Small Non-Coding RNAs in Extracellular Vesicles of Semen: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , ,

Reproductive dysfunction and urogenital malignancies represent a serious health concern in men. This is in part as a result of the absence of reliable non-invasive tests of diagnosis/prognosis. Optimizing diagnosis and predicting the patient’s prognosis will affect the choice of the most appropriate treatment and therefore increase the chances of success and the result of therapy, that is, it will lead to a more personalized treatment of the patient. 

  • urogenital diseases
  • male infertility
  • prostate cancer
  • semen
  • extracellular vesicles
  • sncRNAs
  • non-invasive biomarker
  • diagnosis

1. Semen Extracellular Vesicles

It is important to note that semen contains one of the highest reported concentrations of extracellular vesicles of any body fluid. These sEVs originate from the different organs of the male reproductive system: prostate (these sEVs, also known as prostasomes, represent 40% of semen sEVs), epididymis (epididysomes), seminal vesicles, and testicles [1].
Although it is developmentally mature, sperm exiting the seminiferous tubule still requires further modification. Changes in sperm morphology and post-testicular sperm function rely on sperm interaction with the intraluminal fluid during transit through the epididymis and vas deferens, suggesting that EVs in male reproductive biofluids may participate in intercellular communication after spermatogenesis. Additionally, in the female reproductive system, sperm acquisition of various functions including increased motility, transfer of cargoes, and ability to undertake the acrosome reaction is mediated through the interaction between the sperm and semen EVs derived from the organs of the male reproductive tract. Thus, semen EVs have a relevant physiological function related to the process of fertility [1] (point 1), but they are also involved in pathological states of the male reproductive tract such as the development and dissemination of prostate cancer [2][3] (point 2).

1.1. Semen sEVs and Fertility

Seminal plasma (SP) sEVs have been proposed as a means of selectively transporting and delivering various regulatory molecules to the female reproductive system to facilitate conception and contribute to fertilization [1] and embryo implantation [4], the latter by targeting endometrial stromal cells. Strikingly, as a first step, semen sEVs transfer their content directly to the male germ cell in the acid medium of the vagina [5] to protect sperm and modulate their activity. The numerous enzyme systems, small signaling molecules, and neuroendocrine markers associated with semen sEVs suggest that these vesicles directly or indirectly play a complex role in the regulation of sperm viability and function [6]; (reviewed in [7][8]).
(A)
Direct functions
-
Motility of sperm:
Sperm motility is vital for natural fertility and thus survival after ejaculation into the female reproductive tract [9]. Semen EVs improve the progressive motility of sperm [10] in the acid medium of the vagina [11][12] by increasing intracellular calcium.
-
Sperm capacitation and acrosome reaction:
Both sperm capacitation and the reaction of the acrosome are crucial steps for the sperm to acquire the ability to fertilize the oocyte, by allowing it to cross the zona pellucida of the oocyte to achieve fertilization [13]. Semen sEVs regulate these processes and prevent them from occurring prematurely after fusion between EVs and sperm [14]. On the other hand, this fusion also provides essential hydrolases for the acrosome reaction [15], as well as making the sperm more sensitive to progesterone, one of the stimulators of the acrosome reaction [16][17].
(B)
Indirect functions
Semen sEVs help to protect sperm after ejaculation from the immunological point of view, as they provide CD46 and CD59 proteins that help the sperm regulate complement activity of the female reproductive tract [18]; they also present antioxidant attributes that help avoid oxidative stress of sperm [19], antibacterial properties [20], and coagulant activity [21].

1.2. Semen sEVs and Prostate Cancer

Tumor-derived sEVs have been described as being involved in the development and dissemination of PCa [2][3]. In the tumor environment, EVs derived from malignant cells carry genetic and proteinic messages to the target cells which reduce their immune response. In this way, they influence the homeostasis of the surrounding environment and the progression of the tumor in the donor tissue. Reproductive system sEVs are directly secreted to semen and some of them can also reach blood and plasma. In this way, through the bloodstream, they contribute to the formation of the premetastatic niche in other body tissues (metastasis). The EVs secreted by cancer cells into the blood have a cancer-specific molecule content [22] such as a high proportion of oncoproteins and RNAs [23]. These EVs not only intervene in tumor progression, but also in the communication between tumors and the immune system, contributing to the drug-resistant character of cancer cells.

