Ponad 7000 publikacji medycznych!
Statystyki za 2021 rok:
odsłony: 8 805 378
Artykuły w Czytelni Medycznej o SARS-CoV-2/Covid-19

Poniżej zamieściliśmy fragment artykułu. Informacja nt. dostępu do pełnej treści artykułu
© Borgis - Postępy Nauk Medycznych 12/2016, s. 903-909
*Katarzyna Jankowska, Magdalena Kochman
Endocrine causes of infertility
Endokrynologiczne przyczyny niepłodności
Department of Endocrinology, Centre of Postgraduate Medical Education, Bielański Hospital, Warsaw
Head of Department: Professor Wojciech Zgliczyński, MD, PhD
Streszczenie
Zaburzenia płodności stanowią narastający problem zdrowotny. Powinny być one rozważane zawsze w odniesieniu do obojga partnerów, gdyż dotyczą one w podobnym stopniu kobiet i mężczyzn. Prawidłowy rozwój i funkcjonowanie narządów rodnych kobiety i mężczyzny, procesy tworzenia i rozwoju komórek jajowych i plemników oraz poczęcia dziecka i utrzymania ciąży zależą od współdziałania szeregu czynników, w tym hormonów. Działanie jajników i jąder jest ściśle związane z czynnością innych gruczołów wydzielania wewnętrznego. Dlatego też niepłodność może być skutkiem nieprawidłowego funkcjonowania przysadki, tarczycy czy nadnerczy. W artykule omówiono najczęstsze przyczyny pierwotnej niedoczynności gonad oraz zaburzenia czynności podwzgórza i przysadki prowadzące do hipogonadyzmu, ze szczególnym uwzględnieniem niedoboru gonadotropin i hiperprolaktynemii. Szczególną uwagę zwrócono na endokrynne skutki zaburzeń odżywiania prowadzące do upośledzenia płodności. Omówiono także wpływ na płodność chorób tarczycy i nadnerczy oraz zaburzeń hormonalnych związanych z zespołem policystycznych jajników. Zwrócono uwagę na diagnostykę poszczególnych jednostek chorobowych i ich leczenie w kontekście optymalizacji zdolności prokreacyjnych pacjenta.
Summary
Impairment of fertility is a growing health problem. It should be always considered in relation to both partners, as it affects to a similar extent women and men. Proper development and function of male and female reproductive organs, the processes of egg cell and sperma formation and development as well as of conception and pregnancy maintenance depend on interaction of a number of factors, including hormones. Functioning of ovaries and testes is closely related to the activity of other endocrine glands. Therefore, infertility can be the result of a malfunction of the pituitary, thyroid or adrenals. The article discusses the most common causes of primary hypogonadism as well as hypothalamic and pituitary dysfunctions leading to hypogonadism, in particular gonadotropin deficiency and hyperprolactinemia. Special attention was paid to the endocrine effects of eating disorders, which result in impaired fertility. We also discussed the impact on fertility of the thyroid and adrenal diseases as well as hormonal disorders associated with polycystic ovary syndrome. Attention was paid to diagnosis and treatment of each disease in the context of optimization the patient’s procreative capacity.



