Next Article in Journal
COVID-19 Related Myocarditis and Myositis in a Patient with Undiagnosed Antisynthetase Syndrome
Next Article in Special Issue
Does Nanosilver Exposure Modulate Steroid Metabolism in the Testes?—A Possible Role of Redox Balance Disruption
Previous Article in Journal
Modeling Central Nervous System Injury In Vitro: Current Status and Promising Future Strategies
Previous Article in Special Issue
Effect of High-Fructose Diet-Induced Metabolic Syndrome on the Pituitary-Gonadal Axis in Male Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

The Association between Vitamin D and the Components of Male Fertility: A Systematic Review

by
Daria Adamczewska
*,
Jolanta Słowikowska-Hilczer
and
Renata Walczak-Jędrzejowska
Department of Andrology and Reproductive Endocrinology, Medical University of Lodz, 92-213 Lodz, Poland
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(1), 90; https://doi.org/10.3390/biomedicines11010090
Submission received: 25 November 2022 / Revised: 20 December 2022 / Accepted: 24 December 2022 / Published: 29 December 2022
(This article belongs to the Special Issue Hormonal Regulation of Male Reproductive System)

Abstract

:
Objective: Previous systematic reviews of the effects of vitamin D on the components of male fertility have been inconclusive. This article systematically reviews the latest research to examine the relationship between vitamin D, semen quality parameters, and sex hormones production. Methods: MEDLINE, Cochrane, and Web of Science databases were searched using the appropriate keywords. Results: Observational studies indicate significant correlation between vitamin D levels and sperm parameters, with a particular emphasis on sperm motility, and partially suggest a relationship between higher serum testosterone and vitamin D levels. Additionally, interventional studies confirmed that vitamin D supplementation has a positive effect on sperm motility, especially progressive. However, most randomized clinical trials indicate that vitamin D treatment does not have any significant effect on testosterone or other hormone levels. Conclusions: Although our findings add to the discussion regarding the effect of vitamin D on male fertility, there is still no solid evidence to support the use of vitamin D supplementation to improve the outcomes of patients with impaired sperm parameters and hormonal disorders. Additional dedicated clinical studies are needed to clarify the relationship between vitamin D and male fertility, along with its components.

1. Introduction

The male gonads perform two very important functions that are essential for the male reproductive system; they produce gametes, i.e., spermatozoa, and release sex hormones [1]. Their proper functioning depends on many different factors: age, the influence of nutrition, stimulants, hormones, pharmacological agents, and substances that disrupt the hormonal and vitamin balance [2]. Currently, the research on male reproductive health is focusing on the growing global trend of declining sperm count and increasing male reproductive system abnormalities, which has been related, among others, to lifestyle factors as well as environmental impact. Among these, numerous studies have investigated the role of vitamin D (VD) in the proper functioning of the male reproductive system.
Initially, the action of VD was associated mainly with the maintenance of calcium and phosphorus homeostasis, which have a significant effect on skeleton mineralization. However, in recent years, new knowledge has emerged about its biological function and its potential to reduce the risk of many chronic diseases [3]. The known range of its target organs has expanded, and the discovery of vitamin D receptor (VDR) expression and vitamin D metabolizing enzymes (VDME) in the testes, male reproductive tract, and human sperm suggests that VD plays a role in the male reproductive role [4]. Although several reviews have been made on the subject, no consensus has been reached regarding the role of VD in male fertility. Moreover, new intervention studies in humans have only appeared in the last few years
The aim of this paper is to provide an up-to-date, comprehensive review of the existing literature regarding the experimental and clinical evidence for the effects of VD on the components of male fertility, sperm parameters, and sex hormone production.

2. Background

2.1. Vitamin D Physiology

VD is a fat-soluble vitamin based on a sterol ring. There are two forms of VD: vitamin D2 (ergocalciferol, VD2), found in plants and fungi, and D3 (cholecalciferol, VD3), produced in animals. In humans, it can be obtained through the diet, but about 90% of the VD detectable in human serum is synthesized in the skin from a precursor (provitamin D3) present in epidermal cells. The provitamin D3 is transformed into provitamin D3 under the influence of solar ultraviolet B radiation (wavelength 290–315 nm), and then it is converted to VD3 by heat-dependent isomerization [5,6] (Figure 1). The VD produced in the skin enters the circulation and is transported to the liver by a protein called vitamin D-binding protein (VDBP). In contrast, VD from the diet is absorbed in the small intestine, where it binds to chylomicrons, which are then released into the lymphatic system and enter the venous circulation. These chylomicrons then bind with VDBP and reach the liver [7].
Although VD is classified as a vitamin, in practice it is a prohormone, because in the body it is transformed into a biologically active form, 1alpha,25-dihydroxyvitamin D, 1,25(OH)2D. The first stage of conversion to its active form takes place in the liver. Vitamin 25(OH)D—calcidiol—is produced by enzymatic hydroxylation of a 25-carbon molecule by one of the four hepatic cytochrome P-450 enzymes. Three of them are microsomal forms, CYP2R1, CYP2J2, and CYP3A4. The fourth enzyme CYP27A1 is mitochondrial [8,9]. 25(OH)D is transferred, in combination with VDBP, from the liver to the kidneys, where CYP27B1 creates another form of VD, calcitriol, 1,25(OH)2D [3,10]. Calcitriol is the hormonally active form of VD that is responsible for most of its biological effects.
Calcitriol acts on various cells of the body, mainly through the VDR located in the cell nucleus. The VDR is a nuclear receptor that belongs to the superfamily of steroid hormone receptors, which includes receptors for retinoic acid, thyroid hormone, sex hormones, and adrenal steroids. The VDR is made up of three main domains: a linker, a DNA binding unit, and a ligand binding unit.
Calcitriol can act through VDR by genomic (classical) and non-genomic pathways [11]. In the former, 1,25(OH)2D attaches to a nuclear VDR, which then binds to the retinoid X receptor (RXR). The resulting VDR-RXR heterodimer functions as a transcription factor—it binds to a specific DNA sequence present in the promoter regions, referred to as the vitamin D response element (VDRE), which induces or inhibits the expression of target genes [12]. Advanced genomic techniques such as ChIP-seq have shown that VDR has thousands of genome-wide binding sites influencing the transcription of hundreds of different genes. In a study with human lymphoblastoid cell lines treated with 1,25(OH)2D, 2776 VDR binding sites were found to alter the expression of 229 genes [13]. In contrast, the latter, non-genomic pathway involves the activation of signaling molecules such as phospholipase C and phospholipase A2, phosphatidylinositol-3 kinase and p21ras, and the rapid generation of second messengers (Ca2+, cyclic AMP, fatty acids), accompanied by the activation of protein kinases, such as protein kinase A, src, mitogen-activated protein kinases, protein kinase C (PKC), and Ca2+-calmodulin kinase II [14,15,16,17,18,19]. It also includes the opening of Ca2+ and Cl channels [20]. It has also been recognized that the VDR can mediate rapid non-genomic actions when located in the membrane or cytoplasm [21].
The circulating forms of VD are inactivated in the kidneys and in several target organs [22]. Ultimately, through a series of successive reactions catalyzed by the enzyme 24-hydroxylase (identified as CYP24A1), 1,25(OH)2D is converted into water-soluble calcitric acid, which can be excreted in the urine [23,24].
The level of VD storage in the human body is best reflected by the concentration of 25(OH)D in the serum; 1,25(OH)2D assessment is of limited utility [25,26]. The North American Institute of Medicine (IOM) Dietary Reference Committee (DRI), in cooperation with UK and European Union authorities, established dietary reference values for VD in adults using serum 25(OH)D levels as an indicator of VD status [27]. They came to the conclusion that a serum level of 20 ng/mL is essential for bone health [28]. However, other medical expert committees disagreed, claiming that a value of at least 30 ng/mL is required to prevent skeletal and other system diseases [29,30,31].

2.2. Vitamin D in Male Reproductive System

In the early days of research into the metabolism of VD, the kidney was thought to be the only site of 1,25(OH)2D production. While the kidney is the primary endocrine organ that produces 1,25(OH)2D in response to other calcium- and phosphate-regulating hormones, it is now known that other organs also have the ability to convert 25(OH)D to 1,25(OH)2D [24]. Subsequently, the discovery of the action of VDME and the expression of VDR in the male reproductive system suggested that VD plays a role in male fertility. One of the first significant discoveries was the detection of VDR in human testicular tissue homogenates using titrated VD [32]. In 2006, the presence of VDR was confirmed in human sperm, with VDR expression being observed predominantly in the head and nucleus of fertile men’s sperm, with some localization in the neck region [33]. An investigation, based on electron microscopy and gold labeling, found most of the distribution to occur in the nucleus, with just a minor quantity of staining in the neck [34]. The presence of nuclear VDR in terminally differentiated, transcriptionally inactive cells, such as sperm, suggests that VDR may be a remnant of late spermatogenic processes and may be associated with non-testicular sperm maturation. On the other hand, the presence of VDR can act as a mechanism of genome stability and protection against DNA strand breaks [34,35]. These hypotheses require further research.
Studies on the expression of the VDR and VDME indicate the presence of VDR, CYP2R1, CYP27A1, CYP27B1, and CYP24A1 in human testes, epididymis, prostate, seminal vesicles, and mature sperm. The activity of VDME regulates VD intracellular concentration and, thus, the activation of the VDR. For this reason, local expression of VDME is crucial. Germ cells of adult testes such as spermatogonia, spermatocytes, and round and elongated spermatids primarily express VDRs and metabolizing enzymes, indicating that local regulation of active VD may be important for spermatogenesis and/or sperm function [4,36].
Hormone-producing Leydig cells also express VDR and VDME, indicating a direct link between VD and the synthesis of sex steroids [4]. Indeed, individuals with testicular problems were found to have lower systemic 25(OH)D levels than healthy controls due to reduced local CYP2R1 expression in Leydig cells [37].
The male reproductive function is mediated by the hypothalamic–pituitary–gonad axis. Both the VDR and the enzymes that metabolize VD are found in the central organs of this system. In the brain, VDR and 1α-hydroxylase are most strongly expressed in the substantia nigra and hypothalamus. VDR expression at the gene and protein level has also been confirmed in the pituitary gland [38,39].
Animal studies further support the role of VD in reproduction, revealing a link between VD deficiency and reduced fertility or gonadal dysfunction. VDR has been discovered in the smooth muscles of the epididymis, spermatogonia, and Sertoli cells of male rodents, indicating a role for vitamin D in spermatogenesis and sperm maturation in rats [40]. Early studies have shown that male rats fed a vitamin D-deficient diet have lower fertility than animals fed a vitamin D-rich diet. The number of successful mating, expressed as the presence of sperm in the female reproductive tract, decreased by 45–55% in male rats deficient in VD [41,42]. Additionally, the fertility of normal female rats inseminated by vitamin D-deficient males was reduced by 73% [42]. In both studies, male rats fed a diet deficient in VD showed changes in testicular histology. Males with hypovitaminosis had lower testicular and epididymal sperm counts, Sertoli cell dysfunction, fewer Leydig cells, and degenerative changes in the reproductive epithelium [41,42]. Similar results were obtained in VDR knockout mouse models. Male mice with the VDR null mutation had lower sperm count and motility, as well as testicular histological abnormalities, including a dilated lumen of the seminiferous tubules, a thinner epithelial cell layer, and decreased spermatogenesis [43]. These findings supported the effect of VD on testicular function and, by extension, male infertility.

3. Materials and Methods

A review of relevant literature was conducted using the MEDLINE, Cochrane, and Web of Science databases. The following keywords were used in the search: vitamin D, cholecarciferol, ergocarciferol, vitamin D levels, male fertility, male infertility, semen, sperm, sex hormones, testosterone, estradiol, follicle stimulating hormone, luteinizing hormone, sex hormone binding globulin, inhibin b. During the search, individual keywords and their combinations were typed using AND, OR, or both. The literature search covered papers published from January 2011 to June 2022. Only English-language scientific papers were taken into account. All published randomized clinical trials, retrospective, prospective, observational, and comparative human studies were included, while case reports, comments/letters to the editor, and reviews were omitted. The analysis did not include any in vitro or animal investigations. Title and abstracts were assessed, and articles were classified according to predefined inclusion and exclusion criteria. The eligibility of each article found was assessed independently by two reviewers, and excluded records were verified by another. Full text of selected articles was obtained for further review. Two reviewers independently assessed the quality of the full-text papers describing the strength and level of confidence of the results using the levels of evidence published by The Oxford Centre for Evidence-Based Medicine (CEBM) in 2009 [44]. An Excel file was created containing the following data from selected publications: name of the first author of each study, year of publication, country, sample size, age of participants, health status, study design, variables used in adjustments, reported semen, and sex hormones rates. One reviewer extracted data, the correctness of which was checked by other reviewers, and all discrepancies were resolved through discussion. This review was registered in the INPLASY platform of registered systematic review and meta-analysis protocols (INPLASY2022110151) and followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA statement) guidelines [45].