2. sncRNA EV Content as a Biomarker

sEVs are characterized by a high content of cholesterol and sphingomyelin, as well as by a very complex protein composition. EVs also carry RNA that can be transferred to other cells, modulating the function of the recipient cells [24][25]. This RNA can be found in the form of mRNAs and non-coding regulatory RNAs (including small and long ncRNAs) [24].
sEVs in semen contain a very important population of small ncRNAs (sncRNA, 20 to 100 nucleotides), which have a significant impact on the regulation of gene expression through a variety of epigenetic and post-transcriptional mechanisms [26]. RNA biotypes identified within sncRNAs include microRNAs (miRNAs), PIWI-interacting RNAs (piRNAs), and endogenous interfering RNAs (endo-siRNAs), which are critical regulators of germ cell development. There are also other more recently described molecules such as transfer RNA (tRNA)-derived small RNAs (tsRNAs) and ribosomal RNA (rRNA)-derived small RNAs (rsRNAs), whose role in reproduction and fertility is yet to be fully settled (reviewed in [27]). MiRNAs (21%) and tsRNAs (16%) represent the most abundant sEV sncRNAs in semen [1]. The sncRNA profile of sEVs in semen is unique and differs from the profile found in sEVs from other fluids [1].
Since sncRNAs are encapsulated inside sEVs, they remain stable and protected from RNAse. Due to these attributes, sEV sncRNAs are considered relevant for study as reliable biomarkers. Additionally, EVs contain molecules of the progenitor cell, so these EVs in the fluids can reflect the identity, characteristics, and health of the cell or tissue of origin. This is highly relevant in the context of the study of the content of sEVs as biomarkers. Specifically, in recent years, an increasing number of studies have been published that evaluate the miRNAs of sEVs in fluids as diagnostic biomarkers for physiological processes, such as the immune response, and pathological processes such as cancer, with promising results. The miRNAs participate in the post-transcriptional regulation of the expression of the genes: the specific binding of these small molecules of RNA to the transcripts affects their stability and activates transcript degradation. The tissue-specific expression profile and the high stability of these molecules mean that miRNAs, and other regulatory small RNAs, are becoming more and more relevant as biomarkers for the diagnosis or prognosis of different pathological conditions.
In recent years, it has been shown that ncRNAs play an important role in tumor progression and metastasis by their dissemination through fluids to regulate cancer cells and/or cancer stem cells, which suggests that extracellular ncRNAs in circulating sEVs can be used not only for diagnosis but also as prognostic biomarkers [28].

3. The Small RNA Population Signature Is Different between the Organs of the Reproductive Tract

As previously mentioned, semen contains a heterogeneous composition of sEVs, which are released from the different organs of the male reproductive tract, and these sEVs contain molecules from their cell of origin. Levels of these molecules can reflect the features and the health of that original cell. Certain sncRNA subtypes, such as miRNAs, have been associated with gene deregulation in diseases affecting organs of the male reproductive tract, including prostate cancer [29] and spermatogenic disorders [30]. These attributes support the idea that the study of the small RNA content of semen sEVs can reflect the pathophysiological state of male reproductive organs. The small RNA population varies between tissues; therefore, it is essential to know the expression pattern of the small RNAs in each of the organs of the male reproductive tract to be able to correctly interpret semen sEV sncRNA expression behavior and use it as a potential biomarker of urogenital disease.

