Czasopismo jest indeksowane w Index Copernicus ICV 2017:69,63

Czasopismo Polskiego Towarzystwa Andrologicznego Journal of Polish Society of Andrology

Tom 8 • Numer 1 (Suplement 1) • Czerwiec 2021

STEROIDOGENIC ACTIVITY OF LEYDIG CELLS AND ULTRASTRUCTURE OF THEIR MITOCHONDRIA

Małgorzata Brzoskwinia1, Laura Pardyak2, Alicja Kamińska1, Wacław Tworzydło3, Anna Hejmej1, Sylwia Marek1, Barbara Bilińska1

1Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Poland; 2Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, Poland; 3Developmental Biology and Invertebrate Morphology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Poland e-mail: m.brzoskwinia@doctoral.uj.edu.pl

Androgens, including testosterone, play a vital role in the regulation of male reproduction mediating their biological effects through binding to the androgen receptors (ARs). Leydig cells are the main source of intratesticular testosterone, which is synthesized, like in other steroid-producing cells, from a common substrate cholesterol. Transport of cholesterol into the mitochondria and its conversion to testosterone require the presence of carrier proteins and steroidogenic enzymes (Haider: Int Rev Cytol. 2004, 233, 181–241). It should be emphasized that correct synthesis of androgens depends on the structural condition of Leydig cells, and any disturbance in the biosynthesis and availability of steroids may affect the secretory activity of these cells. In our studies we used flutamide – a pure non-steroidal anti-androgen that blocks receptor binding of androgen, disturbing the action of endogenous testosterone (Labrie: Cancer. 1993, 72, 3816–3827). Although the long-term effects of this anti-androgen on male reproduction are well characterized, its short-term effects on the male gonad are still elusive. Recent findings by Bilinska demonstrated that short-term exposure to flutamide applied to adult rats, alters the blood-testis barrier integrity (Zarzycka et al.: Andrology. 2015, 3, 569-581; Chojnacka et al.: Reprod Biol Endocrinol. 2016, 14, 14). Experiments by Sarabay and coworkers (Sarrabay et al.: Toxicol Appl Pharmacol. 2015, 289, 515–524) showed that blockage of AR leads to inhibition of the negative feedback in the hypothalamic-pituitary-gonadal axis and enhancement of Leydig cell steroidogenic activity. In this context, direct effects of flutamide at the gonadal level, specifically on the steroidogenic function of Leydig cells and its potential autoregulation is of interest. The results of our latest studies (Brzoskwinia et al.: J Mol Sci. 2020, 21, 4439) showed that disruption of androgen signaling by flutamide leads to an increase in plasma luteinizing hormone, cholesterol, and testosterone concentrations. Moreover, we demonstrated increase in the intratesticular levels of testosterone that correspond well to increase in the expression level of several mitochondrial proteins required for the first step of steroidogenesis. Morphometric analysis of semithin testis sections revealed Leydig cells’ hypertrophy, while ultrastructural analysis showed additionally changes in the distribution and morphology of their mitochondria. Mutual relationships between cell organelles, visualized due to a computer aided 3D reconstruction of serial ultrathin sections, revealed that mitochondria fuse forming local mitochondrial networks. Moreover, morphometric analysis confirmed that percentage of the cell area occupied by mitochondria was substantially higher after flutamide exposure. In the light of this, we postulate that enhanced steroidogenic activity of Leydig cells involves multiplication of Leydig cell’ mitochondria and consequent formation of highly active mitochondrial networks. Upregulation of dynamin-related protein 1 (Drp1) expression in Leydig cells supports this assumption. Our results confirm the idea that blockage of AR, initiates a compensatory mechanism that causes enhancement of the steroidogenic activity of Leydig cells. Supported by a grant OPUS12 2016/23/B/NZ4/01788 from National Science Centre.

GENETIC CONTROL OF TESTICULAR CELL FUNCTION IN HEALTH AND DESEASE

Michał Duliban1, Piotr Pawlicki2, Agnieszka Miloń1, Ewelina Górowska-Wójtowicz1, Barbara Bilińska1, Małgorzata Kotula-Balak2

1Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Poland, 2University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow, Poland e-mail: michal.duliban@doctoral.uj.edu.pl

In the male gonad, estrogens act in paracrine and autocrine way to regulate spermatogenesis and steroidogenesis. Nowadays it is known that in Leydig cells estrogen signaling is mediated by various types of estrogen receptors (Pardyak et al.: Tissue Cell. 2016, 48, 432-41; Chimento et al.: Cells. 2020, 9, 2115.). Pathological Leydig cells may produce an abnormal amount of estrogens, e.g. human tumor Leydig cells are characterized by an overproduction of estrogens and their metabolites (Van Der Gucht et al.: Clin. Nuclear Med. 2018, 43, 41–43.). Our research has demonstrated differences in gene expression in healthy and tumor human Leydig cells (Kotula-Balak et al.: unpublished). Detailed analysis revealed disrupted expression of genes associated with such processes as: apoptosis, blood vessel development or cell stress in tumor Leydig cells. A mutation in the TP53 gene, leading to impaired apoptosis, has also been described in patients with testicular tumors (Stecher et al.: Pediatr Blood Cancer. 2008, 50, 701–703). In addition, downregulated expression of genes encoding heat shock proteins was noted. On the basis of next generation sequencing (NGS) analysis (novel research technique that allows for quick and accurate analysis of the entire cell, tissue or organ genome) carried out in mouse testes with disturbed non-classical estrogen signaling (via estrogen-related receptors and membrane estrogen receptors) changes in the expression of genes involved in processes important for individual testicular cell type function e.g. immune response or post-translational protein modifications were revealed (Duliban et al.: Reprod Fertil Dev 2020, 32, 903-913). The breakdown of testis immune balance may contribute to fertility disorders, and disruption of post-translational protein modification that may lead to tumorigenesis (Han et al.: Int. J Oncol. 2018, 52, 1081–1094). It is confirmed by significant changes in the expression of several genes e.g. Bmi1 (polycomb ring finger oncogene) and Npm1 (nucleophosmin 1), which actively participate in acceleration of tumor growth. The application of NGS technique allows to obtain comprehensive data that provide cross-sectional information on the activity of testicular transcriptome and can be used to initiate further studies at the proteome or epigenome levels, including the designing of new diagnostic tests. Supported by grants SONATA BIS5 2015/18/E/NZ4/ 00519 and OPUS12 2016/23/B/NZ4/01788 from National Science Centre, Poland. 27 SYMPOSIUM OF SCIENTIFIC TRANINING OF THE POLISH SOCIETY OF ANDROLOGY – 22 n d DAY OF ANDROLOGY

THE SIGNIFICANCE OF HUMAN SPERM CHROMATIN DAMAGE IN IN VITRO PROCEDURES

Kamil Gill1, Aleksandra Rosiak-Gill1,2, Małgorzata Piasecka1

Department of Histology and Developmental Biology, Pomeranian Medical Uniwersity, Szczecinie, Poland; 2VitroLive Fertility Clinic in Szczecin, Poland e-mail: kamil.gill@pum.edu.pl

Over the last decades, couple infertility has become one of the emerging global public health issue and classified by the World Health Organization as a one of the civilization disease (Agarwal et al.: World J Mens Health. 2019, 37 (3), 296–312). In response to this problem development of the assisted reproductive techniques, especially in vitro fertilization is observed and for many couples this treatment is the only chance to have biological offspring. Although the clinical relevance of basic semen analysis during the in vitro procedure is limited, the influence of the male factor on reproductive success is not completely eliminated. This thesis is confirmed by outcomes of in vitro fertilization procedures in which pregnancy was not achieved despite the lack known female factor. Therefore, many authors underline the need to develop and introduce molecular biomarkers of male infertility assessment Agawrwal i et al.: Int J Mol Sci. 2020, 21(11), 3882; Jerre et al.: Fertil Steril. 2019, 112(1), 46–53; Tang et al.: J Gynecol Obstet Hum Reprod. 2020, 10, 101868). The goal of these methods is to enable the prognosis of the chances of getting pregnant in vitro procedures. In this aspect, the quality of sperm chromatin is of particular importance. Its structural abnormalities can affects fertilization process, creation and development of the pronuclei as well as early embryo development observed before activation of paternal genome (4-cell embryo) – early paternal effect. Moreover, the late paternal effect at the 4–8-cell stage of the embryo (when the paternal genome is fully activated) up to blastocyst stage is found. For that, many researchers indicate that low quality of sperm chromatin may result in: 1) a decrease in the percentage of normal fertilized oocytes and percentage of morphologically normal embryos, 2) a decrease in the percentage of achieved pregnancies, and 3) an increase in proportion of miscarriages or even birth defects and epigenetic disorders in the offspring (Okada and Yamaguchi: Cell. Mol. Life Sci. 2017, 74, 1957–1967; Tesarik: Reprod Biomed Online. 2005, 10(3), 370–375). However, not all researchers confirmed a negative impact of sperm chromatin damages on the reproductive success after in vitro fertilization (Green et al.: J Assist Reprod Genet. 2020, 37(1), 71–76; Yang et al.: Transl Androl Urol. 2019, 8(4), 356–365). This inconsistency may results from different methods of male gametes selection, application of diverse sperm chromatin tests and the use of various statistical tools. Moreover, a large heterogeneity of the groups enrolled to the studies is observed. Also, it cannot be ignored fact that the oocyte is equipped with DNA repair mechanism. First of all, DNA single strand breaks are repaired. This process may begin at the pronuclei stage of zygote and be continued until the blastocyst stage. If the sperm DNA damage does not exceed the repair capacity of the oocyte, the impact of this damage may be limited (Ribas-Maynou and Benet: Genes (Basel). 2019, 10(2), 105). Therefore, the ambiguity of the influence of sperm chromatin abnormalities on reproductive success initiates a discussion about clinical value of sperm nuclear DNA tests. Study supported by the Pomeranian Medical University (no. WNoZ-322-01/S/19/2021).