4. Results

An initial database search found 830 studies. After applying the eligibility criteria to the titles and abstracts, 193 articles were eligible for the full-text review. This number was reduced to 53 eligible studies by a literature search and screening process. The selection approach is shown in the PRISMA chart (Figure 2). All detailed data collected from the studies are presented in Table 1, Table 2, Table 3 and Table 4. Studies on VD included patients from 24 different nations: Argentina, Austria, Bangladesh, Brazil, China, Denmark, Egypt, Germany, Holland, India, Iran, Ireland, Italy, Jordan, Malaysia, Norway, Pakistan, Poland, Qatar, Slovakia, South Korea, Spain, Turkey, and the USA. The age of the participants ranged from 18 to 97 years. Five studies focused on young people, six on elderly males, and the remaining studies examined middle-aged men or patients of various ages. The analyzed participants were drawn from the general population (n = 16), were patients of andrology clinics/had semen abnormalities (n = 27), or were selected on the basis of other criteria (n = 10).
Out of the included 53 articles, 32 focused on the effect of VD on sperm parameters (Table 1 and Table 2) and 34 discussed the effect on sex hormones (Table 3 and Table 4). Furthermore, 36 studies were classified as observational and 17 as interventional. Various techniques were used to assess the level of VD in the serum: chemiluminescence immunoassay (n = 13), electrochemiluminescence (n = 7), enzyme-linked immunosorbent assay (n = 10), isotope dilution liquid chromatography tandem mass spectrometry (n = 8), radioimmunoassay (n = 5), high performance liquid chromatography (n = 4), competitive protein-binding assay (n = 2), enzyme-linked fluorescent assay (n = 1), liquid chromatography mass spectrometry (n = 1), chemiluminescent microparticle immunoassay (n = 1). Four studies did not report the method of measurement.

4.1. Vitamin D and Semen Parameters: Observational Studies

Of the included articles, 23 investigated the relationship between VD levels and semen quality using observational studies. All are cross-sectional studies. The studies are summarized in Table 1.
VD levels can be seen to have a beneficial effect on sperm quality, as indicated by a positive correlation between VD serum levels and total and/or progressive sperm motility [35,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60]. This correlation was significant in men from the general population [35,46], but also in patients of andrology clinics/men with semen abnormalities [47,48,49,50,51,52,53,54,55,56,57,58,59,60]. However, seven studies failed to link VD to sperm motility or returned statistically insignificant results [61,62,63,64,65,66,67].
In contrast, the existence of a correlation between VD levels and sperm count and sperm morphology is uncertain. Only eight studies showed a positive relationship between sperm concentration/total number and VD levels [46,52,53,54,55,57,58,60], and ten publications confirmed a correlation between VD levels and normal sperm morphology [35,47,51,52,53,54,56,57,58]. Only one study found that low VD levels are not a risk factor for poor semen quality in a population of young healthy men, although high VD levels were unexpectedly associated with a lower percentage of normal sperm morphology. However, after the adjustment for potential confounders, the results were found to be insignificant [61].
In contrast, a cross-sectional study of 170 men revealed an association between U-shaped VD levels and semen parameters, showing that both low and high VD levels are associated with poor semen quality [46]. In this study, men with 25(OH)D ≥ 50 ng/mL (≥125 nmol/mL) had lower sperm concentration, sperm progressive motility, sperm morphology, and total progressive motile sperm count compared to men with 20 ng/mL ≤ 25(OH)D < 50 ng/mL (50 nmol/mL ≤ 25(OH)D < 125 nmol/mL) [46].
The first study on the relationship between serum VD levels and sperm DNA fragmentation in humans appeared in 2018. Azizi et al. [51] postulated that there may be a link between serum 25-OHD and reactive oxygen species (ROS) in semen and sperm DNA fragmentation. The results suggest that increasing 25(OH)D levels are associated with a decrease in apoptotic cells and ROS, however, the relationship was not significant. Since then, only two studies have been conducted that showed a significant negative correlation between VD and sperm DNA fragmentation [53,58].

4.2. Vitamin D and Semen Prameters: Interventional Studies

The last few years have seen several studies on the effect of VD supplementation on sperm parameters. This review collated ten such studies, six of which were randomized clinical trials, two prospective interventional studies, one longitudinal observation study, and one case control. The summary and general effect of VD supplementation on semen parameters in clinical trials are presented in Table 2.
Many studies have shown an improvement in motility, especially progressive after supplementation with VD [68,69,70,71,72,73]. However, only four of the intervention studies demonstrated any efficacy in increasing sperm concentration [69,70,71,73]. Only one study found administration of VD to improve sperm morphology [70]. The effects do not seem to be dependent on the length of VD supplementation: both short-term (2–3 months) and long-term (5–6 months) supplementation was able to improve patient outcomes [68,69,70,71,72,73,74,75]. In addition, studies of the same length differed in their results: both positive results [71,72,73] and no changes [75,76] were obtained after three-month supplementation with VD. These differences may be due to adjustment for potential confounders; however, all of the three-month studies were unadjusted for any covariates.
Table 1. Summary of Observational Studies between Vitamin D and Semen Quality Parameters in Men.
Table 1. Summary of Observational Studies between Vitamin D and Semen Quality Parameters in Men.
StudyCountryStudy DesignNumber of PatientsAge
(Years) 1
Characteristics
of Patients
Vitamin D
Measurement Method
Sperm
Parameters
AdjustmentLevel
of Evidence
2022 Güngör et al. [58]TurkeyCross-Sectional108F: 33.2 ± 4.1
I: 34.7 ± 4.0
Fertile and infertile menECLIASC↑ TM↑ NM↑ DF↓Unadjusted3b
2022 Holzer et al. [59] ArgentinaCross-Sectional5620–45Andrological
patients
CLIASC→ TM↑ PM↑ NM→Unadjusted4
2022 Kamal et al. [67]EgyptCross-Sectional10035.0 ± 8.4
(20–50)
Andrological
patients
ELISASC→ PM→ NM→Unadjusted4
2022 Rezayat et al. [60]IranCross-Sectional114F: 34.9 ± 13.5
I: 34.6 ± 12.2
(20–59)
Fertile and infertile menELISASC↑ TM↑ NM→Unadjusted3b
2021 Ciccone et al. [57]BrazilCross-Sectional260SA: 38.7 ± 8.5
NS: 38.0 ± 8.8
(18–60)
Men with semen abnormalities and normozoospermic menECLIA/HPLCSC↑ TM↑ PM↑ NM↑Unadjusted3b
2021 Hajianfar et al. [54]IranCross-Sectional35034.8 ± 0.4
(20–50)
Andrological
patients
ECLIASC↑ TM↑ NM↑Age, Educational Status, Smoking, Alcohol Consumption, Job, Varicocele, BMI, Total Energy Intake4
2021 Hussein et al. [55]EgyptCross-Sectional100F: 31.4 ± 8.9
(19–58)
I: 32.8 ± 6.9
(18–50)
Fertile and infertile menELFASC↑ PM↑ NM→Unadjusted3b
2021 Kumari et al. [56]IndiaCross-Sectional22418–45Men with semen abnormalities and normozoospermic menCMIASC↑ TM↑ PM↑ NM↑Unadjusted3b
2020 Derakhshan et al. [53]IranCross-Sectional70SVD: 33.7 ± 5.7
IVD: 36.3 ± 6.5
(18–60)
Andrological
patients
HPLCSC↑ TM↑ PM↑ NM↑ DF↓Unadjusted4
2020 Rudnicka et al. [66]SpainCross-Sectional19818–23Young men from general populationCLIASC→ TM→ NM→Age, BMI, Smoking, Physical activity, Season, Ejaculation Abstinence Time, Time to Start of Semen Analysis2b
2019 Jueraitetibaike et al. [65]ChinaCross-Sectional22230Andrological
patients
ECLIASC→ TM→ PM→ NM→Unadjusted4
2018 Azizi et al. [51]IranCross-Sectional62NS: 34.1 ± 1.2
OAT: 33.0 ± 0.7
Men with semen abnormalities and normozoospermic menCLIASC→ TM↑ NM↑ DF→Unadjusted3b
2018 Jóźków et al. [64]PolandCross-Sectional17724.6 ± 3.6 (20–35)Young men from general populationECLIASC→ PM→Smoking, Alcohol Consumption, Carrying a Telephone in a Pants Pockets, BMI, WHR, Caffeine Consumption, Physical Activity2b
2018 Rehman et al. [52]PakistanCross-Sectional31325–55Fertile and infertile menNaSC↑ TM↑ NM↑Vitamin D adjusted with BMI3b
2017 Abbasihormozi et al. [49]IranCross-Sectional27833.5 ± 4.8 (20–50)Men with oligoasthenoteratozoospermia and normozoospermic menECLIASC→ TM↑ PM→ NM→Age, BMI, Season3b
2017 Tirabassi et al. [50]ItalyCross-Sectional10433.1 ± 4.8Andrological
patients
CLIASC→ TM↑ PM↑ NM→Age, BMI, PTH, Varicocele4
2016 Blomberg Jensen et al. [48]DenmarkCross-Sectional118934.3Andrological
patients
ID-LC-MS/MSSC→ TM↑ PM↑ NM→Age, BMI, Smoking, Season, Abstinence, Time from Collection, Free Testosterone, Estradiol, Total Estradiol, Testosterone/Estradiol Ratio2b
2016 Neville et al. [62]IrelandCross-Sectional7337.4 ± 4.4Men from couples undergoing IVF/ICSICPBASC→ TM→ PM→ NM→Unadjusted4
2016 Zhu et al. [63]ChinaCase-Control265F: 28.22 ± 0.5
O: 28.8 ± 0.8
A: 28.3 ± 0.6
OA: 27.7 ± 0.6
AZ: 27.2 ± 0.6
NS: 28.4 ± 0.4
Fertile and infertile menELISASC→ TM→ PM→Unadjusted3b
2012 Hammound et al. [46]USACross-Sectional14729.0 ± 8.5 (18–67)General populationCLIASC↑ PM↑ NM→Age, BMI, Season, Alcohol Consumption, Smoking2b
2012 Yang et al. [47]ChinaCross-Sectional55920–40Fertile and infertile menELISATM↑ NM↑Season, Abstinence, Time from Collection3b
2011 Blomber Jensen et al. [35]DenmarkCross-Sectional30019.0General populationID-LC-MS/MSSC→ TM↑ PM↑ NM↑Abstinence, Season, Medication, Fever, Time from Ejaculation to Motility Assessment2b
2011 Ramlau-Hansen et al. [61]DenmarkCross-Sectional30718–21Young men from general populationID-LC-MS/MSSC→ TM→ NM→Season, History of Diseases of the Reproductive Organs, Smoking, Maternal Smoking During Pregnancy, Maternal Alcohol During Pregnancy, Abstinence, Spillage During Collection of The Sample2b
Abbreviations: A: asthenospermia; AZ: azoospermia; CLIA: chemiluminescence immunoassay; CMIA: chemiluminescent microparticle immunoassay; CPBA: competitive protein-binding assay; DF: DNA fragmentation; ECLIA: electrochemiluminescence; ELFA: enzyme-linked fluorescent assay; ELISA: enzyme-linked immunosorbent assay; F: fertile; HPLC: high performance liquid chromatography; I: infertile; ID-LC-MS/MS: isotope dilution liquid chromatography tandem mass spectrometry; IVD: insufficient vitamin D; Na: Not available; NM: normal morphology; NS: normozoospermic; O: oligospermia; OA: oligoasthenospermia; OAT: oligoasthenoteratozoospermic; PM: progressive motility; RIA: radioimmunoassay; SA: seminal abnormalities; SC: sperm concentration; SVD: sufficient vitamin D; TM: total motility. → no correlation; ↑ positive correlation; ↓ negative correlation. 1 Age values are presented as mean ± standard deviation, mean ± standard deviation (range), range only, or median only.
Table 2. Summary of Interventional Studies between Vitamin D and Semen Quality Parameters in Men.
Table 2. Summary of Interventional Studies between Vitamin D and Semen Quality Parameters in Men.
StudyCountryStudy Design (Duration)Vitamin D DoseNumber
of Patients
Age
(Years) 1
Characteristics
of Patients
Vitamin D
Measurement
Method
Sperm
Parameters
AdjustmentLevel
of Evidence
2022 Padmapriya et al. [73]IndiaDBRCT
(3 months)
28,000 IU of VD3 weekly12030–40Men with oligoasthenoteratozoospermiaNaSC↑ TM↑ PM↑Unadjusted1b
2021 Bartl et al. [70]SlovakiaProspective interventional study
(6 months)
17,500 IU of VD3 weekly3436.6Infertile menHPLCSC↑ PM↑ NM↑Unadjusted2b
2021 Begum et al. [71]BangladeshProspective interventional study
(3 months)
40,000 IU of VD3 weekly for six weeks, 14,000 IU of VD3 weekly for another six weeks11033.2 ± 5.8 (25–45)Men with asthenozoospermia and vitamin D deficiencyCLIASC↑ TM↑ PM↑Unadjusted2b
2021 Gheflati et al. [76]IranDBRCT
(3 months)
50,000 IU of VD3 weekly for eight weeks and one maintenance dose of 50,000 IU for another four weeks4418–45Men with asthenozoospermiaELISASC→ PM→ NM→Unadjusted1b
2021 Maghsoumi-Norouzabad et al. [72]IranTBRCT
(3 months)
28,000 IU of VD3 weekly86VD: 35.1 ± 5.5 P: 34.4 ± 5.1Men with asthenozoospermiaELISASC→ TM↑ PM↑ NM→Unadjusted1b
2020 Amini et al. [75]IranTBRCT
(3 months)
50,000 IU of VD3 weekly for eight weeks and one maintenance dose of 50,000 IU for another four weeks6235–39Men with semen abnormalitiesELISASC→ TM→ PM→ NM→Unadjusted1b
2020 Wadhwa et al. [69]IndiaProspective interventional study
(6 months)
60,000 IU of VD3 and 3500 mg of calcium weekly6030.6 ± 4.0 (23–40)Men with asthenozoospermia, oligozoospermia, or bothNaSC↑ PM↑Unadjusted2b
2018 Blomberg Jensen et al. [74]DenmarkTBRCT
(5 months)
single dose of 300,000 IU of VD3, then9800 IU of VD3 and 3500 mg of calcium weekly33034.8 ± 6.6Men with semen abnormalitiesID-LC-MS/MSSC→ TM→ PM→ NM→Unadjusted1b
2017 Alzoubi et al. [68]JordanCase-Control
(2 months)
35,000 IU of VD3 weekly3420–45Men with semen abnormalitiesELISASC→ TM↑ PM↑ NM→Unadjusted2b
Abbreviations: CLIA: chemiluminescence immunoassay; DF: DNA fragmentation; ELISA: enzyme-linked immunosorbent assay; HPLC: high performance liquid chromatography; ID-LC-MS/MS: isotope dilution liquid chromatography tandem mass spectrometry; IVD: insufficient vitamin D; Na: Not available; NM: normal morphology; P: placebo; PM: progressive motility; SC: sperm concentration; SVD: sufficient vitamin D; TM: total motility; VD: vitamin D; → no change; ↑ increase. 1 Age values are presented as mean ± standard deviation, mean ± standard deviation (range), mean only, range only, or median only.