3.1. Profile of Small RNA Populations in Human Testis

Spermatogenesis is a highly orchestrated developmental process which occurs in the testicular seminiferous tubules, and by which primordial germ-cells or spermatogonia develop into haploid spermatozoa. Production of sperm depends on precise developmental stage- and germ-cell type-specific gene expression. Thus, spermatogenesis is heavily dependent on post-transcriptional regulatory processes, and miRNAs have emerged as important regulators of these events [31][32][33]; miRNAs specifically expressed in the germline, such as miR-122, miR-449a, and miR-34b/c, are especially relevant [30][34][35]. In human testes, it is not only miRNAs that are highly abundant but also piRNAs, and tsRNAs at a lesser extent [36]. Interestingly, in physiological conditions, piRNAs are almost exclusively found in the male gonad, mainly in the germline [37][38][39].

3.2. Epididymis

The epididymis is a convoluted tube at the posterior part of the testis, where the sperm mature and are stored. When the sperm leaves the testis, it harbors a specific profile of small RNA, including all major RNA classes, with piRNAs being the most abundant sncRNAs. During its post-testicular maturation, the spermatozoon does not generate sncRNAs, but acquires them exogenously thanks to its fusion with extracellular vesicles [40][41][42][43]. In the epididymal lumen, epididymal epithelial cells secrete epididymosomes that contain small RNAs and proteins. Epididymosomes can deliver miRNAs, tsRNAs, and other sncRNAs to sperm that are transported from the proximal to distal epididymal region (from the caput to the cauda), resulting in changes in sncRNA patterns in mature sperm, with a notable enrichment of tRNA fragments (tRFs) [42] and other miRNAs [44] and a global loss of piRNAs.
Functional studies of epididymal sncRNAs have revealed a relevant role in epididymal region-specific gene regulation and epithelium–sperm interactions. Analysis of epididymosomes revealed that their cargo contains most small RNA species, including miRNAs, tsRNAs, and RNAs derived from snRNAs, snoRNAs, and rRNAs [45]. Some semen EVs contained miRNAs such as the miR-888 family (located in epididymis-enriched cluster of chromosome X), which have been described as being specifically enriched in the distal region of epididymis [34][35][46].

3.3. Prostate

Expression profiles of miRNAs obtained from malignant and benign prostate tissues differ significantly [47][48][49]. In recent years, PCa-related miRNAs have been identified and proposed as a tool for early and specific detection of the disease. Recent studies have revealed that abnormal expression of a large number of tsRNAs also occurs in PCa [50][51], contributing to DNA synthesis, cell viability, and cell proliferation.
Approximately 40% of semen is derived from prostatic tissue. At ejaculation, cauda epididymal spermatozoa are mixed with secretions from the reproductive tract accessory glands such as the prostate. The use of semen, rather than other fluids such as urine after prostate massage, is preferable as a non-invasive source of information of prostate health because semen represents a liquid biopsy from the whole prostatic gland whereas a sample is only obtained from the posterior part of the gland when urine is used [52].

4. Technical Issues to Consider for a Proper Interpretation of Results on Semen sEVs

4.1. Impact of Surgical Procedures for Contraception

The number of couples selecting male vasectomy as a contraceptive method has been increasing in recent years. The practice of vasectomy affects the concentration of certain semen sEV sncRNAs because the fluid from the testis and epididymis cannot reach semen, so whether the subject has undergone vasectomy should be taken into account for the accurate interpretation of results on semen for biomarker discovery/validation in urogenital diseases [53].