INTRODUCTION
According to World Health Organization infertility is a disease (ICD-10 code for female infertility is N97 and for male infertility – N46). It can be diagnosed when there is “the failure to achieve a clinical pregnancy after 12 months or more of regular unprotected sexual intercourse” (1). Therefore, the diagnostic procedures should start after 12 months of unsuccessful efforts to bear a child. From the beginning, it should concern both partners. From the beginning, it should concern both partners, as of infertility cases:
– 30% are due to a problem in the woman,
– 30% – in the man,
– 20% – both in the woman and in the man,
– and 20% remain unexplained (idiopathic).
To conceive and maintain a pregnancy, there is needed not only healthy sperm and healthy egg, properly built and functioning female and male genital organs, but also correct functioning of a number of factors, including hormones, to allow the embryo to nest in the uterine cavity, to be properly nourished, oxygenated, and tolerated by the maternal immune system.
It is crucial to differentiate inability to get pregnant (sterilitas) from inability to maintain pregnancy (infertilitas), usually manifested by recurrent miscarriages.
Recurrent miscarriages are defined as consecutive two or more spontaneous abortions (i.e., terminations of pregnancies prior to 20 weeks). They are mainly due to woman’s health problems. The most common causes are chromosomal abnormalities of egg, sperm or embryo. The probability of an egg cell aneuploidy increases with maternal age. Recurrent miscarriages may be also caused by endocrinopathies (thyroid diseases, PCOS, diabetes, corpus luteum insufficiency), as well as anatomical defects of the uterus, infections (especially Mycoplasma, Ureaplasma, Chlamydia), immunological (systemic lupus erythematosus, antiphospholipid syndrome) or hematologic disorders (thrombophilia). The relationship was also shown between recurrent miscarriages and excessive consumption of caffeine, alcohol and cigarette smoking (2, 3).
Nowadays more and more often the state of reduced fertility (subfertility) is observed, caused mainly by aging. Postponing parenthood “for later” further increases the risk of occurrence of chronic diseases, that affect fertility, and often masks them when potential parents are not aware of them.
The article discusses some causes of infertility and miscarriages, focusing on endocrine disorders.
HYPERGONADOTROPIC HYPOGONADISM
The most important factor affecting woman’s fertility is her age. It is an increasing problem (also a social problem), as the percentage of older women trying to conceive, rises. The function of ovaries is usually preserved until age of 40, afterward more and more cycles are anovulatory, the endocrine function of the ovary decreases and estradiol concentration in the luteal phase lowers. The symptoms of approaching menopause begin at about 47 years of age and last for about 4 years. The last period is usually between the age of 44 and 56, in Poland, statistically at 51.25 years of age (4). In men testosterone concentration may also decrease with age (late onset hypogonadism – LOH), but spermatogenesis remains preserved until senility. Reduced levels of estrogen in women and testosterone in men lead to increased pituitary gonadotropin secretion in a negative feedback mechanism (hypergonadotropic hypogonadism).
Premature ovarian failure, testicular failure
Premature ovarian failure (POF) can be diagnosed when, in a woman under 40 years of age, the concentration of FSH is above 40 IU/L twice at an interval of at least 4-6 weeks at low estradiol concentration and normal TSH and prolactin (PRL) levels. Ovarian reserve is reduced or falling to zero (low AMH – anti-Müllerian hormone level).
The cause of POF is unknown in 90% of cases. The ovarian injury may be the result of viral infections, smoking, and autoimmunity (5-9). POF may be a part of the autoimmune poliglandular syndrome (APS). Therefore, there is recommended screening for other potential autoimmune co-morbidities (Hashimoto’s thyroiditis, Addison’s disease, rheumatoid arthritis, systemic lupus erythematosus, Crohn’s disease, diabetes, celiac disease), including the determination of anti-ovarian antibodies, anti-thyroid peroxidase antibodies (a-TPO), anti-nuclear antibodies (ANA), rheumatoid factor (RF), anti-tissue transglutaminase antibodies, or if need performing a colonoscopy (10, 11).
POF may be also the result of radio- or chemotherapy (tab. 1) as well as surgical treatment of ovaries or testes. In the case of radiotherapy, ovarian failure occurs at doses > 0.06 Gy, and spermatogenesis depletion at doses > 1.20 Gy (12, 13). Irreversible damage to the ovaries occurs at a dose of > 8 Gy, and to testicular Leydig cells at a dose of > 20 Gy (12, 13). There were no reports indicating an increase in the percentage of congenital malformations, perinatal disorders or cancer in naturally conceived children of fathers previously treated for cancer by chemo- or radiotherapy, however the risk may be greater using IVF and ICSI because of a possible damage to the DNA of germ cells and lack of selection of sperm with normal DNA. To reduce the risk, it is recommended a delay of at least 12-18 months from the cessation of treatment to attempt of pregnancy.
Tab. 1. Drugs used in chemotherapy and the probability of fertility impairment
Probability of fertility impairmentDrugs
highcyclophosphamide, melphalan, dacarbazine, busulfan, chlormethine (e.g. MOPP protocol)