DIRECT INTERCELLULAR COMMUNICATION IN SEMINIFEROUS EPITHELIUM AND ITS FUNCTION AFTER ANDROGEN WITHDRAWAL

Anna Hejmej, Alicja Kamińska, Sylwia Marek, Barbara Bilińska

Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University

Intercellular communication in the male gonads is of particular interest due to its key role in maintaining the proper course of spermatogenesis. Initially, research focused on paracrine communication (Bilińska et al.: Biol Cell. 1997, 89, 435–442), but further studies revealed that direct cell-cell interactions are equally important. Direct communication is based on the transport of small molecules through gap junctions or on interactions between molecules located in the membranes of neighboring cells (juxtacrine signaling). Gap junctions consist of proteins connexins, that form channels connecting the cytoplasm of adjacent cells. In the mammalian testis, connexin 43 (Cx43) is the main component of gap junctions. Studies using Sertoli cellspecific Cx43 knockout mice revealed the critical role of this protein in Sertoli cell differentiation and initiation of spermatogenesis (Brehm et al.: Am J Pathol. 2007, 171, 19–31; Chojnacka et al.: Reprod Biol. 2012, 12, 341–346). In human and rodent testes with impaired spermatogenesis, disturbances in Cx43 expression were demonstrated (Kotula-Balak et al.: Eur J Histochem. 2007, 51, 261–268). In rats, androgen signaling inhibition with antiandrogen flutamide resulted in decreased expression and delocalization of Cx43 at the blood-testis barrier. It was accompanied by altered distribution of zonula occludens-1 (ZO-1) and structural abnormalities of basal ectoplasmic specialization. This indicates that Cx43-based communication between Sertoli cells depends on the availability of androgens (Chojnacka et al.: Reprod Biol Endocrinol. 2016, 31, 14, 14). Juxtacrine communication, another mechanism of direct intercellular communication in the testis, involves the Notch signaling pathway. Activation of this pathway is triggered by binding of the membrane receptor Notch to specific ligands located in the membranes of the neighboring cell. This leads to receptor proteolysis and translocation of its intracellular domain into the nucleus where it regulates the transcription of Hes (hairy/enhancer of split) and Hey (Hes-related with YRPW motif) genes. The optimal activity of Notch pathway determines spermatogonial stem cell differentiation, influences further steps of spermatogenesis, and impacts fertility (Garcia et al.: Development. 2014, 141, 4468–4478; Murta et al.: PLoS One. 2014, 9, e113365). Disturbed expression of Notch pathway factors was found in testicular cancer and in men with non-obstructive azoospermia (Hayashi et al.: J Androl. 2001, 22, 999–1011; Hayashi et al.: Tumour Biol. 2004, 25: 99–105). Our recent studies indicate that androgen withdrawal, induced by androgen receptor blockade or selective elimination of Leydig cells, altered the expression of Notch1 and Notch2 receptors, as well as their activated forms in rat testes. Moreover, the expression of the effector genes Hey1, Hes1, and Hes5 was changed following androgen withdrawal, which indicates disturbed activity of Notch pathway. Androgens can therefore be considered as regulators of Notch signaling (Kamińska et al.: Reprod Biol Endocrinol. 2020, 18, 30). Taken together, the control of direct intercellular communication in the seminiferous epithelium is an important aspect of androgen activity during spermatogenesis. Thus, abnormalities in both gap junction communication and juxtacrine signaling may underlie the disturbance of spermatogenic function under conditions of limited availability of androgens. Supported by grants HARMONIA3 2012/06/M/NZ4/00146 and OPUS13 2017/25/B/NZ4/01037 (National Science Centre, Poland).

IMMUNOLOGICAL ASPECTS OF MALE INFERTILITY

Katarzyna Jankowska

The Department of Endocrinology, Center of Postgraduate Medical Education, Bielanski Hospital, Warsaw, Poland e-mail: katarzynakamilajankowska@gmail.com

Male infertility can be caused by an immune disorder. Immune infertility in men is defined as the presence of one or both partners of an immune response against semen (Dondero et al.: In: Oxford Textbook of Endocrinology and Diabetes, Oxford University Press, 2011, DOI: 10.1093/med/9780199235292.003.9100). WHO guidelines list immunological factors as one of the causes of male infertility (WHO: Manual for the Standardized Investigation and Diagnosis of the Infertile Couple. Cambridge University Press, 2000). It is estimated that about 15% of male infertility is immunerelated, either due to recurring infections or an abnormal autoimmune response involving the epididymis, prostate, or testes. This ratio is likely to be underestimated as 30% of male infertility cases are reported to be idiopathic. Unlike orchitis, epididymitis is a common condition. The cause of epididymitis is often bacterial infections, of particular importance are the sexually transmitted Chlamydia trachomatis and Neisseria gonorrhoeae in young men and the intestinal Escherichia coli and Enterococcus faecalis in older men. It has been shown that these infections, even if successfully treated, can cause narrowing of the epididymal duct, decreased sperm count and azoospermia in up to 40% of patients. Clinical data demonstrate the need for an effective and precisely controlled immune response to pathogens in the epididymis. Antisperm antibodies (ASA) may interfere with the fertilizing ability of the sperm: reduce sperm motility, reduce the ability to pass through the secretion of the genital tract in a woman, reduce the ability to penetrate the oocyte, cause sperm agglutination. ASA lead to reduced sperm motility, i.e. asthenozoospermia. However, immunological infertility should also be taken into account in normozoospermia, especially when sperm functional tests are not normal. The guideline of the European Association of Urology (EAU) recommend that diagnostics for immunological disorders should be considered when the semen test result is twice outside the reference range. This examination is performed when other causes of the abnormal seminogram have been excluded (e.g. urogenital infection). It is currently recommended to perform a mixed antiglobulin reaction (MAR test) or an immunobead semen test (Jungwirth et al.: Male Infertility In: EAU Guidelines, http://uroweb.org/guideline/male-infertility). The MAR test is used to detect sperm antibodies in semen. The test should be considered when the examination of semen shows poor sperm motility or agglutination of sperm. Genitourinary tract infection must be ruled out before hand, as it may induce the presence of antibodies. If asthenozoospermia or sperm agglutination are still present, despite the lack of infection or after the infection has healed, immune-mediated infertility should be excluded. The MAR test involves adding latex beads coated with IgA or IgG antibodies and a mixture of anti-IgA or anti-IgG antibodies to the semen sample. If there are IgA or IgG antibodies on the sperm cells, the spermatozoa are joined with the beads, which can be seen in the microscope. A result of more than 50% of sperm associated with the beads is considered positive and indicates the presence of ASA in an amount that interferes with the function of the sperm (possibility of penetration through the cervical mucus and also the ability to fertilize). This test can only be performed when there are sperm cells in the semen that show movement. The immunobead test has a similar meaning. It is more accurate than the MAR test, but the execution is more laborious. The immunobead test is not performed with a fresh sample of semen, but with semen from which elements that may mask the presence of antibodies have been removed. The principle of the test is similar to the MAR test (the connection of sperm with latex beads is observed in a microscope). The evaluation of the immunobead test result is also similar: ≥50% of sperm associated with the beads are positive, indicating the presence of ASA in an amount that interferes with sperm function. This test can only be performed when there are sperm cells in the semen that show movement. The role of epididymides is emphasized in the regulation of the immune response (Voisin et al.: Asian J Androl. 2019, 21(6), 531–539). Epididymal epithelial cells have developed mechanisms that combat pathogens, including the expression of various TLRs (toll-like receptors), antimicrobial molecules (nitric oxide, IDO, β-defensin, etc.), and pro-inflammatory cytokines (IL-1, IL-6, etc.). Interstitial lymphocytes B in the epididymides may be responsible for the secretion of local IgA or they may phagocytose bacteria coated with antibodies and limit their spread in the tissue and induce specific effector lymphocytes T. Activated B cells activate helper T cells. Some an antigen presenting cells (APCs) may migrate to the draining lymph node to trigger the recruitment of effector cells to infected epididymides. When the epithelium is damaged by infection, some CX3CR1+ CD11c+ monocytes may be involved in the elimination of disrupted cells or pathogens. Newly identified lymphocytes T in the epididymis can be activated by APCs or directly by infected cells. Once activated, they can become cytotoxic, contributing to the clearance of bacteria. Probably the epididymis is a potential immune reservoir of cytotoxic cells during infection. Immune infertility is found in 8–20% of infertile men. Sperm antibodies appear as a result of trauma, infection of the testicle or the epididymis that breaks the blood-testicle barrier (e.g. surgery, infections, varicocele). Not all sperm antibodies cause infertility. It is important whether these antibodies bind to antigen epitopes necessary for the fertilization of the egg. Currently, it is recommended to perform tests detecting anti-sperm antibodies: MAR test or immunobead test. Depending on the history (psoriasis, vitiligo, thyroid diseases, inflammatory bowel diseases, rheumatic diseases), screening for other autoimmune diseases (ANA, anti-TPO, antiTg, anti-CCP, celiac disease) may be considered. In many autoimmune diseases, causal treatment (e.g. biological treatment) is possible. Recently, more attention has been paid to the relationship between the state of the immune system and fertility. In 2018 Brubaker et al. (Brubaker et al.: Andrology. 2018, 6(1), 94–98), showed that men with infertility had a significantly higher incidence of autoimmune diseases: rheumatoid arthritis, multiple sclerosis, psoriasis, thyroiditis and Graves’ disease. Hypogonadism may be one of the components of the autoimmune polyendocrine syndrome. On the other hand, other publications (Mouvis et al.: Semin Arthritis Rheum. 2019, 48(5), 911–920) show that in men with rheumatological diseases who are treated with, for example, anti-TNF (anti-tumor necrosis factor) drugs, an improvement in sperm parameters and an increase in the pregnancy rate in partners are observed. This would confirm the importance of autoimmune processes in male infertility. This very interesting information requires further research and clinical observations. Piotr Jędrzejczak AZOOSPERMIA DIAGNOSTICS AND THERAPEUTIC PLANNING Division of Infertility and Reproductive Endocrinology, Poznan University of Medical Sciences, Poland e-mail: piotrjedrzejczak@gmail.com Azoospermia affects about 1% of men. It is a serious challenge for doctors involved in reproductive medicine. During the presentation, the causes of the azoospermia will be presented, as well as diagnostic methods important in detecting the various forms of this disease. Particular attention will be paid to the current therapeutic options in men with a complete absence of sperm in the ejaculate. Finally, an algorithm for the management of patients with azoospermia according to modern recommendations will be presented.