4.3. Vitamin D and Sex Hormones: Observational Studies

Both VDR and the enzymes metabolizing VD are expressed in Leydig cells [4], suggesting that VD plays a direct role in regulating steroidogenesis. Twenty-two of the human studies of the association between 25(OH)D and sex hormones are summarized in Table 3.
The effect of VD on serum levels of male sex hormones has been analyzed in several studies, but with inconsistent results. Most observational studies indicate that serum 25(OH)D3 levels were not associated with circulating total testosterone (TT) or free testosterone (FT) levels [46,47,48,49,50,59,61,77,78,79,80,81], with exceptions in seven studies [52,57,60,82,83,84,85]. In addition, some of these studies revealed a significant association between 25(OH)D and sex hormone binding globulin (SHBG) [48,59,61,77,79]. In addition, two studies found a negative association with FT and a positive relationship with SHBG [48,79]. In such situations, VD deficiency may indirectly affect the hormonal state by modulating the bioavailable fraction of testosterone.
The relationship between VD and testosterone may be age dependent. The majority of studies on older men (over 50) show a positive relationship between 25(OH)D levels and testosterone [82,83,84,86,87]. However, this association disappeared after adjusting for confounding factors such as comorbidities [86]. No association was found between 25(OH)D and testosterone in men aged under 35 [46,48,61,88]. This suggests that the relationship between VD and testosterone may be influenced by age-varying factors such as an increase of SHBG level, as well as age-related comorbidities including endocrine pathologies.
Moreover, studies by Zhao et al. [79] and Blomberg Jensen et al. [48] revealed a negative correlation between 25(OH)D3 and estradiol (E2) level. In contrast, two other studies showed a positive relationship with E2 [83,87]. However, less promising results were noted for other hormones; none of the eligible studies discovered any connection between VD and FSH, and one study found a negative association between LH and VD [52]. In addition, only one study investigated the correlation between VD and inhibin B, and it was found to be positive [48].