4.2. Impact of Extracellular Vesicle Isolation Methods and Preclinical Variables on Downstream sncRNA Analysis in Semen

Several exosome-EV isolation technologies have been optimized for their use in semen, and their EV purifying effectiveness (EV quantity, size, and transmembrane protein composition, as well as the quality of RNA they contain) and the impact on the down-stream analysis of miRNAs, compared against the standard use of ultracentrifugation, have been further evaluated [54]. A study provides evidence that the exosome-EV isolation method has a great impact on the analysis of the miRNAs they contain; it will be important to take this into consideration when implementing the use of semen exosomal/EV miRNAs as a diagnostic tool in the clinical laboratory. The observed over-representation of certain miRNAs from vesicles obtained by a particular isolation method suggests that some miRNAs are likely to be associated with specific vesicle sets enriched by this particular method of isolation. Therefore, the method used for EV isolation can produce variations in EV concentration as well as determine the composition of sEV subpopulations (such as microvesicles and exosomes) in nanovesicle preparation, introducing a bias for subsequent miRNA analysis. Accordingly, until clear markers for delineation between microvesicles and exosomes are established, the results of the analysis of miRNAs contained in EVs cannot be directly extrapolated between different EV isolation methods for clinical application due to the possibility of obtaining misleading results and conclusions.

4.3. Impact of Profiling Methods for Biomarker Testing

Due to the small size of sncRNAs and their low abundance in human fluids, sncRNA expression profiling is technically challenging. Since the discovery of sncRNA, many platforms have been developed for their expression quantification, specially tested for miRNAs, such as small RNA sequencing, microarray hybridization, and reverse transcription-quantitative PCR (RT-qPCR). Several studies in human biofluids, such as serum and plasma, have assessed platform performance in terms of reproductivity, sensitivity, accuracy, and specificity, and indicated that each method has its strengths and weaknesses, which can impact on differential expression analysis [55][56]. Other factors such as normalization and data imputation methods are of relevance to be considered, and will help for a standardized quantification of semen sEV sncRNAs for their clinical use as biomarkers for urogenital diseases.

