients with impaired spermatogenesis. Andrology. 2016;4(6):1020-1027
  63. 63. Stenvang J, Petri A, Lindow M, Obad S, Kauppinen S. Inhibition of microRNA function by antimiR oligonucleotides. Silence. 2012;3(1):1-7
  64. 64. Chen D, Chen Z, Jin Y, Dragas D, Zhang L, Adjei BS, et al. MicroRNA-99 family members suppress Homeobox A1 expression in epithelial cells. PLoS One. 2013;8(12):e80625
  65. 65. Li J, Liu Y, Dong D, Zhang Z. Evolution of an X-linked primate-specific micro RNA cluster. Molecular Biology and Evolution. 2010;27(3):671-683
  66. 66. Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129(7):1401-1414
  67. 67. Belleannée C. Extracellular microRNAs from the epididymis as potential mediators of cell-to-cell communication. Asian Journal of Andrology. 2015;17(5):730
  68. 68. Feng R, Sang Q , Zhu Y, Fu W, Liu M, Xu Y, et al. MiRNA-320 in the human follicular fluid is associated with embryo quality in vivo and affects mouse embryonic development in vitro. Scientific Reports. 2015;5(1):1-9
  69. 69. Zhang LL, Ma J, Yang B, Zhao J, Yan BY, Zhang YQ , et al. Interference with lactate metabolism by mmu-miR-320-3p via negatively regulating GLUT3 signaling in mouse Sertoli cells. Cell Death & Disease. 2018;9(10):1-4
  70. 70. Papaioannou MD, Nef S. microRNAs in the testis: Building up male fertility. Journal of Andrology. 2010;31(1):26-33
  71. 71. Comazzetto S, Di Giacomo M, Rasmussen KD, Much C, Azzi C, Perlas E, et al. Oligoasthenoteratozoospermia and infertility in mice deficient for miR-34b/c and miR-449 loci. PLoS Genetics. 2014;10(10):e1004597
  72. 72. Wu J, Bao J, Kim M, Yuan S, Tang C, Zheng H, et al. Two miRNA clusters, miR-34b/c and miR-449, are essential for normal brain development, motile ciliogenesis, and spermatogenesis. Proceedings of the National Academy of Sciences. 2014;111(28):E2851-E2857
  73. 73. Salas-Huetos A, Blanco J, Vidal F, Mercader JM, Garrido N, Anton E. New insights into the expression profile and function of micro-ribonucleic acid in human spermatozoa. Fertility and Sterility. 2014;102(1):213-222
  74. 74. Lian J, Zhang X, Tian H, Liang N, Wang Y, Liang C, et al. Altered microRNA expression in patients with non-obstructive azoospermia. Reproductive Biology and Endocrinology. 2009;7(1):1
  75. 75. Safarinejad MR, Shafiei N, Safarinejad S. Association of polymorphisms in the estrogen receptors alpha, and beta (ESR1, ESR2) with the occurrence of male infertility and semen parameters. The Journal of Steroid Biochemistry and Molecular Biology. 2010;122(4):193-203
  76. 76. Carreau S, Genissel C, Bilinska B, Levallet J. Sources of oestrogen in the testis and reproductive tract of the male. International Journal of Andrology. 1999;22(4):211-223
  77. 77. Cheng C, Fu X, Alves P, Gerstein M. mRNA expression profiles show differential regulatory effects of microRNAs between estrogen receptor-positive and estrogen receptor-negative breast cancer. Genome Biology. 2009;10(9):1-7
  78. 78. Abhari A, Zarghami N, Shahnazi V, Barzegar A, Farzadi L, Karami H, et al. Significance of microRNA targeted estrogen receptor in male fertility. Iranian Journal of Basic Medical Sciences. 2014;17(2):81
  79. 79. Wu W, Hu Z, Qin Y, Dong J, Dai J, Lu C, et al. Seminal plasma microRNAs: Potential biomarkers for spermatogenesis status. Molecular Human Reproduction. 2012;18(10):489-497
  80. 80. Wu W, Qin Y, Li Z, Dong J, Dai J, Lu C, et al. Genome-wide microRNA expression profiling in idiopathic non-obstructive azoospermia: Significant up-regulation of miR-141, miR-429 and miR-7-1-3p. Human Reproduction. 2013;28(7):1827-1836