ROLE AND HORMONAL REGULATION OF JAGGED AND DELTA-LIKE PROTEINS IN RODENT SERTOLI CELLS

Alicja Kamińska1, Sylwia Marek1, Małgorzata Brzoskwinia1, Laura Pardyak2, Anna Hejmej1, Barbara Bilińska1

1Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland, 2Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, Poland e-mail: ala.kaminska@uj.edu.pl

Mammalian spermatogenesis is a process controlled by Sertoli cells, considered the key mediators of androgen action in the seminiferous epithelium. Androgens act predominantly by activating the classical intracellular androgen receptor (AR). It is now well established that the proper AR signaling in Sertoli cells is required for maintenance of the blood-testis barrier, completion of meiosis and spermiation (O’Hara and Smith: Best Pract. Res. Clin. Endocrinol. Metab. 2015, 29, 595–605). In addition, androgens regulate the process of spermatogenesis through non-classical signaling pathways related to the induction of membrane receptors. In 2014, a member of the zinc transporter family, Zrt- and Irtlike protein 9 (ZIP9), was identified as a novel membrane androgen receptor (Berg et al.: Endocrinology. 2014, 155, 4237–4249). It has been shown that testosterone, acting through ZIP9, is involved in the regulation of the bloodtestis barrier proteins in Sertoli cells (Bulldan et al.: Cell Signal. 2016, 28, 1075–1085). Recent studies have confirmed that ZIP9 is localized in Sertoli cells of mouse and bank vole seminiferous epithelium (Kamińska et al.: Andrology. 2020, 8, 457–472; Profaska-Szymik et al.: Int. J. Mol. Sci. 2020, 21, 6888). Delta-like (DLL) and Jagged (JAG) proteins are membrane ligands activating the Notch signaling pathway involved in juxtacrine communication in the seminiferous epithelium. In mouse and rat testes, the presence of DLL1, DLL4, and JAG1 ligands has been demonstrated in both Sertoli cells and germ cells (Kaminska et al.: Reprod Biol Endocrinol. 2020, 18, 30; Murta et al.: PLoS One. 2013, 8, e72767). Activation of the Notch pathway by these ligands specifically modulates the response of Sertoli cells to androgens, influencing AR and ZIP9 expression. Moreover, it has been found that disturbances in the activity of the Notch pathway led to deregulation of the expression of blood-testis barrier proteins – claudins (Kamińska et al.: Andrology. 2020, 8, 457–472). Therefore, the Notch pathway can be considered as an important factor influencing androgen signaling in Sertoli cells. Further analysis showed that the expression of Notch pathway ligands (DLL1, DLL4, and JAG1) in the male gonad is regulated by androgens. In vitro studies have clarified the mechanism of this regulation, providing evidence for the involvement of both AR and ZIP9 in this process. These results indicate a role of classical and non-classical androgen signaling in the control of the Notch pathway. Summing up, our results demonstrated a bidirectional interaction between Delta-like and Jagged proteins and androgen signaling in rodent seminiferous tubules. The crosstalk between these mechanisms regulates the physiology of Sertoli cells, and its disruption may affect key processes in the seminiferous epithelium, such as the function of blood-testis barrier. Supported by grants MINIATURA1 2017/01/X/NZ4/00285 and OPUS13 2017/25/B/NZ4/01037 (National Science Centre, Poland).

NON-CLASSIC ESTROGEN SIGNALING IN THE TESTICULAR INTERSTITIUM

Małgorzata Kotula-Balak1, Agnieszka Miłoń1, Piotr Pawlicki1, Ewelina Górowska-Wójtowicz2, Michał Duliban2, Barbara Bilińska2



Sposób przygotowania manuskryptu

1University Centre of Veterinary Medicine JU-UA, University of Agriculture in Kraków, Poland, 2Department of Endocrinology, Institute of Zoology and Biomedical Research, Jag

Based on numerous epidemiological and experimental studies, it is known that estrogen signaling through classical and non-classical estrogen receptors is necessary for the proper function of the male reproductive system. At the end of the 20th century, canonical estrogen receptors α and β (ERα and ERβ; estrogen receptor α and β) have been identified and their role in various types of testicular cells was determined using animal experimental models and in vitro studies (Bilińska et al.: Mol Cell Endocrinol. 2001, 10, 189–198; Hess.: Reprod Biol Endocrinol. 2003, 9, 52). The expression of ERα and ERβ genes has been demonstrated in human testis, including pathological conditions, but research aimed at determining the pattern of protein localization of these receptors is still ongoing. Currently, research work is focused on explaining the role of non-classical estrogen receptors, e.g. estrogenrelated receptors (ERRα, ERRβ and ERRγ – estrogenrelated receptor α, β and γ) and membrane estrogen receptor (GPER; G-protein coupled estrogen receptor). In recent years, for the first time, the expression of ERR in mouse Leydig cells in vitro and in vivo conditions has been confirmed. Changes in proliferation, apoptosis and migration were observed in tumor Leydig cells with modulated ERRα, ERRβ or ERRγ activity (Kotula-Balak et al.: Tissue Cell. 2018, 52, 78–91). Species-specific expression of GPER has been demonstrated in all types of mammalian testicular cells, including human ones (Fietz et al.: Methods Mol Biol. 2016, 1366, 189–205; Kotula-Balak et al.: Cell Tissue Res. 2018, 374, 389–412). An increase in GPER expression and changes in estrogen levels were noted in rodent and human tumor Leydig cells (Górowska-Wójtowicz et al.: Tissue Cell. 2019, 61, 51–60). Inactivation of both ERR and GPER in mouse testes resulted in increased volume of interstitial tissue and enhanced expression of ERRα, ERRβ or ERRγ genes. Concomitantly, there was an increase in the number of telocytes – newly discovered regulatory cells of interstitial tissue (Pawlicki et al.: Protoplasma. 2019, 256, 393–408). Due to the unique structure and distribution of telocytes in the interstitial tissue, these cells create structure-communication networks, regulating the steroidogenic function of Leydig cells under the control of ERR and GPER. Thus, overgrowth of the interstitial tissue in pathological testis may result from disturbances in the number and/or function of telocytes and their regulation via ERR and GPER. In the light of presented data, it seems that apart from the ERs, non-classical estrogen receptors are an important components of estrogen signaling in the male gonad. By controlling Leydig cells and telocytes, ERR and GPER regulate the production of sex hormones, which influences spermatogenesis. These receptors can also serve as additional markers of pathological states of the testicular interstitial tissue, especially those associated with changes in the production and concentration of estrogens. Supported by grants 2015/18/E/NZ4/00519 (SONATA BIS5) and 2016/23/B/NZ4/01788 (OPUS12) from the National Science Centre.

NON-CODING RNA OF SEMINAL PLASMA AS A DIAGNOSTIC AND PROGNOSTIC BIOMARKER OF MALE INFERTILITY

Małgorzata Kotwicka, Weronika Tomaszewska

Department of Cell Biology, Poznan University of Medical Sciences, Poland e-mail: mkotwic@ump.edu.pl

In about 60–75% of men diagnosed for infertility, it is impossible to establish a clear causal factor, which means that subjects are diagnosed with idiopathic infertility. The exploration for new biomarkers shows high specificity and sensitivity towards male infertility and provides additional information on the molecular mechanisms underlying this condition, and is therefore necessary. Since semen is a biological material relatively easy to obtain in a non-invasive way, it naturally becomes the object of this research. Numerous body fluids, such as plasma, serum, saliva, tears, cerebrospinal fluid, bronchoalveolar lavage fluid, milk, and urine contain cell-free, non-coding RNA (ncRNA) molecules. Due to the size of the molecule, three main classes of ncRNAs have been distinguished: short non-coding RNA (sncRNA), long non-coding RNA (lncRNA) and very long non-coding RNA (vlncRNA). NcRNAs are not translated into proteins but can modulate cellular messenger RNAs (mRNAs) and protein levels causing mRNA degradation or translation inhibition. Due to the biogenesis and type of proteins interacting with them, sncRNA have been divided into three main classes: microRNA (miRNA), small interference RNA (siRNA) and PIWI-interacting RNA (piRNA). Extracellular sncRNAs may reflect the molecular changes taking place in the cells. This means that profiling their expression can be a good diagnostic and prognostic tool. However, the great number of ncRNA molecules involved in cellular processes make this analysis difficult and time-consuming. Cell-free miRNA and piRNA were found in the seminal plasma. It should be noted that miRNAs can be secreted by many cells and tissues, and the seminal plasma contains fluids secreted by testis, epididymides, seminal vesicles, bulbourethral glands, or the prostate. Thus, seminal plasma miRNAs may not provide unambiguous information about the type and location of the pathological condition. Nevertheless, a change in miRNA expression patterns (e.g., miR-34b/c, miR 334a/b/c, miR449a, miR-19a, miR-31-5p, miR-539-5p, and miR-941) has been linked to various histopathological images of the testes (e.g. Sertoli cell-only syndrome, arrest of maturation of spermatogenesis cells) or semen with the signs of asthenozoospermia, oligozoospermia, oligoasthenozoospermia and azoospermia (Abu-Halima et al.: Fertil Steril. 2013, 99, 1249–1255; Barcelo et al.: Hum Reprod. 2018, 33, 1087–1098; Hu et al.: Clin Biochem. 2014, 47, 967–72; Tian et al.: Forensic Sci Int Genet. 2018, 33, 161–167). On the other hand, piRNA are upregulated especially in pachytene spermatocytes and round spermatids. They seem to play a preliminary role in spermatogenesis, studies showed that silencing of PIWI protein expression inhibited sperm formation and maturation. piRNAs, includes piR-31068, piR-31925, piR43771, piR-43773 and piR-30198, were present in the seminal plasma and their expression is associated with the reduced biological value of the semen (Hong et al.: Scientific Reports. 2016, 6: 224–229). Recent studies also pointed out that to long non-coding RNA and circulating DNA (cfs-DNA) as a potential markers in the diagnosis of infertility. An analysis of the available literature shows that ncRNAs represent a promising complement to traditional male infertility biomarkers.