4.4. Vitamin D and Sex Hormones: Interventional Studies

The search also encompassed the potential impact of VD supplementation on male hormones. The results are given in Table 4.
Unfortunately, intervention studies have not achieved any unequivocal findings on the effects of VD on sex hormones. The effects seemed to be variable and dependent on the length of VD supplementation. Short-term (1.5–4 months) supplementations were not capable to influence circulating levels of TT [72,75,76,89,90,91,92]. Long-term (6–12 months) VD supplementation in different age groups may generate a significant increase in TT [93] and/or FT and SHBG [94] or no change [70,95]. However, even longer (36 months) interventions did not show a positive effect with a decrease in FT levels being noted [96]. One study found VD therapy to have a positive effect on estradiol levels [90] while another found a negative one [93]. SHBG has been found to decrease with VD supplementation [75,90]. The other hormones were not affected by supplementation or were not taken into account in the studies. Ergocalciferol (VD2) supplementation was only used in one study [93]. However, the findings do not indicate that VD3 supplementation yielded any significant benefits in this regard.
Table 3. Summary of observational studies between vitamin D and sex steroids in men.
Table 3. Summary of observational studies between vitamin D and sex steroids in men.
StudyCountryStudy DesignNumber
of Patients
Age
(Years) 1
Characteristics
of Patients
Vitamin D Measurement MethodHormonesAdjustmentLevel
of Evidence
2022 Holzer et al. [59]ArgentinaCross-Sectional5620–45Andrological
patients
CLIATT→ FT→ E2→ LH→ FSH→ SHBG ↑Unadjusted4
2022 Rezayat et al. [60]IranCross-Sectional114F:34.9 ± 13.5
I:34.6 ± 12.2
(20–59)
Fertile and infertile menELISATT↑ LH→ FSH→Unadjusted3b
2022 Talebi et al. [81]IranCross-Sectional22034.5 ± 5.6
(20–45)
Men with semen abnormalitiesNaTT→ FT→ LH→ FSH→Unadjusted4
2021 Ciccone et al. [57]BrazilCross-Sectional260SA: 38.7 ± 8.5NS: 38.0 ± 8.8
(18–60)
Men with semen abnormalities and normospermic menECLIA/HPLCTT↑Unadjusted3b
2021 Książek et al. [80]PolandCross-Sectional17618–35Active young men from general populationECLIATT→ FT→ LH→ FSH→ SHBG →Age, BMI, Smoking, Alcohol Consumption, WHR2b
2019 Chen et al. [97]ChinaCross-Sectional425418–93General populationCLIATT↑Age, Economic Status, Smoking, BMI, Hypertension, Diabetes2b
2018 Rehman et al. [52]PakistanCross-Sectional31325–55Fertile and infertile menNaTT↑ LH↓ FSH→Vitamin D adjusted with BMI3b
2017 Abbasihormozi et al. [49]IranCross-Sectional27833.5 ± 4.8 (20–50)Men with oligoasthenoteratozoospermia and normospermic menECLIATT→ FT→ LH→ FSH→Age, BMI, Season3b
2017 Tirabassi et al. [50]ItalyCross-Sectional10433.1 ± 4.8Andrological
patients
CLIATT→Age, BMI, PTH, Varicocele4
2017 Zhao et al. [79]USACross-Sectional301762.1 ± 10.2 (45–84)General populationHPLC–tandem mass spectrometryTT→ FT↓ E2↓ SHBG ↑Age, Race/Ethnicity, Study Site, BMI, Smoking, Education, Self-Reported Good Health Status, Intentional Physical Activity, Diabetes, Systolic Blood Pressure, Use of Antihypertensive Medications, eGFR, Total Cholesterol, HDL Cholesterol, Use of Lipid Lowering Medication Usage, C-Reactive Protein2b
2016 Anic et al. [85]USACross-Sectional1633≥20General populationRIATT↑ FT→ SHBG ↑Age, Race/Ethnicity, % Body Fat, Smoking2b
2016 Barbonetti et al. [78]ItalyCross-Sectional4947.5 ± 17.3Men with chronic spinal cord injuryCLIATT→ FT↑Age, Smoking, Alcohol Consumption, Coexisting Illness, Homeostatic Model Assessment of Insulin Resistance, Functional Independence Degree (Barthel
Index), BMI, Weekly Leisure Time Physical Activity
4
2016 Blomberg Jensen et al. [48]DenmarkCross-Sectional118934.3Andrological
patients
ID-LC-MS/MSTT→ FT↓ E2↓ LH→ FSH→ SHBG ↑ INHB↑Age, BMI, Smoking, Season, Abstinence, Time from Collection, Free Testosterone, Estradiol, Total Estradiol, Testosterone/Estradiol Ratio2b
2016 Rafiq et al. [84]HollandCohort Study64365–89Older men from general populationCPBATT↑ FT→ E2→ LH→ FSH→ SHBG →Age, BMI, Season, Alcohol Consumption, Smoking, Number of Chronic Diseases, Physical Function, Serum Creatinine2b
2015 Chin et al. [77]MalaysiaCross-Sectional38243.5 ± 15.5General populationELISATT→ FT→ SHBG ↑Age, BMI, Race/Ethnicity2b
2015 Tak et al. [82]South KoreaCross-Sectional65256.7 ± 7.9
(40–80)
Men over 40 years old from general populationCLIATT↑ FT↑ TT: Age, BMI, Waist Circumference, % Body Fat, Fasting Plasma Glucose, Diabetes, Dyslipidemia. FT: Age, Total Muscle Mass, Smooth Muscle Mass, Total Cholesterol, Diabetes, Dyslipidemia, Alcohol Consumption2b
2015 Wang et al. [83]ChinaCross-Sectional285453.0 ± 13.5General populationCLIATT↑ FT→ E2↑ LH→ FSH→ SHBG →Age, BMI, Residence Area, Economic Status, Smoking, HOMA-IR, Diabetes, Systolic Pressure2b
2014 Lerchbaum et al. [88]AustriaCross-Sectional22535Middle-aged men from general populationID-LC-MS/MSTT→ FT→ E2→ LH→ FSH→ SHBG →Unadjusted4
2012 Hammound et al. [46]USACross-Sectional14729.0 ± 8.5
(18–67)
General populationCLIATT→ FT→ E2→ LH→ FSH→ SHBG →Age, BMI, Season, Alcohol Consumption, Smoking2b
2012 Lee et al. [86]EuropeCross-Sectional336940–79General populationRIATT→ FT→ E2→ LH→ FSH→ SHBG →Age, Centre, BMI, Smoking, Alcohol Consumption, Physical Activity, Physical Function, Heart Conditions, Hypertension, Diabetes, Depression2b
2012 Nimptsch et al. [87]USACross-Sectional136240–75Participants selected for a nested case–control study on prostate cancerRIATT↑ FT↑ E2↑Age, Batch, BMI, Season, Geographical Region, Smoking, Physical Activity, Time of Blood Collection. TT Further Adjusted for SHBG3b
2011 Ramlau-Hansen et al. [61]DenmarkCross-Sectional30718–21Young men from general populationID-LC-MS/MSTT→ E2→ LH→ FSH→ SHBG ↑Season, History of Diseases of the Reproductive Organs, Smoking, Maternal Smoking During Pregnancy, Maternal Alcohol During Pregnancy, Abstinence, Spillage During Collection of the Sample2b
Abbreviations: BMI: body mass index; CLIA: chemiluminescence immunoassay; CPBA: competitive protein-binding assay; E2: estradiol; ECLIA: electrochemiluminescence; ELISA: enzyme-linked immunosorbent assay; F: fertile; FSH: follicle stimulating hormone; FT: free testosterone; HOMA-IR: homeostatic model assessment –insulin resistance; HPLC: high performance liquid chromatography; I: infertile; ID-LC-MS/MS: isotope dilution liquid chromatography tandem mass spectrometry; INHB: inhibin B; LH: luteinizing hormone; Na: Not available; NS: normozoospermic; PTH: parathormone; RIA: radioimmunoassay; SA: seminal abnormalities; SHBG: sex hormone binding globulin; TT: total testosterone; WHR: waist-to-hip ratio. → no correlation; ↑ positive correlation; ↓ negative correlation. 1 Age values are presented as mean ± standard deviation, mean ± standard deviation (range), range only, or median only. In one study, the age of patients was presented as greater than/equal to 20 (≥20).
Table 4. Summary of Interventional Studies between Vitamin D and Sex Steroids in Men.
Table 4. Summary of Interventional Studies between Vitamin D and Sex Steroids in Men.
StudyCountryStudy Design (Duration)Vitamin D DoseNumber
of Patients
Age
(Years) 1
Characteristics
of Patients
Vitamin D
Measurement Method
HormonesAdjustmentLevel
of Evidence
2021 Bartl et al. [70]SlovakiaProspective interventional study
(6 months)
17,500 IU of VD3 weekly3436.6Infertile menHPLCTT→ FT→ E2→ LH→ FSH→ SHBG →Unadjusted2b
2021 Gheflati et al. [76]IranDBRCT
(3 months)
50,000 IU of VD3 weekly for eight weeks and one maintenance dose of 50,000 IU for another four weeks4418–45Men with asthenozoospermiaELISATT→ SHBG →Unadjusted1b
2021 Maghsoumi-Norouzabad et al. [72]IranTBRCT
(3 months)
28,000 IU of VD3 weekly86VD:35.1 ± 5.5
P:34.4 ± 5.1
Men with asthenozoospermiaELISATT→ E2→ LH→ FSH→ SHBG →Unadjusted1b
2021 Ulrich et al. [92]GermanyDBRCT
(3 months)
5600 IU of VD3 weekly3520–71General populationLC-MS/MSTT→Unadjusted1b
Dose of VD3 adapted to patients VD serum levels1820–71Hemodialysis patientsCLIATT→Unadjusted1b
2020 Amini et al. [75]IranTBRCT
(3 months)
50,000 IU of VD3 weekly for eight weeks and one maintenance dose of 50,000 IU for another four weeks6235–39Men with semen abnormalitiesELISATT→ FT→ LH→ FSH→ SHBG ↓Unadjusted1b
2019 Lerchbaum et al. [91]AustriaDBRCT
(3 months)
20,000 IU of VD3 weekly9447.0 ± 12.0
(18–70)
Men with serum TT levels < 10.4 nmol/LID-LC-MS/MSTT→ FT→ E2→ LH→ FSH→ SBHG→Unadjusted1b
2019 Zittermann et al. [96]GermanyRCT
(36 months)
28,000 IU of VD3 weekly13318–79Men with advanced heart failure, mean TT level of 11.2 nmol/LCLIATT→ FT SHBG →Unadjusted1b
2017 Canguven et al. [93]QatarProspective interventional study
(12 months)
150,000 IU of VD2 weekly; when serum 25(OH)D level reached 75 nmol/L, VD dose switched to 600,000 IU every eight weeks10253.2 ± 10.5
(35–64)
Men with serum 25(OH)D levels < 30 ng/mLELISATT→ E2 LH→Unadjusted2b
2017 Lerchbaum et al. [90]AustriaDBRCT
(3 months)
20,000 IU of VD3 weekly9818–70Men with serum 25(OH)D levels < 75 nmol/L and TT levels ≥ 10.4 nmol/LID-LC-MS/MSTT→ FT→ E2↑ LH→ FSH→ SHBGUnadjusted1b
2015 Heijboer et al. [89]HollandRCT
(1.5 months)
14,000 IU of VD3 weekly9242–86Male patients with chronic heart failureID-LC-MS/MSTT→Unadjusted1b
RCT
(4 months)
4200 IU of VD3 weekly4971–97Male nursing home residentsRIATT→Unadjusted1b
RCT
(4 months)
8400 IU of VD3 daily4320–70Vitamin D deficient male patientsID-LC-MS/MSTT→Unadjusted1b
2013 Jorde et al. [95]NorwayRCT
(12 months)
20,000–40,000 IU of VD3 and 3500 mg of calcium weekly12948.9 ± 10.6
(21–70)
Men with BMI 28–47 kg/m2CLIATT→ FT→ LH→ FSH→ SHBG →Unadjusted1b
RCT
(6 months)
40,000 IU of VD3 weekly5351.2 ± 10.0
(30–75)
Men with serum 25(OH)D levels < 50 nmol/LLC-MS/MSTT→ FT→ SHBG →Unadjusted1b
RCT
(6 months)
40,000 IU of VD3 weekly10053.0 ±11.1 (30–75)Men with serum 25(OH)D levels < 55 nmol/LLC-MS/MSTT→ FT→ SHBG→Unadjusted1b
2011 Pilz et al. [94]AustriaRCT
(12 months)
23,324 IU of VD3 weekly54VD:49.4 ± 10.2
P:46.8 ± 12.0
Healthy, overweight men, mean TT level of 10.7 nmol/LRIATT↑ FT↑Unadjusted1b
Abbreviations: CLIA: chemiluminescence immunoassay; E2: estradiol; ELISA: enzyme-linked immunosorbent assay; FSH: follicle stimulating hormone; FT: free testosterone; HPLC: high performance liquid chromatography; ID-LC-MS/MS: isotope dilution liquid chromatography tandem mass spectrometry; INHB: inhibin B; LC-MS/MS: liquid chromatography mass spectrometry; LH: luteinizing hormone; P: placebo; RIA: radioimmunoassay; SHBG: sex hormone binding globulin; TT: total testosterone; VD: vitamin D. → no change; ↑ increase; ↓ decrease. 1 Age values are presented as mean only, mean ± standard deviation, mean ± standard deviation (range), or range only.

5. Discussion

Little is known about the role VD plays in male reproduction, despite the identification of VDR, and its associated enzymes, in the reproductive system. Most knowledge in this area has been obtained from cross-sectional observational studies, with interventional studies only appearing recently. As such, the data provides only a vague answer. The main goal of this systematic review was to synthesize evidence showing a link between VD, sperm quality parameters and sex hormone production. Table 5 summarizes the major findings from the studies included in this review.