This entry is adapted from the peer-reviewed paper 10.3390/ijms24065447

References

  1. Vojtech, L.; Woo, S.; Hughes, S.; Levy, C.; Ballweber, L.; Sauteraud, R.P.; Strobl, J.; Westerberg, K.; Gottardo, R.; Tewari, M.; et al. Exosomes in human semen carry a distinctive repertoire of small non-coding RNAs with potential regulatory functions. Nucleic Acids Res. 2014, 42, 7290–7304.
  2. Liu, C.M.; Hsieh, C.L.; Shen, C.N.; Lin, C.C.; Shigemura, K.; Sung, S.Y. Exosomes from the tumor microenvironment as reciprocal regulators that enhance prostate cancer progression. Int. J. Urol. Off. J. Jpn. Urol. Assoc. 2016, 23, 734–744.
  3. Shiao, S.L.; Chu, G.C.; Chung, L.W. Regulation of prostate cancer progression by the tumor microenvironment. Cancer Lett. 2016, 380, 340–348.
  4. Chen, K.; Liang, J.; Qin, T.; Zhang, Y.; Chen, X.; Wang, Z. The Role of Extracellular Vesicles in Embryo Implantation. Front. Endocrinol. 2022, 13, 809596.
  5. Ronquist, G.; Nilsson, B.O.; Hjertën, S. Interaction between prostasomes and spermatozoa from human semen. Arch. Androl. 1990, 24, 147–157.
  6. Kravets, F.G.; Lee, J.; Singh, B.; Trocchia, A.; Pentyala, S.N.; Khan, S.A. Prostasomes: Current concepts. Prostate 2000, 43, 169–174.
  7. Burden, H.P.; Holmes, C.H.; Persad, R.; Whittington, K. Prostasomes--their effects on human male reproduction and fertility. Hum. Reprod. Update 2006, 12, 283–292.
  8. Aalberts, M.; Sostaric, E.; Wubbolts, R.; Wauben, M.W.; Nolte-’t Hoen, E.N.; Gadella, B.M.; Stout, T.A.; Stoorvogel, W. Spermatozoa recruit prostasomes in response to capacitation induction. Biochim. Biophys. Acta 2013, 1834, 2326–2335.
  9. Saez, F.; Frenette, G.; Sullivan, R. Epididymosomes and prostasomes: Their roles in posttesticular maturation of the sperm cells. J. Androl. 2003, 24, 149–154.
  10. Stegmayr, B.; Berggren, P.O.; Ronquist, G.; Hellman, B. Calcium, magnesium, and zinc contents in organelles of prostatic origin in human seminal plasma. Scand. J. Urol. Nephrol. 1982, 16, 199–203.
  11. Fabiani, R.; Johansson, L.; Lundkvist, O.; Ronquist, G. Enhanced recruitment of motile spermatozoa by prostasome inclusion in swim-up medium. Hum. Reprod. 1994, 9, 1485–1489.
  12. Arienti, G.; Carlini, E.; Nicolucci, A.; Cosmi, E.V.; Santi, F.; Palmerini, C.A. The motility of human spermatozoa as influenced by prostasomes at various pH levels. Biol. Cell 1999, 91, 51–54.
  13. Ronquist, G.; Nilsson, B.O. The Janus-faced nature of prostasomes: Their pluripotency favours the normal reproductive process and malignant prostate growth. Prostate Cancer Prostatic Dis. 2004, 7, 21–31.
  14. Arienti, G.; Carlini, E.; Saccardi, C.; Palmerini, C.A. Role of human prostasomes in the activation of spermatozoa. J. Cell. Mol. Med. 2004, 8, 77–84.
  15. Breitbart, H.; Cohen, G.; Rubinstein, S. Role of actin cytoskeleton in mammalian sperm capacitation and the acrosome reaction. Reproduction 2005, 129, 263–268.
  16. Arienti, G.; Carlini, E.; Saccardi, C.; Palmerini, C.A. Nitric oxide and fusion with prostasomes increase cytosolic calcium in progesterone-stimulated sperm. Arch. Biochem. Biophys. 2002, 402, 255–258.
  17. Palmerini, C.A.; Saccardi, C.; Carlini, E.; Fabiani, R.; Arienti, G. Fusion of prostasomes to human spermatozoa stimulates the acrosome reaction. Fertil. Steril. 2003, 80, 1181–1184.
  18. Simpson, K.L.; Holmes, C.H. Presence of the complement-regulatory protein membrane cofactor protein (MCP, CD46) as a membrane-associated product in seminal plasma. J. Reprod. Fertil. 1994, 102, 419–424.
  19. Saez, F.; Motta, C.; Boucher, D.; Grizard, G. Prostasomes inhibit the NADPH oxidase activity of human neutrophils. Mol. Hum. Reprod. 2000, 6, 883–891.