  81. 81. Abu-Halima M, Hammadeh M, Backes C, Fischer U, Leidinger P, Lubbad AM, et al. Panel of five microRNAs as potential biomarkers for the diagnosis and assessment of male infertility. Fertility and Sterility. 2014;102(4):989-997
  82. 82. Xie R, Lin X, Du T, Xu K, Shen H, Wei F, et al. Targeted disruption of miR-17-92 impairs mouse spermatogenesis by activating mTOR signaling pathway. Medicine. 2016;95(7):345-367
  83. 83. Tong MH, Mitchell DA, McGowan SD, Evanoff R, Griswold MD. Two miRNA clusters, Mir-17-92 (Mirc1) and Mir-106b-25 (Mirc3), are involved in the regulation of spermatogonial differentiation in mice. Biology of Reproduction. 2012;86(3):72
  84. 84. Huszar JM, Payne CJ. MicroRNA 146 (Mir146) modulates spermatogonial differentiation by retinoic acid in mice. Biology of Reproduction. 2013;88(1):15
  85. 85. Yang QE, Racicot KE, Kaucher AV, Oatley MJ, Oatley JM. MicroRNAs 221 and 222 regulate the undifferentiated state in mammalian male germ cells. Development. 2013;140(2):280-290
  86. 86. Moritoki Y, Hayashi Y, Mizuno K, Kamisawa H, Nishio H, Kurokawa S, et al. Expression profiling of microRNA in cryptorchid testes: miR-135a contributes to the maintenance of spermatogonial stem cells by regulating FoxO1. The Journal of Urology. 2014;191(4):1174-1180
  87. 87. Bao J, Li D, Wang L, Wu J, Hu Y, Wang Z, et al. MicroRNA-449 and microRNA-34b/c function redundantly in murine testes by targeting E2F transcription factor-retinoblastoma protein (E2F-pRb) pathway. The Journal of Biological Chemistry. 2012;287(26):21686-21698
  88. 88. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843-854
  89. 89. He Z, Jiang J, Kokkinaki M, Tang L, Zeng W, Gallicano I, et al. MiRNA-20 and mirna-106a regulate spermatogonial stem cell renewal at the post-transcriptional level via targeting STAT3 and Ccnd1. Stem Cells. 2013;31(10):2205-2217
  90. 90. Nedaeinia R, Sharifi M, Avan A, Kazemi M, Nabinejad A, Ferns GA, et al. Inhibition of microRNA-21 via locked nucleic acid-antimiR suppressed metastatic features of colorectal cancer cells through modulation of programmed cell death 4. Tumor Biology. 2017;39(3)
  91. 91. Beg MS, Brenner AJ, Sachdev J, Borad M, Kang YK, Stoudemire J, et al. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investigational New Drugs. 2017;35(2):180-188
  92. 92. Bezan A, Gerger A, Pichler M. MicroRNAs in testicular cancer: Implications for pathogenesis, diagnosis, prognosis and therapy. Anticancer Research. 2014;34(6):2709-2713
  93. 93. Costa AL, Lobo J, Jerónimo C, Henrique R. The epigenetics of testicular germ cell tumors: Looking for novel disease biomarkers. Epigenomics. 2017;9(2):155-169
  94. 94. Gillis AJ, Stoop HJ, Hersmus R, Oosterhuis JW, Sun Y, Chen C, et al. High-throughput microRNAome analysis in human germ cell tumours. The Journal of Pathology: A Journal of the Pathological Society of Great Britain and Ireland. 2007;213(3):319-328
  95. 95. Baumann V, Winkler J. miRNA-based therapies: Strategies and delivery platforms for oligonucleotide and non-oligonucleotide agents. Future Medicinal Chemistry. 2014;6(17):1967-1984
  96. 96. Van Rooij E, Kauppinen S. Development of micro RNA therapeutics is coming of age. EMBO Molecular Medicine. 2014;6(7):851-864
  97. 97. Am Hong C, Nam YS. Functional nanostructures for effective delivery of small interfering RNA therapeutics. Theranostics. 2014;4(12):1211

Written By

Mohsin Munawar, Irfana Liaqat and Shaukat Ali

Submitted: 24 May 2022 Reviewed: 26 July 2022 Published: 14 October 2022