middlecisplatin, carboplatin, doxorubicin, BEP, ABVD
lowvincristine, methotrexate, bleomycin, mercaptopurine, vinblastine
Premature ovarian failure occurs in several genetic disorders, such as Turner syndrome (45X0), fragile X syndrome (FMR1 gene), pseudohypoparathyroidism type 1a (GNAS1 gene), galactosemia, Fanconi anemia, Bloom syndrome, Werner syndrome. There were also described rare cases of POF of genetic cause (POF1-7) (14, 15). When POF occurs in a woman under 30 years of age, cytogenetic testing with karyotype analysis is recommended (16).
The chances of getting pregnant in these cases are negligible. For people undergoing chemotherapy or radiotherapy, cryopreservation of ovarian cortex in women and sperm in men is proposed prior to the oncologic treatment. Causal treatment should be applied, if possible. There were case reports of restoring of gonadal function after applying of gluten-free diet in women with celiac disease. There were also trials of dehydroandrostenedione (DHEA) supplementation in women with POF. It was shown, that DHEA therapy in some patients with POF increases the chances of getting pregnant, reduces the risk of miscarriage and improves the results of IVF in women with POF (9, 10). Therefore, it is recommended determine DHEA-S concentration in patients with POF. The use of melatonin in women with POF may be also considered, as it was shown, that this hormone regulates menstruation and gonadotropin concentrations in the perimenopausal period (19-24).
To prevent other health consequences of hypogonadism, hormone replacement therapy is used.
Hypergonadotropic hypogonadism in men should always be an indication to determine the karyotype, because the most frequent (often undiagnosed!) cause of it is Klinefelter syndrome. Subsequently, there should be done screening for causes of secondary testicular failure (tumor, inflammation, toxic damage of the testes, metabolic disorders).
HYPOGONADOTROPIC HYPOGONADISM
Deficiency of pituitary gonadotropins (FSH and LH) leads to secondary ovaries/testes failure, manifested by menstrual disorders in women, erectile dysfunction in men, low libido, lack or loss of tertiary sexual characteristics with thinning or even lack of pubic and axillary hair as well as facial hair in men (19).
Hypogonadotropic hypogonadism (HH) may be caused by tumor, granulomatous or inflammatory diseases, as well as injuries or damage caused by ischemia, radiotherapy or chemotherapy of the area of hypothalamus or pituitary. Gonadotropin deficiency may also be genetically determined (e.g. Kallmann syndrome – an isolated GnRH deficiency, Pasqualini syndrome, GnRH-R mutations) or be the result of developmental disorders (e.g. empty sella syndrome, pituitary hypoplasia).
If the causal treatment is impossible, hormonal replacement therapy is used: sequential estrogen and progestins in women, and testosterone in men. It allows also to achieve normally developed sexual organs. To induce ovulation, a selective estrogen receptor modulator – clomiphene is used, and when this treatment is inefficient, gonadotropins or pulsatile gonadotropin-releasing hormone (GnRH) therapy are administered.
Spermatogenesis can be induced in men by clomiphene, gonadotropins or pulsatile GnRH treatment, however, it may require a long period of time (even up to 2 years).
Dysfunction of the hypothalamic-pituitary-gonadal axis
Impaired activation of the hypothalamic-pituitary-gonadal axis results in alterations of pulsatile GnRH secretion (usually reduced frequency and amplitude of GnRH pulses). In men, the effect is gonadotropin deficiency, which leads to reduction of testosterone secretion and disorders of spermatogenesis. In women, there is no gonadotropin surge, without which ovulation does not occur, or too low FSH and LH concentrations, which result in disturbances in maturation of Graafian follicles or luteal phase insufficiency.
Functional hypothalamic amenorrhea can be diagnosed when, in the absence of organic or anatomical disorders of the hypothalamic-pituitary-ovarian axis, LH and FSH values are within normal limits or are slightly reduced, and there are reduced estrogen concentrations in the second phase of the ovarian cycle (19). The GnRH test is useful in diagnosis. Functional HH is usually the result of negative energy balance, caused by excessive physical effort or significant weight loss, or mental stress (low leptin and kisspeptin concentrations lead to disturbances in GnRH secretion).
HH may be also secondary to chronic diseases: cancer, chronic gastrointestinal diseases causing malnutrition, liver diseases leading to its failure or chronic kidney disease.
Stress, excessive exercise
These factors increase the secretion of corticotropin releasing hormone (CRH) from the hypothalamus, which leads to increased concentrations of ACTH and cortisol. Hypercortisolism reduces the frequency of GnRH pulses. In females under the influence of prolonged stress (businesswomen) or excessive exercise (sportswomen) luteal phase insufficiency, anovulatory cycles, menstrual disorders, amenorrhea and infertility are often observed. In competitive athletes reduced leptin concentrations were also found, which is associated with a small amount of fat and severe energy consumption. The most predisposed to these disorders are women practicing synchronized swimming, artistic gymnastics, ballet and long-distance running.
Emaciation, intensive weight loss