  20. Andersson, E.; Sørensen, O.E.; Frohm, B.; Borregaard, N.; Egesten, A.; Malm, J. Isolation of human cationic antimicrobial protein-18 from seminal plasma and its association with prostasomes. Hum. Reprod. 2002, 17, 2529–2534.
  21. Carson, S.D.; De Jonge, C.J. Activation of coagulation factor X in human semen. J. Androl. 1998, 19, 289–294.
  22. Al-Nedawi, K.; Meehan, B.; Rak, J. Microvesicles: Messengers and mediators of tumor progression. Cell Cycle 2009, 8, 2014–2018.
  23. Melo, S.A.; Sugimoto, H.; O’Connell, J.T.; Kato, N.; Villanueva, A.; Vidal, A.; Qiu, L.; Vitkin, E.; Perelman, L.T.; Melo, C.A.; et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell 2014, 26, 707–721.
  24. Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659.
  25. Zomer, A.; Vendrig, T.; Hopmans, E.S.; van Eijndhoven, M.; Middeldorp, J.M.; Pegtel, D.M. Exosomes: Fit to deliver small RNA. Commun. Integr. Biol. 2010, 3, 447–450.
  26. Storz, G.; Altuvia, S.; Wassarman, K.M. An abundance of RNA regulators. Annu. Rev. Biochem. 2005, 74, 199–217.
  27. Zhu, Q.; Kirby, J.A.; Chu, C.; Gou, L.T. Small Noncoding RNAs in Reproduction and Infertility. Biomedicines 2021, 9, 1884.
  28. Lee, Y.R.; Kim, G.; Tak, W.Y.; Jang, S.Y.; Kweon, Y.O.; Park, J.G.; Lee, H.W.; Han, Y.S.; Chun, J.M.; Park, S.Y.; et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int. J. Cancer 2019, 144, 1444–1452.
  29. Doghish, A.S.; Ismail, A.; El-Mahdy, H.A.; Elkady, M.A.; Elrebehy, M.A.; Sallam, A.M. A review of the biological role of miRNAs in prostate cancer suppression and progression. Int. J. Biol. Macromol. 2022, 197, 141–156.
  30. 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. Sci. Rep. 2015, 5, 17991.
  31. Kotaja, N. MicroRNAs and spermatogenesis. Fertil.Steril. 2014, 101, 1552–1562.
  32. Papaioannou, M.D.; Nef, S. microRNAs in the testis: Building up male fertility. J. Androl. 2010, 31, 26–33.
  33. Hayashi, K.; Chuva de Sousa Lopes, S.M.; Kaneda, M.; Tang, F.; Hajkova, P.; Lao, K.; O’Carroll, D.; Das, P.P.; Tarakhovsky, A.; Miska, E.A.; et al. MicroRNA biogenesis is required for mouse primordial germ cell development and spermatogenesis. PLoS ONE 2008, 3, e1738.
  34. Hu, L.; Wu, C.; Guo, C.; Li, H.; Xiong, C. Identification of microRNAs predominately derived from testis and epididymis in human seminal plasma. Clin. Biochem. 2014, 47, 967–972.
  35. Barceló, M.; Mata, A.; Bassas, L.; Larriba, S. Exosomal microRNAs in seminal plasma are markers of the origin of azoospermia and can predict the presence of sperm in testicular tissue. Hum. Reprod. 2018, 33, 1087–1098.
  36. Rounge, T.B.; Furu, K.; Skotheim, R.I.; Haugen, T.B.; Grotmol, T.; Enerly, E. Profiling of the small RNA populations in human testicular germ cell tumors shows global loss of piRNAs. Mol. Cancer 2015, 14, 153.
  37. Girard, A.; Sachidanandam, R.; Hannon, G.J.; Carmell, M.A. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 2006, 442, 199–202.
  38. Kim, V.N. Small RNAs just got bigger: Piwi-interacting RNAs (piRNAs) in mammalian testes. Genes Dev. 2006, 20, 1993–1997.
  39. Li, X.Z.; Roy, C.K.; Dong, X.; Bolcun-Filas, E.; Wang, J.; Han, B.W.; Xu, J.; Moore, M.J.; Schimenti, J.C.; Weng, Z.; et al. An ancient transcription factor initiates the burst of piRNA production during early meiosis in mouse testes. Mol. Cell 2013, 50, 67–81.
  40. Chen, Q.; Yan, M.; Cao, Z.; Li, X.; Zhang, Y.; Shi, J.; Feng, G.H.; Peng, H.; Zhang, X.; Zhang, Y.; et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science 2016, 351, 397–400.