Four out of six studies comparing infertile and fertile men revealed that the serum vitamin D levels of infertile men were considerably lower than those of fertile men [52,55,58,60]. This finding is in line with previous meta-analysis on this topic and suggests that VD has beneficial effects on male reproduction [98]. The analysis of the correlation between VD and sperm quality parameters showed it to have the most significant impact on sperm motility. In most of the included articles, serum VD levels were significantly associated with total sperm motility [35,47,48,49,50,51,52,53,54,56,57,58,59,60] and progressive motility [35,46,48,50,53,55,56,57,59]. Sperm motility is one of the key elements affecting successful fertilization. Therefore, it is important to know the factors affecting it and the underlying mechanisms. Sperm motility, as an energy-consuming process, is strongly tied to mitochondrial adenosine triphosphate (ATP) synthesis. Mitochondrial function is regulated by many substances, one of which is cyclic adenosine monophosphate (cAMP). cAMP as a cellular second messenger acts by activating various downstream factors such as protein kinase A (PKA) [99]. Jueraitetibaike et al. [65] proposed a mechanism for increasing sperm motility by promoting ATP synthesis through the cAMP/PKA pathway, as well as an increase in intracellular ions, which is influenced by VD. In this in vitro study, sperm kinetic parameters increased after incubation with 1,25(OH)2D, and with them ATP concentrations, cAMP concentrations, PKA activity, and cytoplasmic calcium concentration also increased. This would support the experimental evidence that points to a direct non-genomic effect of VD on human sperm.
There is considerable disagreement among researchers regarding the relationship between VD levels and other sperm parameters. Fewer than half of the studies found a positive and statistically significant correlation between VD levels and sperm concentration [46,52,53,54,55,56,57,58,60] and/or morphology [35,47,51,52,53,54,56,57].
This is the first systematic review to examine the impact of VD on sperm DNA fragmentation; to date, only three articles on this subject appear to have been published, all of which are observational. Only two of them confirmed a negative correlation between the level of sperm DNA fragmentation and the level of VD [53,58]. In addition, in the MOXI study, where men from couples with a mild factor of male infertility were given multivitamin preparations containing, among others, vitamin D, it was not possible to find differences in sperm DNA fragmentation between men with and without vitamin D deficiency [100]. Hence, it has been suggested that the sperm nuclear matrix plays a critical role in regulating DNA fragmentation and degradation. VDR is closely related to the nuclear matrix, and VD is believed to play a significant role in stabilizing the chromosomal structure and thus protecting DNA from harm and breakage. Therefore, it is plausible that VD and its receptor act as protectors of the sperm genome [101,102]. However, this requires further research.
The evidence for the effects of VD on the production of male sex steroid hormones was less apparent. Sex steroids are produced by Leydig cells [103]. On Leydig cells, VDR and VD metabolizing enzymes are also expressed. VD, when bound to VDR, acts as a transcription factor in the promoter regions of the gene encoding the steroidogenesis enzyme. The articles mentioned in the review show an inconsistent relationship between VD and the production of sex hormones. No significant correlation was found between LH, FSH, and VD levels in any of the analyzed studies. However, some studies have found a significant correlation between VD and TT, FT, and SHBG levels. Despite evidence from several studies [93,94,104,105], a recent meta-analysis did not support the hypothesis that VD stimulates testosterone levels [106]. The exact molecular mechanism linking VD to testosterone production is still unknown. Some authors have suggested the hypothesis that genomic stimulation of osteocalcin expression by VD may play a key role in the regulation of testosterone production by the testes [107].
In men, estrogens are mainly derived from circulating androgens and metabolism is regulated by the enzyme aromatase. VD has been functionally related to estrogen signaling and VDR has been found to affect estrogen levels systemically and locally through tissue-specific regulation of aromatase [108]. However, to date, no human studies have shown a convincing causal link between VD status and estrogen production [109]. More research is needed to investigate how VD status affects not only estrogen production but also estrogen signaling, which may be another direct target of VDR.
The role of VD in the male reproductive system is still poorly understood. However, evidence from animal research that connects VD insufficiency with infertility or gonadal malfunction strengthens the significance of VD in reproduction. In a study by Zamani et al. [110], animals kept on a VD-deficient diet demonstrated profoundly impaired sperm concentration, morphology, and motility, and a significant reduction in serum testosterone levels and comparable gonadotropin levels compared to controls. The depletion of VD stores and the induction of moderate degrees of VD deficiency by dietary measures significantly impaired spermatogenesis and the microscopic testicular architecture in rats. These findings can be attributed, at least in part, to decreased androgen production.
The first in vivo study brought attention to the strong relation between D hypovitaminosis in rats and decreased levels of circulating testosterone [111]. However, it is difficult to establish the exact mechanism of the hormonal regulation of testosterone production. Some authors support the theory that in animals, testosterone secretion may be regulated by changes in intracellular calcium homeostasis in Leydig cells. It has been proposed that testosterone synthesis as a result of VD action may be mediated by osteocalcin, a hormone generated by osteoblasts that is involved in bone metabolism [112]. Recent research has revealed the impact of the fat–VD relationship on sperm motility and mitochondrial membrane potential, with animals fed a high-fat diet demonstrating lower reproductive potential than animals fed a control diet; clearly, a high-fat diet and VD deficiency both influence sperm quality, which may therefore affect fertility [113]. Additionally, significantly increased DNA fragmentation was observed in the sperm of animals fed a diet deficient in VD and those that were obese due to diet. Based on this, it can be concluded that the observed DNA damage may be the cause of the low reproductive potential in obese men deficient in VD [104].
Testosterone biosynthesis is closely related to the lipid metabolism in the gonads around the testes. VD regulates the synthesis and metabolism of lipids in adipose tissue through VDR [114]. Vdr knockout animals had impaired testicular and perinuclear adipose tissue histology, indicating a strong relationship between lipid metabolism and VDR-mediated testosterone production. These findings support the belief that VDR plays a crucial role in controlling testicular lipid metabolism, which in turn affects testosterone production and reproduction potential in male mice [115]. Although the results of animal studies are very promising, they have not been reproduced in humans yet. It is possible that the relationship between VD and semen quality parameters noted in rodent studies is not relevant to humans, or that these associations may only be reflected in severe VD deficiency.
The most important study examining whether VD has a direct effect on male reproductive function are randomized clinical trials with VD supplementation. Despite promising findings from animal studies and human observational studies, the results of interventional studies do not yield unequivocal conclusions. Of the 10 intervention studies reviewed, 6 showed that VD supplementation improved progressive sperm motility [68,69,70,71,72,73]. The remaining findings indicated that treatment with VD had no significant effect on changes in other sperm parameters.
To ascertain whether VD supplementation can affect blood testosterone levels, several studies have been performed. One investigation found that treating VD-deficient individuals with VD increased testosterone levels. However, in the majority of randomized clinical trials, no substantial effect of VD supplementation on male testosterone levels was reported. These discrepancies should be regarded with caution. For example, conditions with severe VD3 deficiency are likely to respond more positively to VD3 supplementation on sperm parameters than those with mild or no deficiency. Previous studies on men found a relationship between severe VD3 deficiency and decreased fertility [48,74,116]. Moreover, there is mounting evidence that traditional VD supplementation fails to achieve appropriate 25(OH)D levels. Since VD is fat-soluble, it accumulates mostly in adipose tissue [117]. Obese men are more vulnerable to deficiency in this manner. When VD buildup in adipose tissue limits release into the systemic circulation, individuals with autoimmune disorders may require very large doses of VD treatment to boost blood 25(OH)D levels slightly [118]. Given that obese people with autoimmune disorders frequently have 25(OH)D insufficiency and decreased testosterone levels, situations like these should be examined in future studies [119,120].
The results of the studies selected for this review should be treated with caution due to some limitations. Most studies used a small sample size with high levels of heterogeneity in the study design. In addition, possible confounding factors (other causes of infertility, comorbidities, lifestyle, eating habits, age, season, ethnicity) may hinder the correct interpretation of the research. The inability to make appropriate comparisons between studies is also due to the variability in the choice of cut-off values for determining vitamin D deficiency by the authors. Another limitation that certainly influences the results are the VDBP levels and VDR polymorphisms, neither of which were addressed in the studies. Therefore, no statistical analysis of these results was performed as high heterogeneity would lead to ambiguous or unreliable results. Moreover, most of the studies included in the review are cross-sectional studies, and their nature makes it impossible to delineate a causal relationship between the variables. For these reasons, the clinical relevance of all observations made should be confirmed by further research.