Powyżej zamieściliśmy fragment artykułu, do którego możesz uzyskać pełny dostęp.
Mam kod dostępu
  • Aby uzyskać płatny dostęp do pełnej treści powyższego artykułu albo wszystkich artykułów (w zależności od wybranej opcji), należy wprowadzić kod.
  • Wprowadzając kod, akceptują Państwo treść Regulaminu oraz potwierdzają zapoznanie się z nim.
  • Aby kupić kod proszę skorzystać z jednej z poniższych opcji.

Opcja #1

24

Wybieram
  • dostęp do tego artykułu
  • dostęp na 7 dni

uzyskany kod musi być wprowadzony na stronie artykułu, do którego został wykupiony

Opcja #2

59

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 30 dni
  • najpopularniejsza opcja

Opcja #3

119

Wybieram
  • dostęp do tego i pozostałych ponad 7000 artykułów
  • dostęp na 90 dni
  • oszczędzasz 28 zł
Piśmiennictwo
1. Zegers-Hochschild F, Adamson GD, de Mouzon J et al.: International Committee for Monitoring Assisted Reproductive Technology (ICMART) and the World Health Organization (WHO) revised glossary of ART terminology, 2009. Fertil Steril 2009; 92: 1520-1524.
2. Cicinelli E, Matteo M, Tinelli R et al.: Chronic endometritis due to common bacteria is prevalent in women with recurrent miscarriage as confirmed by improved pregnancy outcome after antibiotic treatment. Reprod Sci 2014 May; 21(5): 640-647.
3. Ford HB, Schust DJ: Recurrent pregnancy loss: etiology, diagnosis, and therapy. Rev Obstet Gynecol 2009 Spring; 2(2): 76-83.
4. Karczmarek M: Określenie wieku menopauzy naturalnej w populacji polskich kobiet. Prz Menopauz 2007; 2: 77-82.
5. Kaufman DW, Slone D, Rosenberg L et al.: Cigarette smoking and age at natural menopause. Am J Public Health 1980; 70: 420-422.
6. Missmer S, Chavarro JE, Malspeis S et al.: A prospective study of dietary fat consumption and endometriosis risk. Hum Reprod 2010; 25: 1528-1535.
7. Klein NA, Battaglia DE, Fujimoto VY et al.: Reproducitve aging: accelerated ovarian follicular development associated with a monotropic follicle-stimulating hormone rise in normal older woman. J Clin Endocrin Metab 1996; 81: 1038.
8. Burger HG, Dudley EC, Hopper JL, Groome N: Prospectively measured levels of serum follicle-stimulating hormone, estradiol, and the dimeric inhibins during the menopausal transition in a population-based cohort of women. J Clin Endocrin Metabol 1999; 84: 4025.
9. Grynnerup AG, Lindhard A, Sorensen S: The role of anti-Müllerian hormone in female fertility and infertility – an overview. Acta Obstet Gyn Scand 2012 Nov; 91(11): 1252-1260.
10. Santoro N: Mechanisms of premature ovarian failure. Ann Endocrinol 2003; 64: 87-92.
11. van der Woude CJ, Kolacek S, Dotan I et al.: European Crohn’s Colitis Organisation (ECCO). J Crohns Colitis 2010 Nov; 4(5): 493-510.
12. Howell SJ, Shalet SM: Spermatogenesis after cancer treatment: damage and recover. J Nat Cancer Inst Monogr 2005; 34: 12-17.
13. Dębski R: Niepłodność. [W:] Antczak A, Myśliwiec M, Pruszczyk P (red.): Wielka Interna. Endokrynologia część I. Tom 12, Wyd. I. Medical Tribune, Warszawa 2011: 581-592.
14. Simpson JL, Rajkovic A: Ovarian differentiation and gonadal failure. Am J Med Genet 1999; 89: 186-200.