  41. García-López, J.; Hourcade Jde, D.; Alonso, L.; Cárdenas, D.B.; del Mazo, J. Global characterization and target identification of piRNAs and endo-siRNAs in mouse gametes and zygotes. Biochim. Biophys. Acta 2014, 1839, 463–475.
  42. Peng, H.; Shi, J.; Zhang, Y.; Zhang, H.; Liao, S.; Li, W.; Lei, L.; Han, C.; Ning, L.; Cao, Y.; et al. A novel class of tRNA-derived small RNAs extremely enriched in mature mouse sperm. Cell Res. 2012, 22, 1609–1612.
  43. Sharma, U.; Conine, C.C.; Shea, J.M.; Boskovic, A.; Derr, A.G.; Bing, X.Y.; Belleannee, C.; Kucukural, A.; Serra, R.W.; Sun, F.; et al. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science 2016, 351, 391–396.
  44. Amanai, M.; Brahmajosyula, M.; Perry, A.C. A restricted role for sperm-borne microRNAs in mammalian fertilization. Biol. Reprod. 2006, 75, 877–884.
  45. Trigg, N.A.; Eamens, A.L.; Nixon, B. The contribution of epididymosomes to the sperm small RNA profile. Reproduction 2019, 157, R209–R223.
  46. Belleannee, C.; Calvo, E.; Thimon, V.; Cyr, D.G.; Legare, C.; Garneau, L.; Sullivan, R. Role of microRNAs in controlling gene expression in different segments of the human epididymis. PLoS ONE 2012, 7, e34996.
  47. Stuopelyte, K.; Daniunaite, K.; Bakavicius, A.; Lazutka, J.R.; Jankevicius, F.; Jarmalaite, S. The utility of urine-circulating miRNAs for detection of prostate cancer. Br. J. Cancer 2016, 115, 707–715.
  48. Song, C.J.; Chen, H.; Chen, L.Z.; Ru, G.M.; Guo, J.J.; Ding, Q.N. The potential of microRNAs as human prostate cancer biomarkers: A meta-analysis of related studies. J. Cell. Biochem. 2018, 119, 2763–2786.
  49. Kristensen, H.; Thomsen, A.R.; Haldrup, C.; Dyrskjøt, L.; Høyer, S.; Borre, M.; Mouritzen, P.; Ørntoft, T.F.; Sørensen, K.D. Novel diagnostic and prognostic classifiers for prostate cancer identified by genome-wide microRNA profiling. Oncotarget 2016, 7, 30760–30771.
  50. Zhao, C.; Tolkach, Y.; Schmidt, D.; Muders, M.; Kristiansen, G.; Müller, S.C.; Ellinger, J. tRNA-halves are prognostic biomarkers for patients with prostate cancer. Urol. Oncol. 2018, 36, 503.e1–503.e7.
  51. Olvedy, M.; Scaravilli, M.; Hoogstrate, Y.; Visakorpi, T.; Jenster, G.; Martens-Uzunova, E.S. A comprehensive repertoire of tRNA-derived fragments in prostate cancer. Oncotarget 2016, 7, 24766–24777.
  52. Barceló, M.; Castells, M.; Pérez-Riba, M.; Bassas, L.; Vigués, F.; Larriba, S. Seminal plasma microRNAs improve diagnosis/prognosis of prostate cancer in men with moderately altered prostate-specific antigen. Am. J. Transl. Res. 2020, 12, 2041–2051.
  53. Barceló, M.; Castells, M.; Bassas, L.; Vigués, F.; Larriba, S. Semen miRNAs Contained in Exosomes as Non-Invasive Biomarkers for Prostate Cancer Diagnosis. Sci. Rep. 2019, 9, 13772.
  54. Mercadal, M.; Herrero, C.; López-Rodrigo, O.; Castells, M.; de la Fuente, A.; Vigués, F.; Bassas, L.; Larriba, S. Impact of Extracellular Vesicle Isolation Methods on Downstream Mirna Analysis in Semen: A Comparative Study. Int. J. Mol. Sci. 2020, 21, 5949.
  55. Mestdagh, P.; Hartmann, N.; Baeriswyl, L.; Andreasen, D.; Bernard, N.; Chen, C.; Cheo, D.; D’Andrade, P.; DeMayo, M.; Dennis, L.; et al. Evaluation of quantitative miRNA expression platforms in the microRNA quality control (miRQC) study. Nat. Methods 2014, 11, 809–815.
  56. Hong, L.Z.; Zhou, L.; Zou, R.; Khoo, C.M.; Chew, A.L.S.; Chin, C.L.; Shih, S.J. Systematic evaluation of multiple qPCR platforms, NanoString and miRNA-Seq for microRNA biomarker discovery in human biofluids. Sci. Rep. 2021, 11, 4435.
More
This entry is offline, you can click here to edit this entry!
Video Production Service