THE INFLUENCE OF SRAS-COV2- (COVID-19) VIRUS INFECTION ON MALE REPRODUCTIVE HEALTH

Maciej Kurpisz, Monika Frączek

Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland e-mail: maciej.kurpisz@igcz.poznan.pl

Severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) can be now presented as a common pandemic microbial agent with adverse multi-organ effects directed, among the others, to male reproductive system. Its promiscuity within organism can be linked to numerous receptors mediating its harmful actions to the host. One of them and seemingly a prominent player appears to be, ACE-2, (angiotensin-converting enzyme 2) which is expressed almost on the all morphotic elements of the male gonad. Another enzyme, expressed on adult testis spermatogonia is TMPRSS2 (transmembrane serine protease 2 enzyme) which cleaves capsid S of the virus. The other assisting receptors for Cov-2 are BSG (basigin) inducing metalloproteases of extracellular matrix, expressed on oolemma and trophectoderm membranes which presents opportunity for early embryonal damage, ANPEP (alanyl aminopeptidase N) is overexpressed in Parkinson disease as well through epigenetic mechanisms may induce cancerogenesis in a prostate gland (it is also expressed in endometrium). In turn, receptor DPP4/CD26 (dipeptidyl peptidase-4) has been expressed on epithelial and endothelial cells of vasculature belonging to circulatory system of lung, kidneys, intestine and heart – thus could potentially induce cytokine storm or thrombotic process. Interaction of SARS-Cov-2 with ACE-2 receptor may lead to dysfunction in hypothalamus-pituitary-testis axis. Principally it is due to primary dysfunction of Sertoli cells – secondarily it involves germ cells. SARS-Cov-2 does not primarily trigger autoimmune orchitis but it acts altering endocrine, inflammatory and innate immunity within male gonad – often exaggerated through release of reactive oxygen species (ROS) associated with pro-inflammatory stage. Other, visible disturbing syndromes in male reproductive ability are associated rather with therapeutic process than virus presence per se. Adverse effects may be caused by anti-viral drugs and glucocorticosteroids. They both may exert cytotoxic actions towards actively dividing cells thus impairing spermatogenesis. Also polyclonal antibodies occasionally applied may be harmful due to possible cross reactivity. Additionally, glucocorticosteroids destroy cell junctions in spermatogenetic epithelium affecting blood-testis barrier. Infiltrating leucocytes (due to inflammatory state invoked by SARS-Cov-2) may produce reactive oxygen species and cytokines as well as may augment cytokine secretion and synthesis in male gonad itself giving a ground for initiation of local cytokine storm. Due to all these circumstances the classical sperm parameters (sperm quality) may deteriorate which makes necessity of careful semen oxidative stress monitoring as well as application of antioxidants. Testosterone supplementation should be also considered. Particularly when applying anti-viral drugs a grace period in trying for offspring should be considered for at least few month (epigenetic phenomena or sperm DNA damage should be considered which strictly speaking is a prerequisite to abandon procreative activities during that time. Infertility may be induced primarily due to Sertoli cells dysfunction. Another problem could be asymptomatic infections in children of pre-pubertal or early reproductive age. Long-term carriership of the SARS-Cov-2 in semi-privileged site should be taken into attention.

SPERM DNA FRAGMENTATION – DIAGNOSTICS AND TREATMENT

Sposób przygotowania manuskryptu



Krzysztof Łukaszuk1,2

INVICTA Fertility and Reproductive Center, Gdansk, Warsaw, Wroclaw, Bydgoszcz, Poland; 2Medical University of Gdansk, Faculty of Health Sciences Department of Obstetrics and Gynecological Nursing, Gdansk, Poland e-mail: krzysztof.lukaszuk@invicta.pl

Globally, about 140 million people (13–15% of couples) remain childless after a year of trying. Only a small minority seeks any treatment. The male factor is the least known aspect of the diagnosis of the causes of infertility. So far, there are no clearly defined cut off values for identifying sperm reproductive potential based on standard semen analysis. The value of the World Health Organization (WHO) standards for semen analysis is still being questioned. The most common deviations from the test standard – in terms of sperm count, motility and morphology do not withstand a detailed analysis of their causal relationship with pregnancy. Based on current research, it is even difficult to determine whether standard semen analysis parameters are clearly related to the cause of infertility. Thus the search continues for tests that would facilitate diagnostics and treatment management of an infertile couple. The most convincing is the analysis of sperm DNA fragmentation and other abnormalities related to the genetic material contained in the sperm. Properly chromatin remodeling, from histones to protamines, in the nucleus of mature spermatozoa has a protective function for DNA. Sperm chromatin integrity is essential for normal reproductive function. Its disorders cause: infertility, poorer embryo quality, lower implantation rate and an increased risk of miscarriage or having a sick child. The oocyte can effectively repair sperm chromatin damage to some extent, but the effectiveness of the repair mechanisms in the oocytes declines with age. The mechanisms of sperm DNA damage known so far include: apoptosis or anomalies arising during the spermatogenesis process, DNA strand breaks during chromatin remodeling in the process of spermiogenesis, non-nuclear DNA damage mainly by oxygen free radicals during transport through the seminal tubules and in the epididymis, DNA fragmentation induced by endogenous caspases and endonuclease, damaging effects of radio- and chemotherapy and environmental toxins. The main one appears to be the oxidative stress. It can be related to internal factors (such as protamine deficiency and DNA packaging defects) affecting up to 15% of infertile men. We should also take into account external factors – inflammation of the genital organs, heat, radiation, chemotherapy, smoking. Smoking generates high levels of oxidative stress and reduces the concentration of antioxidants in semen plasma. Unfortunately, sperm DNA fragmentation does not correlate with the basic sperm parameters. Increased sperm DNA damage was observed in approximately 5% of infertile men with normal sperm parameters compared with 25% in the group of patients with abnormal sperm parameters. Increased sperm DNA fragmentation index indicates reduced male fertility regardless of sperm count, motility and morphology. The results of the assessment of the sperm chromatin structure assay (SCSA) allowed for the best assessment of the chances of a successful pregnancy in the case of planning the first pregnancy, also in couples undergoing intrauterine insemination (IUI). That shows why it is so important to use sperm DNA fragmentation analysis in the evaluation of an infertile couple. In order to be able to effectively diagnose the cause of infertility, one must have an appropriate diagnostic tool. We currently have several methods for determining sperm DNA damage. Unfortunately, the increased popularity of such test and their commercialization lead to introduction of unreliable tests to the market, that easy to perform and inexpensive to buy. As a result, a group of clinics charges unaware patients for the unreliable tests the fees comparable to reliable tests for, obtaining a 1000% margin. The tests that have proven reliability and quality include SCSA, TUNEL (TdT-mediated dUTP nick-end labeling) and comet assay. Among the tests not confirmed by independent studies, one should mention the commercial hit – sperm chromatin dispersion (SCD) test– falsely called the sperm DNA fragmentation test. So far, it has not been clearly established where to place the sperm DNA fragmentation in the treatment algorithm – does it belong in the initial diagnosis, extended diagnosis, before in vitro fertilization or after its failure. We currently have a tool for at least partial separation of sperm with high fragmentation of sperm DNA, so the question arises whether we should use it also after sperm separation for in vitro fertilization.

ANDROGENETIC ALOPECIA AND MEN’S HEALTH

Mariola Marchlewicz1, Ewa Duchnik2

Department of Dermatology and Venereology, Pomeranian Medical University in Szczecin, Poland, 2Department of Aesthetic Dermatology, Pomeranian Medical University in Szczecin, Poland e-mail: mariola.marchlewicz@pum.edu.pl