15. Lass A, Silye R, Abrams D-C et al.: Follicular density in ovarian biopsy of infertile women: a novel method to assess ovarian reserve. Hum Reprod 1997; 12: 1028.
16. Conway GS, Kaltsas G, Patel A et al.: Characterization of idiopathic premature ovarian failure. Fertil Steril 1996; 64: 337-341.
17. Mamas L, Mamas E: Premature ovarian failure and dehydroepiandrosterone. Fertil Steril 2009; 91: 644-666.
18. Gleicher N, Weghofer A, Barad DH: Dehydroepiandrosterone (DHEA) reduces embryo aneuploidy: direct evidence from preimplantation genetic screening (PGS). Reprod Biol Endocrinol 2010; 8: 140.
19. Yong PY, Baird DT, Thong KJ et al.: Prospective analysis of the relationships between the ovarian follicle cohort and basal FSH concentration, the inhibin response to exogenous FSH and ovarian follicle number at different stages of the normal menstrual cycle and after pituitary down-regulation. Hum Reprod 2003; 18: 35-44.
20. Iguchi H, Kato K, Ibayashi H: Are dependent reduction in serum-melatonin concentration in healthy human subjects. J Clin Endocrinol Metab 1982; 55: 27.
21. Bellipani G, Bianchi P, Pierpaoli W et al.: Effects of melatonin in perimenopausal and menopausal women: a randomized and placebo colntrolled study. Exp Gerontol 2001; 36(2): 297-310.
22. Cagnaci A, Paoletti AM, Soldani R et al.: Melatonin enhances the luteinizing hormone and follicle-stilulation hormone response to gonadotropin-releasing hormone in the follicular, but not in the luteal menstrual phase. J Clin Endocrinolo Metab 1995; 80(4): 1095-1099.
23. Carrillo-Vico A, Guerrero JM, Lardone PJ, Reiter RJ: A reviev of the multiple actions of melatonin on the immune system. Endocrine 2005; 27(2): 189-200.
24. Fisher TW, Zmijewski MA, Zbytek B et al.: Oncostatic effects of the indole melatonin and expression of its cytosolic and muclear receptors in cultured human melanoma cell lines. Int J Oncol 2006; 29(3): 665-672.
25. Becker A: Eating Disorders. N Engl J Med 1999; 341: 614-616.
26. Chan YM, Butler JP, Sidhoum VF et al.: Kisspeptin administration to wo-men: a window into endogenous kisspeptin secretion and GnRH respon-siveness across the menstrual cycle. J Clin Endocrinol Metab 2012; 97: E1458-E1467.
27. Fauser BC: Observations in favor of normal early follicle development and disturbed dominant follicle selection in polycystic ovary syndrome. Gynecol Endocrinol 1994; 8: 75-82.
28. Fauser BC, Tarlatzis BC, Rebar RW et al.: Consensus on women’s health aspects of polycystic ovary syndrome (PCOS). Fertil Steril 2012; 97: 28-38.
29. Domecq JP, Prutsky G, Mullan RJ et al.: Lifestyle modification programs in polycystic ovary syndrome: systematic review and meta-analysis. J Clin Endocrinol Metab 2013; 98: 4655-4663.
30. Michaliszyn SF, Lee S, Tfayli H, Arslanian S: Polycystic ovary syndrome and nonalcoholic fatty liver in obese adolescents: association with metabolic risk profile. Fertil Steril 2013; 100: 1745-1751.
31. Chavarro JE, Rich-Edwards JW, Rosner BA, Willett WC: Diet and lifestyle in the prevention of ovulatory disorder infertility. Obstet Gynecol 2007; 110: 1050-1058.