6. Conclusions

The data presented in this review demonstrate that VD exerts a beneficial effect on sperm quality, particularly sperm motility, which is likely driven by the modulation of factors related to sperm function, namely the direct non-genomic effect of VD on sperm in men. In addition, none of the included studies have been able to exhibit a clear connection between testicular hormone production and VD levels. This evidence is mainly based on small association studies in humans and rodent models. However, there is still a lack of solid evidence from interventional studies, as these tend to be based on small sample sizes and demonstrate considerable variability regarding the optimal dose and duration of VD supplementation. Due to the increasing number of men suffering from reproductive problems and endocrine disorders, more high-quality RCTs are needed to elucidate the effects of VD supplementation on sperm parameters, especially sperm DNA fragmentation, and sex hormones.

Author Contributions

Conceptualization, D.A., J.S.-H. and R.W.-J.; investigation, D.A. and R.W.-J.; writing—original draft preparation, D.A.; writing—review and editing J.S.-H. and R.W.-J.; supervision, J.S.-H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Medical University of Lodz, Statute No. 503/1-089/503-11001-19-00.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Edward Lowczowski, a native speaker from the Foreign Language Center of the Medical University of Lodz, for proofreading of this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Weinbauer, G.F.; Luetjens, C.M.; Simoni, M.; Nieschlag, E. Physiology of testicular function. In Andrology; Springer: Berlin/Heidelberg, Germany, 2010; pp. 11–59. [Google Scholar] [CrossRef]
  2. Mortimer, D.; Barratt, C.L.R.; Björndahl, L.; de Jager, C.; Jequier, A.M.; Muller, C.H. What should it take to describe a substance or product as “sperm-safe”. Hum. Reprod. Update 2013, 19 (Suppl. S1), i1–i45. [Google Scholar] [CrossRef] [Green Version]
  3. Battault, S.; Whiting, S.J.; Peltier, S.L.; Sadrin, S.; Gerber, G.; Maixent, J.M. Vitamin D metabolism, functions and needs: From science to health claims. Eur. J. Nutr. 2013, 52, 429–441. [Google Scholar] [CrossRef]
  4. Blomberg Jensen, M.; Nielsen, J.E.; Jørgensen, A.; Rajpert-De Meyts, E.; Kristensen, D.M.; Jørgensen, N.; Skakkebaek, N.E.; Juul, A.; Leffers, H. Vitamin D receptor and vitamin D metabolizing enzymes are expressed in the human male reproductive tract. Hum. Reprod. 2010, 25, 1303–1311. [Google Scholar] [CrossRef] [Green Version]
  5. Holick, M.F.; Maclaughlin, J.A.; Clark, M.B.; Holick, S.A.; Potts, J.T.; Anderson, R.R.; Blank, I.H.; Parrish, J.A.; Elias, P. Photosynthesis of Previtamin D3 in Human Skin and the Physiologic Consequences. Science 1980, 210, 203–205. [Google Scholar] [CrossRef]
  6. Chang, S.W.; Lee, H.C. Vitamin D and health—The missing vitamin in humans. Pediatr. Neonatol. 2019, 60, 237–244. [Google Scholar] [CrossRef] [Green Version]
  7. Bouillon, R.; Schuit, F.; Antonio, L.; Rastinejad, F. Vitamin D Binding Protein: A Historic Overview. Front. Endocrinol. (Lausanne) 2020, 10, 910. [Google Scholar] [CrossRef]
  8. Cheng, J.B.; Motola, D.L.; Mangelsdorf, D.J.; Russell, D.W. De-orphanization of Cytochrome P450 2R1. J. Biol. Chem. 2003, 278, 38084–38093. [Google Scholar] [CrossRef] [Green Version]
  9. Schuster, I. Cytochromes P450 are essential players in the vitamin D signaling system. Biochim. Biophys. Acta 2011, 1814, 186–199. [Google Scholar] [CrossRef]
  10. Gil, Á.; Plaza-Diaz, J.; Mesa, M.D. Vitamin D: Classic and Novel Actions. Ann. Nutr. Metab. 2018, 72, 87–95. [Google Scholar] [CrossRef]
  11. McDuffie, J.R.; Calis, K.A.; Booth, S.L.; Uwaifo, G.I.; Yanovski, J.A. Effects of orlistat on fat-soluble vitamins in obese adolescents. Pharmacotherapy 2002, 22, 814–822. [Google Scholar] [CrossRef]
  12. Haussler, M.R.; Haussler, C.A.; Jurutka, P.W.; Thompson, P.D.; Hsieh, J.C.; Remus, L.S.; Selznick, S.H.; Whitfield, G.K. The vitamin D hormone and its nuclear receptor: Molecular actions and disease states. J. Endocrinol. 1997, 154 (Suppl. S3), S57–S73. [Google Scholar] [PubMed]
  13. Ramagopalan, S.V.; Heger, A.; Berlanga, A.J.; Maugeri, N.J.; Lincoln, M.R.; Burrell, A.; Handunnetthi, L.; Handel, A.E.; Disanto, G.; Orton, S.M.; et al. A ChIP-seq defined genome-wide map of vitamin D receptor binding: Associations with disease and evolution. Genome Res. 2010, 20, 1352–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Dwivedi, P.P.; Hii, C.S.T.; Ferrante, A.; Tan, J.; Der, C.J.; Omdahl, J.L.; Morris, H.A.; May, B.K. Role of MAP kinases in the 1,25-dihydroxyvitamin D3-induced transactivation of the rat cytochrome P450C24 (CYP24) promoter. Specific functions for ERK1/ERK2 and ERK5. J. Biol. Chem. 2002, 277, 29643–29653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Fleet, J.C. Rapid, membrane-initiated actions of 1,25 dihydroxyvitamin D: What are they and what do they mean? J. Nutr. 2004, 134, 3215–3218. [Google Scholar] [CrossRef] [Green Version]
  16. Nutchey, B.K.; Kaplan, J.S.; Dwivedi, P.P.; Omdahl, J.L.; Ferrante, A.; May, B.K.; Hii, C.S.T. Molecular action of 1,25-dihydroxyvitamin D3 and phorbol ester on the activation of the rat cytochrome P450C24 (CYP24) promoter: Role of MAP kinase activities and identification of an important transcription factor binding site. Biochem. J. 2005, 389, 753–762. [Google Scholar] [CrossRef] [Green Version]
  17. Dwivedi, P.P.; Gao, X.H.; Tan, J.C.T.; Evdokiou, A.; Ferrante, A.; Morris, H.A.; May, B.K.; Hii, C.S.T. A role for the phosphatidylinositol 3-kinase--protein kinase C zeta--Sp1 pathway in the 1,25-dihydroxyvitamin D3 induction of the 25-hydroxyvitamin D3 24-hydroxylase gene in human kidney cells. Cell. Signal. 2010, 22, 543–552. [Google Scholar] [CrossRef]
  18. Doroudi, M.; Schwartz, Z.; Boyan, B.D. Membrane-mediated Actions of 1,25-Dihydroxy Vitamin D3: A Review of the Roles of Phospholipase A2 Activating Protein and Ca2+/Calmodulin-dependent Protein Kinase II. J. Steroid Biochem. Mol. Biol. 2015, 147, 81–84. [Google Scholar] [CrossRef] [Green Version]
  19. Hii, C.S.; Ferrante, A. The Non-Genomic Actions of Vitamin D. Nutrients 2016, 8, 135. [Google Scholar] [CrossRef] [Green Version]
  20. Norman, A.W. Minireview: Vitamin D receptor: New assignments for an already busy receptor. Endocrinology 2006, 147, 5542–5548. [Google Scholar] [CrossRef]
  21. Haussler, M.R.; Jurutka, P.W.; Mizwicki, M.; Norman, A.W. Vitamin D receptor (VDR)-mediated actions of 1α,25(OH)2vitamin D3: Genomic and non-genomic mechanisms. Best Pract. Res. Clin. Endocrinol. Metab. 2011, 25, 543–559. [Google Scholar] [CrossRef]
  22. Veldurthy, V.; Wei, R.; Campbell, M.; Lupicki, K.; Dhawan, P.; Christakos, S. 25-Hydroxyvitamin D3 24-Hydroxylase: A Key Regulator of 1,25(OH)2D3 Catabolism and Calcium Homeostasis. Vitam. Horm. 2016, 100, 137–150. [Google Scholar] [CrossRef] [PubMed]
  23. Prosser, D.E.; Jones, G. Enzymes involved in the activation and inactivation of vitamin D. Trends Biochem. Sci. 2004, 29, 664–673. [Google Scholar] [CrossRef] [PubMed]
  24. Henry, H.L. Regulation of vitamin D metabolism. Best Pract. Res. Clin. Endocrinol. Metab. 2011, 25, 531–541. [Google Scholar] [CrossRef] [PubMed]
  25. Hollis, B.W. Assessment of circulating 25(OH)D and 1,25(OH)2D: Emergence as clinically important diagnostic tools. Nutr. Rev. 2007, 65 (Suppl. S2), S87–S90. [Google Scholar] [CrossRef]
  26. Hollis, B.W. Assessment of vitamin D status and definition of a normal circulating range of 25-hydroxyvitamin D. Curr. Opin. Endocrinol. Diabetes. Obes. 2008, 15, 489–494. [Google Scholar] [CrossRef]
  27. Cashman, K.D.; van den Heuvel, E.G.H.M.; Schoemaker, R.J.W.; Prévéraud, D.P.; Macdonald, H.M.; Arcot, J. 25-Hydroxyvitamin D as a Biomarker of Vitamin D Status and Its Modeling to Inform Strategies for Prevention of Vitamin D Deficiency within the Population. Adv. Nutr. 2017, 8, 947–957. [Google Scholar] [CrossRef] [Green Version]
  28. Institute of Medicine (US) Committee to Review Dietary Reference Intakes for Vitamin D and Calcium. Dietary Reference Intakes for Calcium and Vitamin D; Ross, A.C., Taylor, C.L., Yaktine, A.L., Del Valle, H.B., Eds.; National Academies Press (US): Washington, DC, USA, 2011. [Google Scholar] [CrossRef]
  29. Dawson-Hughes, B.; Mithal, A.; Bonjour, J.P.; Boonen, S.; Burckhardt, P.; Fuleihan, G.E.H.; Josse, R.G.; Lips, P.; Morales-Torres, J.; Yoshimura, N. IOF position statement: Vitamin D recommendations for older adults. Osteoporos. Int. 2010, 21, 1151–1154. [Google Scholar] [CrossRef] [Green Version]
  30. Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.; Weaver, C.M. Evaluation, treatment, and prevention of vitamin D deficiency: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2011, 96, 1911–1930. [Google Scholar] [CrossRef]
  31. Recommendations Abstracted from the American Geriatrics Society Consensus Statement on Vitamin D for Prevention of Falls and Their Consequences. J. Am. Geriatr. Soc. 2014, 62, 147–152. [CrossRef]
  32. Habib, F.K.; Maddy, S.Q.; Gelly, K.J. Characterisation of receptors for 1,25-dihydroxyvitamin D3 in the human testis. J. Steroid Biochem. 1990, 35, 195–199. [Google Scholar] [CrossRef]
  33. Corbett, S.T.; Hill, O.; Nangia, A.K. Vitamin D receptor found in human sperm. Urology 2006, 68, 1345–1349. [Google Scholar] [CrossRef] [PubMed]
  34. Aquila, S.; Guido, C.; Perrotta, I.; Tripepi, S.; Nastro, A.; Andò, S. Human sperm anatomy: Ultrastructural localization of 1alpha,25-dihydroxyvitamin D receptor and its possible role in the human male gamete. J. Anat. 2008, 213, 555–564. [Google Scholar] [CrossRef] [PubMed]
  35. Blomberg Jensen, M.; Bjerrum, P.J.; Jessen, T.E.; Nielsen, J.E.; Joensen, U.N.; Olesen, I.A.; Petersen, J.H.; Juul, A.; Dissing, S.; Jørgensen, N. Vitamin D is positively associated with sperm motility and increases intracellular calcium in human spermatozoa. Hum. Reprod. 2011, 26, 1307–1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Blomberg Jensen, M.; Jørgensen, A.; Nielsen, J.E.; Steinmeyer, A.; Leffers, H.; Juul, A.; Meyts, E.R. De Vitamin D Metabolism and Effects on Pluripotency Genes and Cell Differentiation in Testicular Germ Cell Tumors In Vitro and In Vivo. Neoplasia 2012, 14, 952. [Google Scholar] [CrossRef] [Green Version]
  37. Foresta, C.; Strapazzon, G.; De Toni, L.; Perilli, L.; Di Mambro, A.; Muciaccia, B.; Sartori, L.; Selice, R. Bone Mineral Density and Testicular Failure: Evidence for a Role of Vitamin D 25-Hydroxylase in Human Testis. J. Clin. Endocrinol. Metab. 2011, 96, E646–E652. [Google Scholar] [CrossRef] [Green Version]
  38. Pérez-Fernandez, R.; Alonso, M.; Segura, C.; Muñoz, I.; García-Caballero, T.; Diéguez, C. Vitamin D receptor gene expression in human pituitary gland. Life Sci. 1996, 60, 35–42. [Google Scholar] [CrossRef] [PubMed]
  39. Eyles, D.W.; Smith, S.; Kinobe, R.; Hewison, M.; McGrath, J.J. Distribution of the vitamin D receptor and 1 alpha-hydroxylase in human brain. J. Chem. Neuroanat. 2005, 29, 21–30. [Google Scholar] [CrossRef]
  40. Johnson, J.A.; Grande, J.P.; Roche, P.C.; Kumar, R. Immunohistochemical detection and distribution of the 1,25-dihydroxyvitamin D3 receptor in rat reproductive tissues. Histochem. Cell Biol. 1996, 105, 7–15. [Google Scholar] [CrossRef]
  41. Sood, S.; Marya, R.K.; Reghunandanan, R.; Singh, G.P.; Jaswal, T.S.; Gopinalhan, K. Effect of vitamin D deficiency on testicular function in the rat. Ann. Nutr. Metab. 1992, 36, 203–208. [Google Scholar] [CrossRef]
  42. Uhland, A.M.; Kwiecinski, G.G.; DeLuca, H.F. Normalization of serum calcium restores fertility in vitamin D-deficient male rats. J. Nutr. 1992, 122, 1338–1344. [Google Scholar] [CrossRef]
  43. Kinuta, K.; Tanaka, H.; Moriwake, T.; Aya, K.; Kato, S.; Seino, Y. Vitamin D Is an Important Factor in Estrogen Biosynthesis of Both Female and Male Gonads. Endocrinology 2000, 141, 1317–1324. [Google Scholar] [CrossRef] [PubMed]
  44. Oxford Centre for Evidence-Based Medicine: Levels of Evidence (March 2009)—Centre for Evidence-Based Medicine (CEBM), University of Oxford. Available online: https://www.cebm.ox.ac.uk/resources/levels-of-evidence/oxford-centre-for-evidence-based-medicine-levels-of-evidence-march-2009 (accessed on 15 November 2022).