The presence of healthy, properly growing hair is very important for the well-being of men. The processes of hair growth and replacement are regulated, among other factors by androgens, in some places androgens have no effect, in other stimulate or inhibit hair growth. In men, high levels of androgens stimulate the growth of facial, suprapubic and chest hair. However, in the scalp androgens can cause baldness. Hair follicles are androgensensitive due to the expression of Androgen Receptor (AR) in dermal papilla cells (DPCs). The AR expression is significantly higher in DPCs of the chin and areas subject to androgenetic alopecia than in non-balding skin of the occipital region. Androgenetic Alopecia (AGA) affects approximately 50% of adult men under the age of 50 and over 70% over the age of 70. The clinical presentation of AGA is that hair lost is initially in the frontal area and then in the top of the head. The severity of the disease is assessed on the Hamilton – Norwood scale. The primary pathogenetic process in AGA, is mediated by dihydrotestosterone (DHT), hair follicle miniaturization in androgen-dependent areas. Genetic (single nucleotide polymorphism in exon 1 of the AR gene within the long arm of X chromosome), hormonal (testosterone, DHT) and environmental factors are important in the pathogenesis of AGA. It is thought that in AGA there is, within the hair follicle, an altered interaction between DPCs and keratinocytes. In the DPCs the androgen/AR complex influences the synthesis of factors that regulate both the function of these cells autocrinally and the function of follicular epithelial cells paracrinally, e.g. through IGF-1 (insulinlike growth factor 1), within DPCs, in areas of baldness skin, the level of secretion of this cytokine is as much as 6 times lower than in non-baldness skin, what confirms the important role of IGF-1 in inhibiting baldness. Androgens also alter the transcription of genes, in DPCs, regulating the hair growth cycle. They are responsible for reducing the number of cells in the hair follicle, as high levels of DHT induce apoptosis of DPCs and loss of their proliferative properties. As a consequence, there is a shortening of the anagen phase and a reduction in the size of the follicles. The activity of DPCs, keratinocytes and melanocytes changes, which leads to the transformation of the final hair follicles into the follicle in which shorter, thinner and discolored hair are formed (Marchlewicz et al.: Post Androl Online. 2014, 1(2), 14–24). In addition to the negative impact of AGA on quality of life, a growing number of studies have shown its association with increased cardiovascular risk. Men with AGA are more common to have metabolic syndrome, obesity, hypertension, dyslipidemia, insulin resistance and accelerated atherosclerosis. The criteria for diagnosing the metabolic syndrome are met by 20-57% of patients with AGA. The patomechanism of the coexistence of AGA and cardiovascular diseases is complex and includes genetic factors (increased sensitivity to androgens and increased 5α - reductase activity), hormonal (increased insulin, aldosterone, leptin) and inflammatory factors (cytokines, reactive oxygen species). Patients with AGA have an inflammatory reaction around the hair follicles, and serum levels of oxidative stress markers have been found to be increased in patients with early onset AGA. Studies show an association of AGA with increased levels of cholesterol and triglycerides in the blood serum and higher values of systolic and diastolic blood pressure in these men (Wernicka et al.: Przegl Dermatol. 2018, 105, 716–725).

PROGNOSTIC SIGNIFICANCE OF FUNCTIONAL TESTS IN REPRODUCTION

Katarzyna Marchlewska

Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Poland e-mail: katarzyna.marchlewska@umed.lodz.pl

In recent years, a lot of attention has been paid to study the sperm quality. A significant progress has been made in understanding the importance of sperm chromatin integrity as well as the impact of oxidative stress on male fertility. However, the assessment of sperm function seems to be no less important. One of parameters describing sperm function in the basic semen analysis is motility. Often, for prognostic purposes, an index called total motile sperm count (TMSC) is also calculated. The reference values for this parameter are still under discussion. Some authorssuggest the cut-off value above 10, others above 5 or even above 1 million. This index is usually used when qualifying a couple for intrauterine insemination (IUI), but also to predict the chances of natural fertilization. Many studies has shown that the motility does not ensure that the sperm is involved in the reproductive process. Under natural conditions, about 1% of spermatozoa deposited during sexual intercourse enter the uterus. This number is constantly reduced as sperm migrate through subsequent sections of the female reproductive system. That is why the assessment of their migration ability is such an important parameter related to male gametes hyperactivation, chemo- and thermotaxis. The capability of sperm to migration is assessed using a functional test such as the migration test, often called the “swim up” test. The result of the test indicates the number of sperm that migrate in 1 hour from 1 mL of native semen sample to 1.2 mL of human tubal fluid-like medium and is expressed as the motile sperm concentration (million/mL) after the preparation. The test is used to predict the chances of natural fertilization, IUI and classic in vitro fertilization (IVF). It has no cut-off value because during interpretation it should be remembered that result does not take into account the total ejaculate volume so it should be considered individually. In the literature the limit values for number of motile sperms that will be transfer into the uterus during evaluation of fertilization effectiveness using IUI method ranges from 0.8 to 20 million sperm. However, the American Society for Reproductive Medicine (ASRM) published data showing that the effectiveness of insemination increases with the number of sperm transferred to the uterus, but only to 9 million and then remains constant. On the other hand, if qualification for the IVF procedure is considered, the sperm number of 70–100 thousand per each oocyte ensure the optimal conditions. Fertilization is a multistep and complex process and there is a paucity of diagnostic methods that are dedicated to evaluate the function of male gametes at each stage. One of these tests indicates sperm maturity and assesses their ability to bind to hyaluronic acid (hyaluronan binding assay – HBA). Physiologically, hyaluronic acid is found in the corona radiata of the oocyte and is degraded by an enzyme hyaluronidase located on the sperm heads. As recommended by the test manufacturer, a value of ≥80% of the motile sperm responding to the test was considered normal. However, other studies suggest that HBA value of <

60% significantly indicates decreased sperm maturity (Huszar et al., Reprod Biomed, 2007, 14, 650–663), while patients with a binding score ≤65% have a reduced chance of having mature sperm randomly selected for ICSI (Worrilow et al., Hum Reprod, 2013, 28, 306–314). The data of our own studies concerning the frequency of impairment of sperm function in the population of men referred for semen analysis in the Lodz agglomeration will be presented.

FOLLICLE-STIMULATING HORMONE (FSH) REGULATES NOTCH SIGNALING IN RAT TESTIS DURING PUBERTY

Sylwia Marek1, Alicja Kamińska1, Małgorzata Brzoskwinia1, Laura Pardyak2, Anna Hejmej1, Barbara Bilińska1

Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Poland; 2Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, Poland e-mail: s.marek@doctoral.uj.edu.pl

The function of the mammalian male gonad is maintained by the hypothalamic-pituitary axis, whose activation during puberty is associated with pulsatile secretion of gonadotropin-releasing hormone (GnRH) from hypothalamus. Follicle-stimulating hormone (FSH) released in response to this signal, is involved (along with androgens) in the control of spermatogenesis. Studies on rodent models have shown that FSH, by regulating Sertoli cell function, reduces apoptosis of germ cells during the first wave of spermatogenesis, and influences the proliferation and differentiation of spermatogonia (O’Shaughnessy: Semin Cell Dev Biol. 2014, 29, 55–65). Sertoli cells support spermatogenesis through mutual interactions and communication with germ cells. One type of direct intercellular interactions in the seminiferous epithelium is communication via the Notch signaling pathway (Murta et al.: PLoS One. 2013, 8, e72767). Although in recent years the role of Notch signaling in spermatogenesis has been the subject of in-depth research, the regulation of this pathway in the seminiferous epithelium is still poorly understood. Our recent studies using pubertal rats exposed to GnRH antagonist followed by FSH supplementation, have shown that the function of Notch pathway in the testes is modulated by FSH. The expression of Notch1, Notch2, and Notch3 receptors, as well as the levels of their active forms, were altered in the seminiferous epithelium, both in Sertoli cells and in germ cells, of FSHtreated rats. The expression of Notch pathway effector genes Hes1 and Hey1 was also investigated, and the localization of HES1 and HEY1 proteins in the seminiferous tubule was confirmed by immunohistochemical analysis. Decreased Hes1 expression was found after FSH signaling activation, which may indicate a reduction in the Notch pathway activity in the seminiferous epithelium. The results presented herein demonstrate that in seminiferous epithelium FSH regulates direct intercellular communication based on the Notch pathway. The interplay between FSH and Notch pathways may be a molecular mechanism responsible for the effect of FSH on the ability of Sertoli cells to support germ cells. These observations may therefore contribute to elucidating the basis of impaired germ cell development associated with disturbed FSH action. Supported by a grant OPUS13 2017/25/B/NZ4/01037 (National Science Centre, Poland).

THERE WAS SEX AND TESTOSTERONE REMAINED

Marek Mędraś

Department of Sports Medicine and Dietetics, University School of Physical Education, Wrocław, Poland, Polish Anti-Doping Laboratory, POLADA e-mail: m.medras@gmail.com

During the lecture, the information on sex differentiation and then into the problem of gender verification in competitive sport from a historical perspective will be presented. Moreover, the issue of testosterone levels in healthy women and with hyperandrogenism will be analyzes. These data will be confronted with recommendations of sports organizations regarding acceptable testosterone levels in female athletes. Finally, the cases of sex development disorders (46,XY DSD) encountered in competitive sports (androgen insensitivity syndrome, 5α-reductase deficiency, 17-β hydroxysteroid dehydrogenase deficiency, congenital adrenal hyperplasia, Leydig cell hypoplasia) will be presented.

THE IMPORTANCE OF HORMONAL BALANCE IN THE REPRODUCTIVE TISSUES OF THE TURKEY WITH YELLOW SEMEN SYNDROME

Laura Pardyak1, Alicja Kaminska2, Malgorzata Brzoskwinia2, Anna Hejmej2, Malgorzata Kotula-Balak3, Jan Jankowski4, Mariola Slowinska5, Andrzej Ciereszko4, Barbara Bilinska2

1Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, Poland, 2Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Poland, 3University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Poland, 4Department of Poultry Science, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Poland, 5Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland e-mail: laura.pardyak@urk.edu.pl

It is well established that some male turkeys produce yellow semen instead of the normal white semen (WNS). Yellow semen syndrome (YSS) in domestic turkey (Meleagris gallopavo) was detected over thirty years ago by identifying seminal plasma having both a yellow color and a high protein concentration (Thurston et al.: Poult Sci. 1982, 61, 1905–1911). Moreover, the YSS has been characterized as containing abnormal sperm, spermatids and causes reduced fertilizing capacity and hatchability compared to the WNS produced by most turkeys. More recently, spermatozoa motility characteristics has been disturbed in YSS turkeys as demonstrated by a computerassisted sperm analysis (Slowinska et al.: Poult Sci, 2011, 90, 181–190). Although semen quality parameters have been presented in detail, the cause of YSS still remains unclear. It is noteworthy that male turkeys producing yellow semen are of special interest as an animal model for the investigation of reproductive changes associated with unexplained low sperm quality. In many mammalian species the balance between testosterone and estradiol is crucial for spermatogenesis, normal sperm maturation within the epididymis, and proper functioning of the ductus deferens. Compared with extensive studies on mammals, very little is known about the importance of testosterone metabolism and the androgen:estrogen ratio within the reproductive tissues of domestic birds. Thus, the aim of the present study was placed on understanding a connection between the hormone levels and the lower quality of sperm occurring in YSS male. The study was performed on testicular, epididymal, ductal and semen samples collected from adult YSS and WNS turkeys using several techniques such as immunohistochemistry, Western blotting, qRT-PCR (quantitative real-time polymerase chain reaction), and electron microscopy (Pardyak et al.: Br Poult Sci, 2018, 59, 591–603; Pardyak et al.: Poult Sci, 2020, 99, 555–566). The results revealed a non-homogeneous distribution of aromatase and its markedly enhanced expression levels in all reproductive tissues and sperm of YSS males compared to WNS ones, what may reflect a higher endogenous synthesis of estrogens in the males with abnormal yellow semen. This suggestion was confirmed by increased estradiol concentration measured in YSS testes compared to the value of WNS testis samples. Additionally, a positive correlation between increased estradiol concentration and elevated expression of estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) in YSS testes and epididymis was observed. The in vivo results were in line with ex vivo experiments where YSS and WNS testis explants were incubated with estradiol. Moreover, transmission electron microscopy observations revealed that YSS spermatozoa have a reduced fibrous sheath thickness compared with WNS spermatozoa, which may suggest impaired sperm motility in turkeys with the yellow semen syndrome. Taken together, our data suggest that the androgen:estrogen ratio provides a mechanistic basis for amplification of differences between turkeys with white and yellow semen. It may indicate a potential relationship between hormonal imbalance and lower semen quality in turkeys with YSS as indicated by structural disorders of the proximal part of the YSS spermatozoa tail. Supported by a grant 2017/25/N/NZ9/00585 (PRELUDIUM 13) from the National Science Centre to L.P.

LATE ONSET HYPOGONADISM – CURRENT OPINIONS ON DIAGNOSIS AND TREATMENT

Michał Rabijewski

Department of Reproductive Health, Centre for Postgraduate Medical Education, Warsaw, Poland e-mail: mirab@cmkp.edu.pl

Late-onset hypogonadism (LOH) is a syndrome characterized by clinical and biochemical evidence of low testosterone (T) levels with advancing age, and is promoted by senescence. LOH affects the hypothalamicpituitary-testis axis. Additionally, metabolic disorders can have a greater impact on causing hypogonadism than tissue senescence, which is reflected in decreased male quality of life, mood, sexual performance as well as increased morbidity and mortality. On the other hand, T deficiency is a risk factor for the metabolic disorders, diabetes, and atherosclerosis (vicious cycle). Testosterone replacement therapy (TRT) is widely used to restore T levels and can improve symptoms and signs of T deficiency including decreased libido, erectile dysfunction, depressed mood, loss of muscle and bone mass, by increasing serum T levels to physiologic range. In recent years, several guidelines for TRT have been develop, but despite similar principles, there are still important differences both for the diagnostic workup and follow-up. Discrepancies were reported both for total and free T levels for diagnosis and for total testosterone for monitoring. Numerous preparations of T have been developed to improve pharmacokinetics and patient compliance. It is important to use the appropriate and safe preparation for each patient. Longterm TRT in patients with LOH is safe in terms of its impact on heart disease and the risk of prostate diseases. Although, the effects of TRT on the cardiovascular system are complex and rather positive, but several clinical observations reveal neutral or occasionally detrimental effects, mostly due to confounding factors. It appears, that TRT is safe once other comorbidities are addressed.

ANDROGEN RECEPTOR AND ANDROGENS INSENSITIVITY SYNDROMES

Michał Rabijewski

Department of Reproductive Health, Centre for Postgraduate Medical Education, Warsaw, Poland e-mail: mirab@cmkp.edu.pl

The androgen receptor (AR) play a significant role in male sexual differentiation and the development and function of male reproductive and organs. Because of AR’s widely varied and important roles, its abnormalities have been identified in various diseases. Androgen insensitivity syndrome (AIS) is a genetic condition where affected people have male chromosomes and male gonads with complete or partial feminization of the external genitals. AIS results from mutations in the androgen receptor gene, located on the long arm of the X chromosome (Xq11-q12) which iss associated with functioning Y sex chromosome and abnormality on X sex chromosome. AIS is devided into CAIS (complete androgen insensitivity syndrome) with external female genitalia and lacking female internal organs, PAIS (partial androgen insensitivity syndrome) with external genitalia appearance on a spectrum (male to female), and MAIS (mild androgen insensitivity syndrome) with impaired sperm development and/or impaired masculinization. MAIS is the most common cause of mild hypogonadism at elevated testosterone (T) levels. in some men we observe the AR gene polymorphism leading to a slight but clinically significant decrease in T action, which causes clinical symptoms of hypogonadism at normal T levels. The negative influence of AR gene polymorphism on bone mineral density, lipids and mood was demonstrated but positive influence on prostate volume. AR gene polymorphism should be taken into account in patients with symptoms of hypogonadism and the normal T levels, and may also modulate the risk of side effects during testosterone replacement therapy.

INTESTIN MICROFLORA, DISEASES OF THE INTESTINES AND CANCER AND BENIGN HYPERPLASIA OF THE PROSTATE GLAND

Weronika Ratajczak1,2, Olimpia Sipak3, Maria Laszczyńska2

1Department of General Pharmacology and Pharmacoeconomics, 2Department of Histology and Development Biology, 3Department of Obstetrics and Pathology of Pregnancy, Pomeranian Medical University in Szczecin, Poland e-mail: veronica.ratajczak@gmail.com

Benign prostatic hyperplasia (BPH) is one of the most commonly diagnosed urological diseases in men over 50 years of age. BPH is characterized by prostatic stromal cell proliferation, leading to prostatic bladder obstruction (BOO) and lower urinary tract symptoms (LUTS), which together reduce quality of life (QoL). The development of BPH is very often associated with the existence of comorbidities, such as diabetes, cardiovascular diseases, and even neurological diseases (Cho et al.: Curr Bladder Dysfunct Rep. 2020, 15, 60–65). Many studies also indicate a relationship between the metabolic syndrome (MetS) and the risk of LUTS and BPH (Zhao et al.: Urol J. 2016, 13, 2717–2726). The factor that contributes to the initiation of pathological changes in the prostate, and consequently its benign hyperplasia, is chronic inflammation resulting, among others, from metabolic disorders (Gacci et al.: BMC Urol. 2017, 17:22). Moreover, an additional factor that influences the occurrence of inflammation in the body is disturbances in the intestinal microflora. The intestinal microflora is one of the elements of the bacterial ecosystem in mammals. The microorganisms that inhabit the gut are one of the key elements involved in modulating the immune response from the moment of birth. More and more research is currently 37 SYMPOSIUM OF SCIENTIFIC TRANINING OF THE POLISH SOCIETY OF ANDROLOGY – 2 being carried out on the effect of bacterial metabolites, including short-chain fatty acids (SCFA), on homeostasis, not only in the intestinal microenvironment, but also in cells and tissues of other organs. SCFAs are a group of compounds made up of six carbon atoms (C1 - C6), with the majority of acids being: butyric acid (C4), propionic acid (C3), and acetic acid (C2) (Tramontano et al.: Nat Microbiol. 2018, 3, 514–522). So far, disturbances in the intestinal microflora and its impact on inflammation and prostate diseases have not yet been thoroughly analyzed. The influence of SCFAs on the development of BPH has also not been studied. Only a few publications on the impact of the intestinal microflora on the prostate are available in scientific data bases. They mainly concern the influence of intestinal bacteria on the synthesis of metabolites and androgens, which may be associated with the development of prostate cancer in humans (Liss et al.: Eur Urol. 2018, 74, 575–582). A study conducted in mice, on the other hand, revealed that pathogenic intestinal bacteria can promote the development of prostate cancer by activating systemic immune cells (Poutahidis et al.: PLoS One. 2013; 8:e73933). There are also reports of the impact of inflammatory bowel disease (IBD) on the risk of prostate cancer (Burns et al.: Eur Urol. 2019, 75, 846–852). So far, differences in the composition of the intestinal microflora have only been confirmed in a pilot study, in which the composition of the intestinal microflora was analyzed in patients with prostate cancer, and with BPH (Golombos et al.: Urology. 2018, 111, 122–128). The results of the research by Liss et al. indicate that bacteria predominating in men diagnosed with prostate cancer (PCa) were Bacteroides and Streptococcus spp. The exact mechanism by which the intestinal microflora affects the prostate gland has not so far been fully elucidated. It seems very likely, however, that disturbed intestinal microflora does not directly affect the prostate gland, but contributes to the development of chronic systemic inflammation. Inflammatory Cells and inflammatory factors (including cytokines) from the intestinal environment, along with the circulation, can get into the gland and there cause ‘local’ inflammation and stimulate the growth factors of the prostate stroma, which in turn may lead to prostatic diseases. The own study compared the SCFAs profile between healthy patients and patients diagnosed with BPH, with regard to MetS as a factor predisposing to the development of prostate hyperplasia. The conducted study is the first to show changes in the tested SCFA levels between these two groups. Nevertheless, further research is needed (including testing in animal models) to determine whether there is a ‘microbiota-gut-prostate axis’ and whether the intestinal microflora and its metabolites contribute to the development of BPH. Study supported by the Pomeranian Medical University (no. WNoZ-322-03/S/16/2020).