32. Belanger CF, Hennekens CH, Rosner B, Speizer FE: The nurses' health study. Am J Nurs 1978; 78: 1039-1040.
33. Pasquali R, Pelusi C, Genghini S et al.: Obesity and reproductive disorders in women. Hum Reprod Update 2003; 9: 359-372.
34. Isidori AM, Caprio M, Strollo F et al.: Leptin and androgens in male obesity: evidence for leptin contribution to reduced androgen levels. J Clin Endocrinol Metab 1999; 84: 3673-3680.
35. Zgliczyński W, Witek P, Zdunowski P: Guzy przysadki. Prolaktynoma i hiperprolaktynemia. [W:] Antczak A, Myśliwiec M, Pruszczyk P (red.): Wielka Interna. Endokrynologia część I. Tom 11. Wyd. I. Medical Tribune, Warszawa 2011: 67-74.
36. Casey BM, Dashe JS, Wells CE et al.: Subclinical hypothyroidism and pregnancy outcomes. Obstet Gynecol 2005; 105: 239-245.
37. Stagnaro-Green A, Abalovich M, Alexander E et al.: Guidelines of the American Thyroid Association for the diagnosis and management of thyroid disease during pregnancy and postpartum. Thyroid 2011; 21: 1081-1125.
38. Haddow JE, Palomaki GE, Allan WC et al.: Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child. N Engl J Med 1999; 341: 549-555.
39. Hubalewska-Dydejczyk A, Lewiński A, Milewicz A et al.: Postępowanie w chorobach tarczycy u kobiet w ciąży. Pol J Endocrinol 2011; 62: 362-381.
40. Glinoer D, de Nayer P, Bourdoux P et al.: Regulation of maternal thyroid during pregnancy. J Clin Endocrinol Metab 1990; 71: 276-287.
41. Negro R, Formoso G, Mangieri T et al.: Levothyroxine treatment in euthyroid pregnant women with autoimmune thyroid disease: effects on obstetrical complications. J Clin Endocrinol Metab 2006; 91: 2587-2591.
42. Männistö T, Vääräsmäki M, Pouta A et al.: Thyroid dysfunction and autoantibodies during pregnancy as predictive factors of pregnancy complications and maternal morbidity in later life. J Clin Endocrinol Metab 2010; 95: 1084-1094.
43. Negro R, Schwartz A, Stagnaro-Green A: Impact of levothyroxine in miscarriage and preterm delivery rates in first trimester thyroid antibody-positive women with TSH less than 2.5 mIU/L. J Clin Endocrinol Metab 2016; 101: 3685-3690.
44. Fauser BC: Observations in favor of normal early follicle development and disturbed dominant follicle selection in polycystic ovary syndrome. Gynecol Endocrinol 1994; 8: 75-82.
45. Lord JM, Flight IH, Norman RJ: Metformin in polycystic ovary syndrome: systematic review and meta-analysis. BMJ 2003; 327: 951-953.
46. Speroff L, Fritz Marc A (red.): Kliniczna endokrynologia ginekologiczna i niepłodność. Wyd. I. Medipage, Warszawa 2007: 1271, 1385.
47. Nestler JE, Jakubowicz DJ, Evans WS, Pasquali R: Effects of metformin on spontaneous and clomiphene-induced ovulation in the polycystic ovary syndrome. N Engl J Med 1998; 338: 1876-1880.
otrzymano: 2016-11-03
zaakceptowano do druku: 2016-11-30

Adres do korespondencji:
*Katarzyna Jankowska
Department of Endocrinology Centre of Postgraduate Medical Education Bielański Hospital
Cegłowska 80, 01-809 Warszawa
tel./fax +48 (22) 834-31-31
katarzynakamilajankowska@gmail.com

Postępy Nauk Medycznych 12/2016
Strona internetowa czasopisma Postępy Nauk Medycznych