  45. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  46. Hammoud, A.O.; Wayne Meikle, A.; Matthew Peterson, C.; Stanford, J.; Gibson, M.; Carrell, D.T. Association of 25-hydroxy-vitamin D levels with semen and hormonal parameters. Asian J. Androl. 2012, 14, 855–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Yang, B.; Sun, H.; Wan, Y.; Wang, H.; Qin, W.; Yang, L.; Zhao, H.; Yuan, J.; Yao, B. Associations between testosterone, bone mineral density, vitamin D and semen quality in fertile and infertile Chinese men. Int. J. Androl. 2012, 35, 783–792. [Google Scholar] [CrossRef] [PubMed]
  48. Blomberg Jensen, M.; Lawaetz, J.G.; Andersson, A.M.; Petersen, J.H.; Nordkap, L.; Bang, A.K.; Ekbom, P.; Joensen, U.N.; Prætorius, L.; Lundstrøm, P.; et al. Vitamin D deficiency and low ionized calcium are linked with semen quality and sex steroid levels in infertile men. Hum. Reprod. 2016, 31, 1875–1885. [Google Scholar] [CrossRef] [Green Version]
  49. Abbasihormozi, S.; Kouhkan, A.; Alizadeh, A.R.; Shahverdi, A.H.; Nasr-Esfahani, M.H.; Sadighi Gilani, M.A.; Salman Yazdi, R.; Matinibehzad, A.; Zolfaghari, Z. Association of vitamin D status with semen quality and reproductive hormones in Iranian subfertile men. Andrology 2017, 5, 113–118. [Google Scholar] [CrossRef]
  50. Tirabassi, G.; Cutini, M.; Muscogiuri, G.; delli Muti, N.; Corona, G.; Galdiero, M.; Pivonello, R.; Colao, A.; Balercia, G. Association between vitamin D and sperm parameters: Clinical evidence. Endocrine 2017, 58, 194–198. [Google Scholar] [CrossRef] [Green Version]
  51. Azizi, E.; Naji, M.; Shabani-Nashtaei, M.; Sobhani, A.; Najafi, A.; Amidi, F. Association of serum content of 25-hydroxy vitamin D with semen quality in normozoospermic and oligoasthenoteratozoospermic men. Int. J. Reprod. Biomed. 2018, 16, 689. [Google Scholar]
  52. Rehman, R.; Lalani, S.; Baig, M.; Nizami, I.; Rana, Z.; Gazzaz, Z.J. Association Between Vitamin D, Reproductive Hormones and Sperm Parameters in Infertile Male Subjects. Front. Endocrinol. (Lausanne) 2018, 9, 607. [Google Scholar] [CrossRef]
  53. Derakhshan, M.; Derakhshan, M.; Omidi, E.; Heidarpour, M. The association between serum vitamin D level and sperm parameters; A pilot study in a subset of Iranian infertile males. Immunopathol. Persa 2020, 6, e30. [Google Scholar] [CrossRef]
  54. Hajianfar, H.; Karimi, E.; Mollaghasemi, N.; Rezaei, S.; Arab, A. Is there a relationship between serum vitamin D and semen parameters? A cross-sectional sample of the Iranian infertile men. Basic Clin. Androl. 2021, 31, 29. [Google Scholar] [CrossRef] [PubMed]
  55. Hussein, T.M.; Eldabah, N.; Zayed, H.A.; Genedy, R.M. Assessment of serum vitamin D level and seminal vitamin D receptor gene methylation in a sample of Egyptian men with idiopathic infertility. Andrologia 2021, 53, e14172. [Google Scholar] [CrossRef]
  56. Kumari, S.; Singh, K.; Kumari, S.; Nishat, H.; Tiwary, B. Association of Vitamin D and Reproductive Hormones with Semen Parameters in Infertile Men. Cureus 2021, 13, e14511. [Google Scholar] [CrossRef] [PubMed]
  57. Ciccone, I.; Costa, E.; Pariz, J.; Teixeira, T.; Drevet, J.; Gharagozloo, P.; Aitken, R.; Hallak, J. Serum vitamin D content is associated with semen parameters and serum testosterone levels in men. Asian J. Androl. 2021, 23, 52–58. [Google Scholar] [CrossRef]
  58. Güngör, K.; Güngör, N.D.; Başar, M.M.; Cengiz, F.; Erşahin, S.S.; Çil, K. Relationship between serum vitamin D levels semen parameters and sperm DNA damage in men with unexplained infertility. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 499–505. [Google Scholar] [CrossRef]
  59. Holzer, M.; Massa, E.; Ghersevich, S. Relationship between serum vitamin D concentration and parameters of gonadal function in infertile male patients. Curr. Urol. 2022, in press. [CrossRef]
  60. Rezayat, A.A.; Asadpour, A.A.; Yarahmadi, A.; Ahmadnia, H.; Hakkak, A.M.; Soltani, S. Association Between Serum Vitamin D Concentration with Spermiogram Parameters and Reproductive Hormones Among Infertile Iranian Males: A Cross-sectional Study. Reprod. Sci. 2022, 29, 270–276. [Google Scholar] [CrossRef]
  61. Ramlau-Hansen, C.H.; Moeller, U.K.; Bonde, J.P.; Olsen, J.; Thulstrup, A.M. Are serum levels of vitamin D associated with semen quality? Results from a cross-sectional study in young healthy men. Fertil. Steril. 2011, 95, 1000–1004. [Google Scholar] [CrossRef]
  62. Neville, G.; Martyn, F.; Kilbane, M.; O’Riordan, M.; Wingfield, M.; McKenna, M.; McAuliffe, F.M. Vitamin D status and fertility outcomes during winter among couples undergoing in vitro fertilization/intracytoplasmic sperm injection. Int. J. Gynaecol. Obstet. 2016, 135, 172–176. [Google Scholar] [CrossRef]
  63. Zhu, C.L.; Xu, Q.F.; Li, S.X.; Wei, Y.C.; Zhu, G.C.; Yang, C.; Shi, Y.C. Investigation of serum vitamin D levels in Chinese infertile men. Andrologia 2016, 48, 1261–1266. [Google Scholar] [CrossRef]
  64. Józków, P.; Slowinska-Lisowska, M.; Zagrodna, A.; Medras, M.; Lwow, F. Vitamin D and semen quality in urban, young, healthy men (Androls). J. Mens. Health 2018, 14, 1–7. [Google Scholar] [CrossRef] [Green Version]
  65. Jueraitetibaike, K.; Ding, Z.; Wang, D.D.; Peng, L.P.; Jing, J.; Chen, L.; Ge, X.; Qiu, X.H.; Yao, B. The effect of vitamin D on sperm motility and the underlying mechanism. Asian J. Androl. 2019, 21, 400–407. [Google Scholar] [CrossRef]
  66. Rudnicka, A.; Adoamnei, E.; Noguera-Velasco, J.A.; Vioque, J.; Cañizares-Hernández, F.; Mendiola, J.; Jørgensen, N.; Chavarro, J.E.; Swan, S.H.; Torres-Cantero, A.M. Vitamin D status is not associated with reproductive parameters in young Spanish men. Andrology 2020, 8, 323–331. [Google Scholar] [CrossRef]
  67. Kamal, E.E.; Bakry, R.M.; Danyail, M.Y.; Badran, A.Y. Nonsignificant relation of seminal and serum vitamin D levels and semen parameters of males in Upper Egypt. Egypt. J. Dermatol. Venerol. 2022, 42, 27. [Google Scholar] [CrossRef]
  68. Alzoubi, A.; Mahdi, H.; Al Bashir, S.; Halalsheh, O.; Ebbini, M.A.; Alzarir, M.; Al-Ahmar, K.; Alfaqih, M.; Al-Hadidi, A.H. Normalization of Serum Vitamin D Improves Semen Motility Parameters in Patients with Idiopathic Male Infertility. Acta Endocrinol. (Bucharest Rom. 2005) 2017, 13, 180–187. [Google Scholar] [CrossRef]
  69. Wadhwa, L.; Priyadarshini, S.; Fauzdar, A.; Wadhwa, S.N.; Arora, S. Impact of Vitamin D Supplementation on Semen Quality in Vitamin D-Deficient Infertile Males with Oligoasthenozoospermia. J. Obstet. Gynaecol. India 2020, 70, 44–49. [Google Scholar] [CrossRef]
  70. Bartl, I.; Ondrušová, M.; Kužma, M.; Jackuliak, P.; Gažová, A.; Kyselovič, J.; Payer, J. Treatment With Cholecalciferol Leads to Increase Of Selected Semen Parameters in Young Infertile Males: Results of a 6-month Interventional Study. Physiol. Res. 2021, 70, S99–S107. [Google Scholar] [CrossRef]
  71. Amina Begum, M.; Ishrat, S.; Rani Saha, M.; Parveen, F.; Shah Alam, M.; Deeba, F.; Fatima, P.; Professor, A. Vitamin D Supplementation Improves Sperm Motility in Infertile Males with Asthenozoospermia: A Prospective Observational Study Scholars International Journal of Obstetrics and Gynecology Abbreviated Key Title: SchInt J ObstetGynec Vitamin D Supplementation Improves Sperm Motility in Infertile Males with Asthenozoospermia: A Prospective Observational Study. SchInt. J. Obs. 2021, 4, 331–336. [Google Scholar] [CrossRef]
  72. Maghsoumi-Norouzabad, L.; Zare Javid, A.; Mansoori, A.; Dadfar, M.; Serajian, A. The effects of Vitamin D3 supplementation on Spermatogram and endocrine factors in asthenozoospermia infertile men: A randomized, triple blind, placebo-controlled clinical trial. Reprod. Biol. Endocrinol. 2021, 19, 102. [Google Scholar] [CrossRef]
  73. Padmapriya, D.; Archana, R.; Sharma, N.; Vijayaraghavan, R. Effects of Vitamin D3 Supplementation in Male Reproductive Function: A Double-Blinded Randomized Controlled Trial. Curr. Top. Nutraceutical Res. 2022, 20, 334–339. [Google Scholar] [CrossRef]
  74. Blomberg Jensen, M.; Gerner Lawaetz, J.; Petersen, J.H.; Juul, A.; Jørgensen, N. Effects of Vitamin D Supplementation on Semen Quality, Reproductive Hormones, and Live Birth Rate: A Randomized Clinical Trial. J. Clin. Endocrinol. Metab. 2018, 103, 870–881. [Google Scholar] [CrossRef]
  75. Amini, L.; Mohammadbeigi, R.; Vafa, M.; Haghani, H.; Vahedian-Azimi, A.; Karimi, L.; Jahanfar, S.; Jamialahmadi, T.; Talebi, A.; Sahebkar, A. Evaluation of the effect of vitamin D3 supplementation on quantitative and qualitative parameters of spermograms and hormones in infertile men: A Randomized controlled trial. Complement. Ther. Med. 2020, 53, 102529. [Google Scholar] [CrossRef]
  76. Gheflati, A.; Mirjalili, S.A.M.; Kaviani, M.; Salehi-Abargouei, A.; Hosseini-Marnani, E.; Nadjarzadeh, A. Effects of Vitamin D Supplementation on Semen Quality and Reproductive Hormones in Patients with Asthenozoospermia: A Randomized Double-Blind Placebo-Controlled Clinical Trial. J. Nutr. Food Secur. 2021, 6, 31–42. [Google Scholar] [CrossRef]
  77. Chin, K.-Y.; Ima-Nirwana, S.; Wan Ngah, W.Z. Vitamin D is significantly associated with total testosterone and sex hormone-binding globulin in Malaysian men. Aging Male 2015, 18, 175–179. [Google Scholar] [CrossRef]
  78. Barbonetti, A.; Vassallo, M.R.C.; Felzani, G.; Francavilla, S.; Francavilla, F. Association between 25(OH)-vitamin D and testosterone levels: Evidence from men with chronic spinal cord injury. J. Spinal Cord Med. 2016, 39, 246–252. [Google Scholar] [CrossRef] [Green Version]
  79. Zhao, D.; Ouyang, P.; de Boer, I.H.; Lutsey, P.L.; Farag, Y.M.K.; Guallar, E.; Siscovick, D.S.; Post, W.S.; Kalyani, R.R.; Billups, K.L.; et al. Serum vitamin D and sex hormones levels in men and women: The Multi-Ethnic Study of Atherosclerosis (MESA). Maturitas 2017, 96, 95–102. [Google Scholar] [CrossRef] [Green Version]
  80. Książek, A.; Mędraś, M.; Zagrodna, A.; Słowińska-Lisowska, M.; Lwow, F. Correlative studies on vitamin D and total, free bioavailable testosterone levels in young, healthy men. Sci. Rep. 2021, 11, 20198. [Google Scholar] [CrossRef]
  81. Talebi, A.; Amini, L.; Sahebkar, A.; Paper, R. Association between Serum Vitamin D3 Levels and Sex Hormones in Infertile Men. J. Babol Univ. Med. Sci. 2022, 24, 88–94. [Google Scholar]
  82. Tak, Y.J.; Lee, J.G.; Kim, Y.J.; Park, N.C.; Kim, S.S.; Lee, S.; Cho, B.M.; Kong, E.H.; Jung, D.W.; Yi, Y.H. Serum 25-hydroxyvitamin D levels and testosterone deficiency in middle-aged Korean men: A cross-sectional study. Asian J. Androl. 2015, 17, 324. [Google Scholar] [CrossRef]
  83. Wang, N.; Han, B.; Li, Q.; Chen, Y.; Chen, Y.; Xia, F.; Lin, D.; Jensen, M.D.; Lu, Y. Vitamin D is associated with testosterone and hypogonadism in Chinese men: Results from a cross-sectional SPECT-China study. Reprod. Biol. Endocrinol. 2015, 13, 74. [Google Scholar] [CrossRef]
  84. Rafiq, R.; van Schoor, N.M.; Sohl, E.; Zillikens, M.C.; Oosterwerff, M.M.; Schaap, L.; Lips, P.; de Jongh, R.T. Associations of vitamin D status and vitamin D-related polymorphisms with sex hormones in older men. J. Steroid Biochem. Mol. Biol. 2016, 164, 11–17. [Google Scholar] [CrossRef] [PubMed]
  85. Anic, G.M.; Albanes, D.; Rohrmann, S.; Kanarek, N.; Nelson, W.G.; Bradwin, G.; Rifai, N.; McGlynn, K.A.; Platz, E.A.; Mondul, A.M. Association between serum 25-hydroxyvitamin D and serum sex steroid hormones among men in NHANES. Clin. Endocrinol. (Oxf.) 2016, 85, 258–266. [Google Scholar] [CrossRef] [Green Version]
  86. Lee, D.M.; Tajar, A.; Pye, S.R.; Boonen, S.; Vanderschueren, D.; Bouillon, R.; O’Neill, T.W.; Bartfai, G.; Casanueva, F.F.; Finn, J.D.; et al. Association of hypogonadism with vitamin D status: The European Male Ageing Study. Eur. J. Endocrinol. 2012, 166, 77–85. [Google Scholar] [CrossRef] [Green Version]
  87. Nimptsch, K.; Platz, E.A.; Willett, W.C.; Giovannucci, E. Association between plasma 25-OH vitamin D and testosterone levels in men. Clin. Endocrinol. (Oxf.) 2012, 77, 106–112. [Google Scholar] [CrossRef] [Green Version]