NGS (NEXT GENERATION SEQUENCING) ANALYSIS IN FERTILITY DIAGNOSTICS

Marzena Skrzypczak-Zielinska, Monika Frączek

Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland e-mail: mskrzypczakzielinska@gmail.com; framon@man.poznan.pl

Genetic analyzes play an important role in determining the causes of reproductive failure in couples. Genetic factors are considered responsible in at least 10–15% of cases of male infertility (Ambulkar et al. J. Clin. Diagn. Res. 2014, 8, 88–91). Karyotype analysis has become a standard. Chromosome aberrations occur in 7% of infertile men, which is 30 times more frequent than in the general population. Another cause of male infertility are microdeletions of the Y chromosome or aberrations and mutations of genes responsible for male sexual development, e.g. located in the Yp11.2 region. On the other hand, patients with bilateral or unilateral absence or obstruction of the vas deferens should be ordered to check for the presence of mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene (Łukaszuk et al. Gin Perinat Prakt. 2018, 3, 112–140; Kaminski et al. Int J Mol Sci. 2020, 21, 5274). However, the possibilities of genetics do not end there. Modern technologies open up new areas of research in the diagnosis of infertility. Next generation sequencing (NGS) is such a technology, which is used, e.g., in genetic preimplantation diagnostics (PDG) as part of the in vitro procedure in order to reduce the risk of serious chromosomal defects in the embryo (before a woman becomes pregnant), and in the detection of monogenic diseases and disorders related to the occurrence of translocation in the offspring. Performing the NGS analysis is especially recommended for individuals with a positive genetic history, couples who have experienced recurrent spontaneous abortions or whose offspring have been diagnosed with genetic disorders, as well as women deciding to become pregnant after 35 years of age (Łukaszuk et al.: Gin Perinat Prakt. 2018, 3, 112–140). With PGD diagnosis significantly increases the percentage of embryo implantation and birth of healthy children. Another crucial aspect of the use of NGS in the diagnosis of infertility is the characteristics of the microbiome of the reproductive system. Because the human body contains more microbes than human cells, the microbiome (being termed the ‘second human genome’) has a huge potential to influence human physiology (Tsonis et al.: Ann Transl Med. 2020, 8, 1707). To date, the use of NGS supports the explanation of the functional, quantitative and mechanistic aspects of the complex microorganism-host interactions, particularly in relation to the gut microbiome. Meanwhile, the sperm microbiome is an area of growing scientific interest due to important implications for male reproductive health, the health of couples, and even the health of the offspring by transmitting microorganisms to the partner and offspring (Baud et al. Front Microbiol. 2019, 10, 234; Chen et al. Exp Ther Med. 2018, 15, 2884–2890). An indication of a particular species of bacteria having an impact on sperm function may facilitate the development of new therapies (eg. probiotics). Currently, however, the data are inconclusive and more research is needed (Farahanii et al.: Andrology. 2021, 9, 115–144). Nevertheless, it should be expected that NGS analyzes are the future of broadly understood molecular and genetic diagnostics also in infertility research.

EAA GUIDELINES ON KLINEFELTER SYNDROME

Jolanta Słowikowska-Hilczer

Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, Poland e-mail: jolanta.slowikowska-hilczer@umed.lodz.pl

Klinefelter’s syndrome is the most common (0.1–0.2% of male newborns) genetically determined cause of infertility and hypergonadotrophic hypogonadism in men. It develops as a result of numerical aberration of X chromosomes (most often 47,XXY). During childhood and early puberty, the hypothalamic-pituitary-testis axis is usually normal. The clinical picture of hypergonadotropic hypogonadism develops from mid-puberty (stage G III according to Tanner’s classification) due to progressive degeneration of the structure and impairment of testicular function (Wikström and Dunkel: Horm Res. 2008, 69, 317–326). The phenotype is varied, ranging from nearly normal to significantly abnormal. The phenotype in newborns with Klinefelter syndrome is usually normal male. Often the only clinical feature is small testes, which are most often not identified until puberty. Patients with this syndrome are most often infertile, but in about half of the cases it is possible to find spermatozoa in the testicles (Rohayem et al.: Andrology. 2015, 3, 868–875), and they also have a higher risk of developing, e.g. for breast cancer, metabolic syndrome, cardiovascular diseases, osteopenia / osteoporosis, autoimmune diseases. Additionally, there are various levels of cognitive, social and behavioral disorders as well as learning difficulties. Early diagnosis for this syndrome is recommended in order to implement early therapeutic and preventive treatment against comorbidities (Zitzmann et al.: Andrology. 2021, 9, 145–167).

ANTI-MÜLERIAN HORMON AND INHIBIN B – THEIR ROLE IN ANDROLOGICAL DIAGNOSTIC

Renata Walczak-Jędrzejowska

Department in Andrology and Reproductive Endocrinology, Medical University of Lodz e-mail: renata.walczak-jedrzejowska@umed.lodz.pl

Anti-Müllerian hormone (AMH), also called Müllerian inhibiting factor (MIF), and Inhibin B (INHB) are glycoproteins belonging to the transforming growth factor β (TGFβ, transforming growth factor β). The main physiological role of AMH is the regression of the Müllerian ducts in the fetal period, while INHB regulates FSH secretion from the pituitary by negative feedback. In the male, they are produced by Sertoli cells in seminiferous tubules starting from the fetal period and are considered to be markers of their function (Iliadou et al.: Hormones. 2015, 14(4), 504–514). The dynamics of secretion of both hormones changes throughout the life span of men, which determines the possibility of using their concentrations measurement in andrological diagnostic. Measurements of AMH and INHB concentrations in blood serum is used in pediatric patients. In the pre-pubertal period, the concentrations of both hormones remain at relatively constant level. During socalled “minipuberty”, i.e. the temporary activation of the hypothalamic-pituitary-gonadal axis, the assessment of their concentration is an additional marker of the proper development of the testicles and function of Sertoli cells. In turn, after its completion, i.e. during so-called “hormonal silence” period, AMH and INHB become the only hormonal markers of testicular activity. Currently, both hormones are used, for example, in the differential diagnosis of testicular failure, congenital hypogonadotropic hypogonadism or sexual differentiation disorders (Rey: Adv Lab Med. 2020, doi.org/10.1515/ almed-2020-0024). Recently, the possibility of using the assessment of AMH and INHB levels in boys in the prepuberty and pubertal period in order to early diagnose isolated Sertoli cell dysfunction is suggested (LaVignera et al.: J Clin Med. 2019, 8(5), 636–342).. With the beginning of puberty and the activation the hypothalamic-pituitary-testicular axis, changes in the secretion of AMH and INHB occur. The AMH concentration decreases to the values corresponding to the concentration values in pre-pubertal girls, while INHB concentration increases to the values observed in adult men. Measurement of the concentration of INHB along with the measurement of the concentration of follicle stimulating hormone (FSH) in adult men has been used for years as a marker of the proper function of Sertoli cells, and thus testicular spermatogenic activity. However, despite many attempts, so far it has not been possible to established the cut-off value clearly indicating an impartment in spermatogenesis or reduced sperm quality. Recent studies also question previous reports showing a higher predictive value of INHB concentration compared to other sex hormones in predicting the chance of obtaining sperm from testicular biopsy in men with obstructive azoospermia (NOA) (Arshad et al.: Int Urol Nephrol. 2020, 52(11), 2015–2038). Although attempts to use serum AMH measurements in adult men as an additional marker of Sertoli cell function and spermatogenesis did not bring expected results, it was possibly to demonstrate a relationship between semen quality and AMH concentrations in seminal plasma (Vitku et al.: Basic Clinl Androl, 2017, 27, 19). When it comes to the use of AMH concentration as a predictor of sperm biopsy in men with NOA, the results are inconclusive (Alfano i et al.: Sci Rep. 2017, 7(1), 17638; La Marca et al.: Hum Reprod Update. 2010, 16(2), 113–130).

OAT – NEW RECOMMENDATIONS BY THE EUROPEAN ACADEMY OF ANDROLOGY

Artur Wdowiak

Diagnostic Techniques Unit, Medical University of Lublin, Lublin, Polnad e-mail: wdowiakartur@gmail.com

Oligoasthenoteratozoospermia (OAT) is a decrease in the amount, progressive motility, and percentage of morphologically normal spermatozoa, diagnosed based on two semen analyses. The etiology of OAT varies and is most often multifactorial. Abnormalities of semen parameters in OAT are most frequently explained by the concept of oxidative stress. Recommendations of the European Academy of Andrology suggest, after making the diagnosis of OAT, conducting a thorough medical history, physical examination, as well as USG and hormonal diagnostics (Colpi et al.: Andrology. 2018, 6, 513–524). Appropriately performed diagnostics allows the selection of a proper treatment method. Gonadotropins, antiestrogens, aromatase inhibitors, and antioxidants are used in the therapy of OAT (Majzoub et al.: Arab Jour Urol. 2018, 2; 16(1), 113–124). The treatment undertaken should be accompanied by the potential change of life style. Management of a patient with OAT should also include the reproductive capacity of the female partner, which is determined primarily by women age. The lack of improvement of semen parameters, or no pregnancy after treatment and low reproductive potential of the partner are an indication for applying the assisted reproductive technology as soon as possible. At present, the most effective treatment method is intracytoplasmic sperm injection (ICSI) (Janicka et al.: Gin Pol. 2021, 92(1), 7–15).

MICROSCOPIC TESTICULAR SPERM EXTRACTION

Jan Karol Wolski1,2

1Fertility Clinic “nOvum”, Warsaw, Poland, 2Department of Urological Cancer, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland e-mail: jkwolski@op.p

In 1998 Peter Schlegel described for the first time the use of an operating microscope testicular biopsy in a man with azoospermia (Schlegel: Hum Reprod. 1999;14(1), 131–135). The microscopic testicular sperm extraction (m-TESE), microsurgical method allows to find groups of promising tubules within damaged gonads where spermatogenesis may occur, giving an advantage over classical surgical (open) and percutaneous needle biopsy. According to guidelines from the European Association of Urology Guidelines on Sexual and Reproductive Health (EAU) 2020, m-TESE is particularly useful for non-obstructive azoospermia. An magnification of 20–25× results in at least 1.5-fold greater effectiveness in sperm obtains compared to a classic biopsy. The Sperm Retrieval Rate (SSR) is 52% (Bernie et al.: Fertil Steril, , 2015, 104, 1099–1103). Between 06.10.2012 and 05.03.2021, 396 m-TESE biopsies were performed in the non-homogeneous group of men with azoospermia: an average age of 34 years (range: 17–44 years). M-TESE was the first procedure performed in 55/396 (14%) patients. The Klinefelter Group consisted of 47/396 (12%). Most patients recived stimulation of spermatogenesis before surgery: androgen + antiestrogen (Adamopoulos et al.: Fertil Steril. 1997. 67(4), 756–762), or gonadotropin (LH, FSH) or clomiphene. In addition, antioxidants were included. The procedures were carried out under general anesthesia, as the one–day-surgery. Type of microscope: Seiler Evolution XR6 (94), Carl Zeiss S7 (6), Leica M860 2 × 2 (296) and the magnification 25× were used. Access to the testes – cutting in the middle line through the scrotum seam (lac. raphe scrotum). After the procedure, prophylactic antibiotic therapy (cephalosporin) was administered for 5 days. SSR achieved in the years 2012–2018 – 30.5%; 2019 – 36,7% and in the year 2020 – 42%. In the Klinefleter syndrome group sperm was found in 8/47 subjects (17%). Complications: 3/396 (0.76%) scrotum hematomas requiring surgical intervention, 5/396 (1,26%) massive scrotum and penile ecchymosis, 3/396 (0.76%) secondary wound healing. Until January 1, 2021, m-TESE sperm were used in 132 IVF/ICSI procedures, 19 pregnancies were obtained after fresh mTESE and 23 from frozen sperm (42/132 – 31.8%). 27 children were born (including one twin pregnancy). Embryo transfer (ET) miscarriages: 5 after fresh ET and 6 after frozen ET. Four pregnancies still developing. M-TESE, testicular biopsy using the operating microscope, increases the chance to find sperm in men with nonobstructive azoospermia, finally allows incorporated them into the reproduction protocol IVF/ICSI.

SEX LIFE OF HYPOGONADAL MEN

Kornelia Zaręba

Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, Warsaw, Poland e-mail: konelia3@poczta.onet.pl

The appropriate level of testosterone in adulthood influences the majority of the sex life aspects in men: dreaming and fantasizing about sex, sex drive, arousal, spontaneous erections, erections in the mechanism of sensory stimulation and the appropriate response to drugs used in sexology. Psychogenic erections are also partially due to the level of testosterone. Hypogonadism occurs considerably more commonly due to the mechanism of body aging (LOH, late-onset hypogonadism) apart from the classic forms which result from the abnormal function of the testes, hypothalamus and the pituitary gland. It requires treatment not only because of the deteriorating quality of life, but also due to high mortality (almost 5.5-fold increase). In the long-term perspective it leads to osteoporosis, metabolic syndrome, insulin resistance, cognitive disorders, infertility and anemia. Sexologists also meet patients with hypogonadism resulting from body weight increase and prolonged exposure to stress. The most common symptoms reported by men with hypoandrogenism include: decreased libido (91%), energy loss (89%), erectile dysfunction (79%), somnolence in the afternoon (77%), and reduced physical endurance (66%). The treatment of sexual disorders in hypogonadal men is a complex and individually-tailored task. It involves the introduction of lifestyle changes (reduction in alcohol consumption, smoking cessation, diet and physical activity influencing the vascular status), psychotherapy and pharmacological treatment. The first-line intervention should involve the correction of potentially-reversible causes. Pharmacological treatment consists both in the substitution of testosterone and stimulating the male gonad to produce testosterone, which is preferred by men trying to get offspring. Normal testosterone concentrations are achieved after 2–3 weeks of therapy. Its effect on the sexual function depends on the dose and differs in various individuals from 2 to 4.5 ng/mL. Inhibitors of phosphodiesterase type-5 (iPDE5) remain first-line treatment in patients reporting erectile dysfunction. Second-line treatment involves the local administration of prostaglandin E1 (PGE1) (alprostadil). Numerous studies confirmed the effectiveness of iPDE5 combined with testosterone in patients in whom monotherapy was unsuccessful (with the minimal treatment duration of 3 months). Concomitant diseases are another important aspect, especially in men with the diagnosis of LOH, as they may be a contraindication for the use of testosterone and require alternative therapeutic solutions. Sexual disorders may frequently result from depressive disorders and depression which may also be due to hypogonadism. The treatment should then include antidepressive drugs with a negligible effect on libido, e.g. bupropion which influences the dopaminergic receptor and is used at a dose of 150 mg/day. In case of a marked anxiety component it is recommended to use medications such as agomelatine, bupropion and moclobemide. In many cases it is necessary to include psychotherapy, predominantly cognitive behavioral therapy.

REKLAMA



andrologia
andrologia


Oddajemy do rąk czytelników starannie przygotowany podręcznik „Andrologii” przygotowany przez najlepszych specjalistów w kraju. To pierwsze tak obszerne opracowanie na temat zdrowia mężczyzn na rynku wydawniczym. Obszerne, całościowe ujęcie zagadnień andrologicznych przygotowane przez andrologów, endokrynologów, urologów, seksuologów, chirurgów, pediatrów, genetyków, biologów, diagnostów, a także psychologów i prawników. Andrologia jest dziedziną medycyny, która zajmuje się męskim układem płciowym i zdrowiem mężczyzn w zakresie prawidłowego rozwoju płciowego, płodności, sprawności seksualnej i starzenia się, z zachowaniem dobrej jakości życia. Jako dziedzina medycyny andrologia wyodrębniła się na pograniczu endokrynologii, urologii, seksuologii, medycyny rozrodu i pediatrii. Obejmuje takie problemy zdrowotne mężczyzn jak: niepłodność, niedobór androgenów, zaburzenia seksualne i zaburzenia rozwoju płciowego. W ostatnich latach obserwuje się niezwykle dynamiczny rozwój andrologii spowodowany zastosowaniem nowych metod badawczych z dziedziny biochemii, biologii molekularnej i genetyki oraz powrót do bardziej intensywnych badań nad zaburzeniami męskiego układu płciowego. Powstają coraz doskonalsze metody diagnostyki laboratoryjnej i obrazowej. Pojawiają się nowe preparaty stosowane w terapii zaburzeń hormonalnych, seksualnych i niepłodności. Opracowywane są rekomendacje dotyczące postępowania w zaburzeniach andrologicznych oparte na coraz silniejszych dowodach naukowych. Praktyczna księga postępowania diagnostycznego i terapeutycznego w zaburzeniach andrologicznych.

Ewa Rajpert-De Meyts, MD, PhD, DMSc

Konsultant naukowy, Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Sekretarz Europejskiej Akademii Andrologii (EAA)

Znakomity podręcznik andrologii obejmujący wszystkie aspekty tej multidyscyplinarnej dziedziny, która zajmuje się problemami zdrowia specyficznymi dla mężczyzn, więc wymaga znajomości wielu aspektów endokrynologii, urologii, medycyny rozrodu, seksuologii, diagnostyki laboratoryjnej, genetyki, onkologii i chorób wewnętrznych, szczególnie kardiologii. Gorąco polecam książkę specjalistom różnych dyscyplin. Uważam również, że powinna być obowiązkową lekturą dla każdego praktykującego androloga, a także dla specjalistów w leczeniu niepłodności, włączając ginekologów. Książka będzie niezwykle pomocna dla młodych lekarzy, którzy planują specjalistyczne egzaminy z andrologii. Wielką zaletą książki jest kompleksowe podejście do tematu, ze szczegółowym omówieniem najnowszej wiedzy dotyczącej męskiej fizjologii i patologii od wczesnego okresu rozwojowego aż do wieku starczego, co uczyni ją bardzo przydatną również dla pediatrów, urologów dziecięcych i lekarzy ogólnych. Nie pominięto również ważnych aspektów psychologicznych, etycznych i prawnych. Redaktor naukowy i autorzy są ekspertami na najwyższym poziomie w skali międzynarodowej i ich zalecenia są oparte na najnowszych doniesieniach ze światowego piśmiennictwa i przyjętych dowodach naukowych, z komentarzami i doświadczeniami dostosowanymi do specyfiki sytuacji w kraju. Książka jest dowodem postępów polskiej andrologii i na pewno przyczyni się do dalszego rozwoju tej dynamicznej dyscypliny. Książka, którą przeczytałam z wielkim zainteresowaniem, jest jednym z najlepszych podręczników andrologii, jakie kiedykolwiek widziałam – i to w skali międzynarodowej. Gratulacje za wybór tematów i autorów należą się redaktorowi naukowemu, prof. Jolancie Słowikowskiej-Hilczer, długoletniej przewodniczącej Polskiego Towarzystwa Andrologicznego. Imponująca jest wszechstronność tematyki pasująca do nowoczesnego holistycznego podejścia do pacjenta.

prof. Aleksander Giwercman

Uniwersytet w Lund, Malmö, Szwecja

Trzymanie w ręku nowego podręcznika z andrologii klinicznej zdecydowanie nie jest codziennym wydarzeniem! Jest to doskonała publikacja nie tylko pod względem treści, lecz także pedagogicznego sposobu przedstawienia informacji, zarówno w odniesieniu do zestawu tematów, struktury poszczególnych rozdziałów, jak i szerokiego wyboru wysokiej jakości ilustracji i tabel. Wszystkie rozdziały są napisane przez ekspertów w swojej dziedzinie. Książka jest nie tylko doskonałym narzędziem dla tych lekarzy, którzy chcą zostać andrologami klinicznymi, lecz także klinicystów reprezentujących inne specjalności oraz badaczy zainteresowanych zagadnieniami związanymi z chorobami męskiego układu płciowego. Jestem przekonany, że niezależnie od tego, do której z tych kategorii należysz, ta książka udzieli odpowiedzi na wiele pytań z dziedziny andrologii. Gratuluję moim polskim Koleżankom i Kolegom opracowania, które z pewnością będzie dobrym dodatkiem do ich innych działań mających na celu rozwój polskiej andrologii. Miejmy nadzieję, że w przyszłości pojawią się kolejne wydania książki, która może stać się inspiracją dla andrologów z innych krajów do stworzenia podobnych publikacji w różnych językach.