  88. Lerchbaum, E.; Pilz, S.; Trummer, C.; Rabe, T.; Schenk, M.; Heijboer, A.C.; Obermayer-Pietsch, B. Serum vitamin D levels and hypogonadism in men. Andrology 2014, 2, 748–754. [Google Scholar] [CrossRef]
  89. Heijboer, A.C.; Oosterwerff, M.; Schroten, N.F.; Eekhoff, E.M.W.; Chel, V.G.M.; De Boer, R.A.; Blankenstein, M.A.; Lips, P. Vitamin D supplementation and testosterone concentrations in male human subjects. Clin. Endocrinol. (Oxf.) 2015, 83, 105–110. [Google Scholar] [CrossRef]
  90. Lerchbaum, E.; Pilz, S.; Trummer, C.; Schwetz, V.; Pachernegg, O.; Heijboer, A.C.; Obermayer-Pietsch, B. Vitamin D and Testosterone in Healthy Men: A Randomized Controlled Trial. J. Clin. Endocrinol. Metab. 2017, 102, 4292–4302. [Google Scholar] [CrossRef] [Green Version]
  91. Lerchbaum, E.; Trummer, C.; Theiler-Schwetz, V.; Kollmann, M.; Wölfler, M.; Heijboer, A.C.; Pilz, S.; Obermayer-Pietsch, B. Effects of vitamin D supplementation on androgens in men with low testosterone levels: A randomized controlled trial. Eur. J. Nutr. 2019, 58, 3135–3146. [Google Scholar] [CrossRef] [Green Version]
  92. Ulrich, C.; Trojanowicz, B.; Fiedler, R.; Kraus, F.B.; Stangl, G.I.; Girndt, M.; Seibert, E. Serum Testosterone Levels Are Not Modified by Vitamin D Supplementation in Dialysis Patients and Healthy Subjects. Nephron 2021, 145, 481–485. [Google Scholar] [CrossRef]
  93. Canguven, O.; Talib, R.A.; El Ansari, W.; Yassin, D.J.; Al Naimi, A. Vitamin D treatment improves levels of sexual hormones, metabolic parameters and erectile function in middle-aged vitamin D deficient men. Aging Male 2017, 20, 9–16. [Google Scholar] [CrossRef]
  94. Pilz, S.; Frisch, S.; Koertke, H.; Kuhn, J.; Dreier, J.; Obermayer-Pietsch, B.; Wehr, E.; Zittermann, A. Effect of vitamin D supplementation on testosterone levels in men. Horm. Metab. Res. 2011, 43, 223–225. [Google Scholar] [CrossRef] [PubMed]
  95. Jorde, R.; Grimnes, G.; Hutchinson, M.; Kjærgaard, M.; Kamycheva, E.; Svartberg, J. Supplementation with vitamin D does not increase serum testosterone levels in healthy males. Horm. Metab. Res. 2013, 45, 675–681. [Google Scholar] [CrossRef] [PubMed]
  96. Zittermann, A.; Ernst, J.B.; Prokop, S.; Fuchs, U.; Dreier, J.; Kuhn, J.; Knabbe, C.; Berthold, H.K.; Gouni-Berthold, I.; Gummert, J.F.; et al. Vitamin D supplementation does not prevent the testosterone decline in males with advanced heart failure: The EVITA trial. Eur. J. Nutr. 2019, 58, 673–680. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, C.; Zhai, H.; Cheng, J.; Weng, P.; Chen, Y.; Li, Q.; Wang, C.; Xia, F.; Wang, N.; Lu, Y. Causal Link Between Vitamin D and Total Testosterone in Men: A Mendelian Randomization Analysis. J. Clin. Endocrinol. Metab. 2019, 104, 3148–3156. [Google Scholar] [CrossRef]
  98. Arab, A.; Hadi, A.; Moosavian, S.P.; Askari, G.; Nasirian, M. The association between serum vitamin D, fertility and semen quality: A systematic review and meta-analysis. Int. J. Surg. 2019, 71, 101–109. [Google Scholar] [CrossRef]
  99. Nolan, M.A.; Babcock, D.F.; Wennemuth, G.; Brown, W.; Burton, K.A.; McKnight, G.S. Sperm-specific protein kinase A catalytic subunit Calpha2 orchestrates cAMP signaling for male fertility. Proc. Natl. Acad. Sci. USA 2004, 101, 13483–13488. [Google Scholar] [CrossRef] [Green Version]
  100. Banks, N.; Sun, F.; Krawetz, S.A.; Coward, R.M.; Masson, P.; Smith, J.F.; Trussell, J.C.; Santoro, N.; Zhang, H.; Steiner, A.Z. Male vitamin D status and male factor infertility. Fertil. Steril. 2021, 116, 973–979. [Google Scholar] [CrossRef]
  101. Cui, X.; Pertile, R.; Eyles, D.W. The vitamin D receptor (VDR) binds to the nuclear matrix via its hinge domain: A potential mechanism for the reduction in VDR mediated transcription in mitotic cells. Mol. Cell. Endocrinol. 2018, 472, 18–25. [Google Scholar] [CrossRef]
  102. Merino, O.; Sánchez, R.; Gregorio, B.M.; Sampaio, F.J.; Risopatrón, J. Effects of Diet-Induced Obesity and Deficient in Vitamin D on Spermatozoa Function and DNA Integrity in Sprague-Dawley Rats. Biomed Res. Int. 2018, 2018, 5479057. [Google Scholar] [CrossRef]
  103. Skakkebaek, N.E.; Rajpert-De Meyts, E.; Buck Louis, G.M.; Toppari, J.; Andersson, A.M.; Eisenberg, M.L.; Jensen, T.K.; Jørgensen, N.; Swan, S.H.; Sapra, K.J.; et al. Male Reproductive Disorders and Fertility Trends: Influences of Environment and Genetic Susceptibility. Physiol. Rev. 2016, 96, 55–97. [Google Scholar] [CrossRef]
  104. Hofer, D.; Münzker, J.; Schwetz, V.; Ulbing, M.; Hutz, K.; Stiegler, P.; Zigeuner, R.; Pieber, T.R.; Müller, H.; Obermayer-Pietsch, B. Testicular synthesis and vitamin D action. J. Clin. Endocrinol. Metab. 2014, 99, 3766–3773. [Google Scholar] [CrossRef] [Green Version]
  105. de Angelis, C.; Galdiero, M.; Pivonello, C.; Garifalos, F.; Menafra, D.; Cariati, F.; Salzano, C.; Galdiero, G.; Piscopo, M.; Vece, A.; et al. The role of vitamin D in male fertility: A focus on the testis. Rev. Endocr. Metab. Disord. 2017, 18, 285–305. [Google Scholar] [CrossRef] [PubMed]
  106. Hosseini Marnani, E.; Mollahosseini, M.; Gheflati, A.; Ghadiri-Anari, A.; Nadjarzadeh, A. The effect of vitamin D supplementation on the androgenic profile in men: A systematic review and meta-analysis of clinical trials. Andrologia 2019, 51, e13343. [Google Scholar] [CrossRef] [PubMed]
  107. Oury, F.; Ferron, M.; Huizhen, W.; Confavreux, C.; Xu, L.; Lacombe, J.; Srinivas, P.; Chamouni, A.; Lugani, F.; Lejeune, H.; et al. Osteocalcin regulates murine and human fertility through a pancreas-bone-testis axis. J. Clin. Investig. 2013, 123, 2421–2433. [Google Scholar] [CrossRef] [PubMed]
  108. Krishnan, A.V.; Swami, S.; Peng, L.; Wang, J.; Moreno, J.; Feldman, D. Tissue-selective regulation of aromatase expression by calcitriol: Implications for breast cancer therapy. Endocrinology 2010, 151, 32–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Lorenzen, M.; Boisen, I.M.; Mortensen, L.J.; Lanske, B.; Juul, A.; Blomberg Jensen, M. Reproductive endocrinology of vitamin D. Mol. Cell. Endocrinol. 2017, 453, 103–112. [Google Scholar] [CrossRef]
  110. Zamani, A.; Saki, F.; Hatami, N.; Koohpeyma, F. Stereological assessment of the effects of vitamin D deficiency on the rat testis. BMC Endocr. Disord. 2020, 20, 162. [Google Scholar] [CrossRef]
  111. Sonnenberg, J.; Christakos, S.; Krey, L.C. 1,25-Dihydroxyvitamin D3 treatment results in increased choline acetyltransferase activity in specific brain nuclei. Endocrinology 1986, 118, 1433–1439. [Google Scholar] [CrossRef] [PubMed]
  112. Zanatta, L.; Zamoner, A.; Zanatta, A.P.; Bouraïma-Lelong, H.; Delalande, C.; Bois, C.; Carreau, S.; Silva, F.R.M.B. Nongenomic and genomic effects of 1α,25(OH)2 vitamin D3 in rat testis. Life Sci. 2011, 89, 515–523. [Google Scholar] [CrossRef]
  113. Merino, O.; Sánchez, R.; Gregorio, M.B.; Sampaio, F.; Risopatrón, J. Effect of high-fat and vitamin D deficient diet on rat sperm quality and fertility. Theriogenology 2019, 125, 6–11. [Google Scholar] [CrossRef]
  114. Xu, Y.; Lou, Y.; Kong, J. VDR regulates energy metabolism by modulating remodeling in adipose tissue. Eur. J. Pharmacol. 2019, 865, 172761. [Google Scholar] [CrossRef]
  115. Wang, L.; Lu, H.; Wang, S.; Liu, H.; Guo, M.; Bai, H.; Zeng, W.; Zhang, T. Vitamin D Receptor affects male mouse fertility via regulation of lipid metabolism and testosterone biosynthesis in testis. Gene 2022, 834, 146589. [Google Scholar] [CrossRef]
  116. Blomberg Jensen, M. Vitamin D and male reproduction. Nat. Rev. Endocrinol. 2014, 10, 175–186. [Google Scholar] [CrossRef]
  117. Albahrani, A.A.; Rotarou, V.; Roche, P.J.; Greaves, R.F. A simultaneous quantitative method for vitamins A, D and E in human serum using liquid chromatography-tandem mass spectrometry. J. Steroid Biochem. Mol. Biol. 2016, 159, 41–53. [Google Scholar] [CrossRef]
  118. Agmon-Levin, N.; Theodor, E.; Segal, R.M.; Shoenfeld, Y. Vitamin D in systemic and organ-specific autoimmune diseases. Clin. Rev. Allergy Immunol. 2013, 45, 256–266. [Google Scholar] [CrossRef]
  119. de Lorenzo, A.; Noce, A.; Moriconi, E.; Rampello, T.; Marrone, G.; di Daniele, N.; Rovella, V. MOSH Syndrome (Male Obesity Secondary Hypogonadism): Clinical Assessment and Possible Therapeutic Approaches. Nutrients 2018, 10, 474. [Google Scholar] [CrossRef] [Green Version]
  120. Santos, H.O.; Howell, S.; Nichols, K.; Teixeira, F.J. Reviewing the Evidence on Vitamin D Supplementation in the Management of Testosterone Status and Its Effects on Male Reproductive System (Testis and Prostate): Mechanistically Dazzling but Clinically Disappointing. Clin. Ther. 2020, 42, e101–e114. [Google Scholar] [CrossRef]
Figure 1. Overview of the vitamin D pathway. The main endogenous source of vitamin D is sunlight, which stimulates the synthesis of previtamin D3. Vitamin D can also be absorbed in the intestines from food. From the skin or intestines, vitamin D goes into the blood where it is bound to protein. It is then hydroxylated in the liver and kidneys to produce 1,25-dihydroxyvitamin D, a physiologically active form of vitamin D that functions in many parts of the body, including the skeletal system, immune system, reproductive system, and others. Abbreviations: CYP: cytochrome P-450; UVB: ultraviolet B; VDBP: vitamin D-binding protein.
Figure 1. Overview of the vitamin D pathway. The main endogenous source of vitamin D is sunlight, which stimulates the synthesis of previtamin D3. Vitamin D can also be absorbed in the intestines from food. From the skin or intestines, vitamin D goes into the blood where it is bound to protein. It is then hydroxylated in the liver and kidneys to produce 1,25-dihydroxyvitamin D, a physiologically active form of vitamin D that functions in many parts of the body, including the skeletal system, immune system, reproductive system, and others. Abbreviations: CYP: cytochrome P-450; UVB: ultraviolet B; VDBP: vitamin D-binding protein.
Biomedicines 11 00090 g001
Figure 2. PRISMA flow chart of the literature search and screening process.
Figure 2. PRISMA flow chart of the literature search and screening process.
Biomedicines 11 00090 g002
Table 5. Summary table of major conclusions.
Table 5. Summary table of major conclusions.
Hypothesis SupportedNot Supported
VD has a positive effect on sperm motilityobservational studies[35,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60][61,62,63,64,65,66,67]
interventional studies[68,69,70,71,72,73][74,75,76]
VD increases sperm concentrationobservational studies[46,52,53,54,55,56,57,58,60][35,48,49,50,51,59,61,62,63,64,65,66,67]
interventional studies[69,70,71,73][68,72,74,75,76]
VD improves sperm morphologyobservational studies[35,51,52,53,54,56,57,58][46,48,49,50,55,59,60,61,62,65,67]
interventional studies[70][68,72,73,74,75,76]
VD reduces sperm DNA fragmentationobservational studies[53,58][51]
interventional studiesNaNa
VD positively influences testosterone levelsobservational studies[52,57,60,78,82,83,84,85,87,97][46,48,59,61,77,80,81,86,88]
interventional studies[94][70,72,75,76,91,92,93,95,96]
VD affects the bioavailability of androgens by increasing the level of SHBGobservational studies[48,59,61,77,79,85][46,49,50,79,80,83,84,86,88]
interventional studiesNa[70,72,75,76,89,90,91,95,96]
Abbreviations: Na: not applicable; SHBG: sex hormone binding globulin; VD: vitamin D.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Adamczewska, D.; Słowikowska-Hilczer, J.; Walczak-Jędrzejowska, R. The Association between Vitamin D and the Components of Male Fertility: A Systematic Review. Biomedicines 2023, 11, 90. https://doi.org/10.3390/biomedicines11010090

AMA Style

Adamczewska D, Słowikowska-Hilczer J, Walczak-Jędrzejowska R. The Association between Vitamin D and the Components of Male Fertility: A Systematic Review. Biomedicines. 2023; 11(1):90. https://doi.org/10.3390/biomedicines11010090

Chicago/Turabian Style

Adamczewska, Daria, Jolanta Słowikowska-Hilczer, and Renata Walczak-Jędrzejowska. 2023. "The Association between Vitamin D and the Components of Male Fertility: A Systematic Review" Biomedicines 11, no. 1: 90. https://doi.org/10.3390/biomedicines11010090

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop