Next Article in Journal
Influence of Psychological Factors on Vaccination Acceptance among Health Care Workers in Slovenia in Three Different Phases of the COVID-19 Pandemic
Previous Article in Journal
A Mathematical Model of Vaccinations Using New Fractional Order Derivative
Previous Article in Special Issue
Immune Persistence against SARS-CoV-2 after Primary and Booster Immunization in Humans: A Large-Scale Prospective Cohort Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effect of Long COVID-19 Infection and Vaccination on Male Fertility; A Narrative Review

by
Soheila Pourmasumi
1,2,
Alireza Nazari
1,3,
Zahra Ahmadi
4,
Sophia N. Kouni
5,
Cesare de Gregorio
6,
Ioanna Koniari
7,
Periklis Dousdampanis
8,
Virginia Mplani
9,
Panagiotis Plotas
10,
Stelios Assimakopoulos
11,
Christos Gogos
12,
Georgios Aidonisdis
13,
Pavlos Roditis
14,
Nikos Matsas
15,
Dimitrios Velissaris
16,
Gianfranco Calogiuri
17,18,
Ming-Yow Hung
19,20,21,
Servet Altay
22 and
Nicholas G. Kounis
23,*
1
Social Determinants of Health Research Center, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
2
Clinical Research Development Unit, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
3
Department of Surgery, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
4
Pistachio Safety Research Center, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
5
Speech Therapy Practice, 26221 Patras, Greece
6
Department of Clinical and Experimental Medicine, University of Messina Medical School, 98122 Messina, Italy
7
Department of Internal Medicine, Division of Cardiology, University Hospital of South Manchester NHS Foundation Trust, Manchester M23 9LT, UK
8
Department of Nephrology, Saint Andrews State General Hospital, 26221 Patras, Greece
9
Intensive Care Unit, Patras University Hospital, 26500 Patras, Greece
10
Department of Speech Therapy, University of Patras, 26500 Patras, Greece
11
Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, 26500 Patras, Greece
12
COVID-19 Unit, Papageorgiou General Hospital, 56403 Thessaloniki, Greece
13
Interbalkan Medical Center, 57001 Thessaloniki, Greece
14
Department of Cardiology, Mamatsio Kozanis General Hospital, 50100 Kozani, Greece
15
Cardiology Private Practice, 30131 Agrinion, Greece
16
Department of Internal Medicine, University of Patras, 26500 Patras, Greece
17
Pneumonology Department, Civil Hospital “Ninetto Melli”, Pietro Vernoti, 72027 Brindisi, Italy
18
Department of Internal Medicine, Immunology and Infectious Diseases, Section of Allergology and Clinical Immunology, University of Bari Medical School, 70121 Bari, Italy
19
Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
20
Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
21
Taipei Heart Institute, Taipei Medical University, Taipei 110, Taiwan
22
Department of Cardiology, Faculty of Medicine Trakya University, Edirne 22030, Turkey
23
Department of Internal Medicine, Division of Cardiology, University of Patras Medical School, 26500 Patras, Greece
*
Author to whom correspondence should be addressed.
Vaccines 2022, 10(12), 1982; https://doi.org/10.3390/vaccines10121982
Submission received: 28 October 2022 / Revised: 14 November 2022 / Accepted: 17 November 2022 / Published: 22 November 2022
(This article belongs to the Special Issue Immune Response against SARS-CoV-2)

Abstract

:
Earlier research has suggested that the male reproductive system could be particularly vulnerable to SARS-CoV-2 (COVID-19) infection, and infections involving this novel disease not only pose serious health threats but could also cause male infertility. Data from multi-organ research during the recent outbreak indicate that male infertility might not be diagnosed as a possible consequence of COVID-19 infection. Several review papers have summarized the etiology factors on male fertility, but to date no review paper has been published defining the effect of COVID-19 infection on male fertility. Therefore, the aim of this study is to review the published scientific evidence regarding male fertility potential, the risk of infertility during the COVID-19 pandemic, and the impact of COVID-19 vaccination on the male reproductive system. The effects of COVID-19 infection and the subsequent vaccination on seminal fluid, sperm count, sperm motility, sperm morphology, sperm viability, testes and sex hormones are particularly reviewed.

1. Introduction

Infertility constitutes an important problem for families around the world [1,2]. Infertility can create social, economic and psychological distress to infertile patients, also affecting the economic burden ofhealth and clinical systems [3,4]. Based on the World Health Organization (WHO) definition, when couples have unprotected sexual intercourse for at least 1 year and pregnancy does not occur, they are categorized in the infertility group [5]. It is estimated that about 8–12% of couples suffer from infertility problems [6]. Infertility is due to male and female disorders and both genders are involved in infertility. It is estimated that in all infertility cases worldwide, about 50% of infertility is related to male factors and 50% to female factors [7]. A Global Burden of Disease (GBD) analysis reported that over the last three decades, infertility prevalence has increased annually in both men and women (∼0.3% vs. 0.4% respectively) [8].
Male infertility can result from three main parts: (A) testicular part, (B) ejaculated part and (C) hormonal part. Approximately, 50% of all male infertility is caused by abnormal sperm production (defects in spermatogenesis process) and defective sperm parameters (count, motility and morphology) [9,10]. These parts can be affected by several factors, including systematic disease, infections, medications, occupational parameters, psychological factors, lifestyle, etc. Consequently, these factors can have negative effects on male reproductive potential [11].
Since 2020, COVID-19 has rapidly spread across the world and infected hundreds of millions of people globally. This was an unknown virus and the experience of the scientific community concerning both short-term and long-term complications of COVID-19 infection continues to improve [12]. Laboratory assessments confirmed that COVID-19 has 46 different varieties and is detectable in human and animal body tissue and secretions [13].
COVID-19 infection can induce a variety of manifestations in the human body via the attack on respiratory, cardiac, immunology, digestive, urinary and neural systems. The effects of COVID-19 on fertility potential at the start of virus spread were unexpected and mysterious. In the second peak, several studies were designed to determine the effects of COVID-19 on fertility, confirming that this virus can also affect male fertility [14].
When the human body is infected by a virus, infection might further induce immune system response against the virus. An important signof infection is spiked temperature (>38 °C), which can have further harmful effects on the male reproductive system andsperm quality. It has been established that 4 to 11 weeks post fever, the quality of sperm decreases and the number of impaired sperm in seminal fluid increases. Based on scientific reports, three mechanisms may be responsible for negative COVID-19 effects on the male reproductive system: First, high testis temperature post fever is a defective factor for germ cell lines (spermatogonia cell line); Second, fever can affect other cells in the testis and seminiferous tubules (Sertoli and Leydig cells) and consequently create defects in exocrine and endocrine functions; Third, fever can decrease accessory gland secretion and this might be harmful for sperm viability and quality [15,16].
Both human genders are susceptible to COVID-19 infection, but it has been shown that men are at higher risk of infection, and the rate of infected men with COVID-19 is higher than in women [17].
Hence, infertility and its treatment have been affected during the COVID-19 pandemic, as in the first peak there was a lack of knowledge about the COVID-19 transmission process and its appearance in human reproductive secretions; and also, in different countries, infertility clinical centers were completely closed.
A study reported that COVID-19 infection was harmful for reproductive potential in male patients, affecting sex hormone level, testicular integrity and seminal quality [18].
After the COVID-19 pandemic and the death of millions of people around the world, serious efforts were made globally to produce a vaccine against COVID-19 [19]. For the first time, a Pfizer vaccine received emergency use approval by the Food and Drug Administration (FDA) and was rapidly administered for the reduction of infection severity [20]. The safety of the COVID-19 vaccine was identified by several laboratory tests, and the clinical and self-reports of cases who received both the first and second dose of the vaccine were evaluated [21,22].
In spite of no confirmed reports on the results of the vaccine on human physiology systems, there was public fear about the potential adverse effects of the vaccine on both the male and female reproductive systems [23]. Based on several fertility and reproductive reports, COVID-19 vaccines demonstrate no harmful effects on fertility, but to date many reproductive-age people are reluctant to receive the vaccine [24,25].
Therefore, in this study, we aimed to review scientific evidence about male fertility potential, the risk of infertility during the COVID-19 pandemic, and the effects of COVID-19 vaccination on the male reproductive system. Given the global burden of the COVID-19 pandemic, there is tremendous interest in understanding the potential health disparities in the offspring of men with a history of COVID-19. The effects of COVID-19 infection on male fertility are shown in Figure 1.

2. Search Methods

We searched related keywords in Medline (PubMed) based on the study purpose. The keywords were SARS-CoV-2 OR COVID-19, AND male infertility, OR sperm, OR sex hormone, AND testis OR orchitis, AND seminal fluid, OR fertility potential, AND COVID-19 vaccine OR SARS-CoV-2 vaccination. Studies reporting the effect of either SARS-CoV2 infection or COVID-19 vaccination on qualitative factors such as demographic parameters and quantitative markers, such as seminal fluid quality, sperm count, sperm motility, sperm morphology elements, sperm viability, testicular and sex hormone function, were analyzed.

3. SARS-CoV2

SARS-CoV-2 caused the COVID-19 disease pandemic at the end of 2019 [26]. There are several symptoms associated with COVID-19 infection, including a cough, gut dysfunction, shortness of breath, and fever. Furthermore, 80% of patients after infection with SARS-CoV-2 reported approximately mild symptoms, 5% of them severe, and death can occur in 3% of cases [27]. The COVID-19 epidemic has been associated with infertility [28]. Recent reports have confirmed that the COVID-19 virus is harmful for sperm parameters, sex hormone levels and fertility potential. There is considerable evidence reporting over 25 viruses, such as Herpes Simplex Virus (HSV) and Human Immunodeficiency Virus (HIV), that can pass the genital tract and negatively impact spermatogenesis [29,30,31].

Possible Mechanisms of SARS-CoV-2 Affecting Male Reproduction

How can a virus cause damage to the reproductive system? This is an important scientific question and several hypotheses have been raised to answer this question. The main proposed mechanisms by which a virus impairs fertility include: (I) The virus can attack the spermatogonia germ cell line directly and impair sperm production; (II) The virus can spread in reproductive fluids and be transmitted by sexual intercourse; (III) The virus can induce inflammation in the reproductive tract. This inflammation can cause tissue damage, cell damage and sex hormone secretion damage in testis; (IV) The virus can cause male accessory gland infection, where secretion is an important substance to maintain male fertility potential; (V) The virus is a cause of infection-induced inflammation response and indirectly exerts negative effects on male fertility via the elevation of oxidative stress in the human body and the increase of reactive oxygen species; (VI) Finally, virus infection can trigger fever, and high body temperature could prove harmful for the reproductive system [32]. In conclusion, the inflammation chain can create damage to the body, and perhaps some of these mechanisms overlap.

4. COVID-19 Vaccination and Male Reproductive Potential

At the present time, limited studies have assessed the effects of the COVID-19 vaccine on male fertility factors including sperm parameters, hormonal level and testis impairment. In the scientific reports, researchers have confirmed that vaccination against COVID-19 did not exert harmful effects on male fertility. In a recent study, Adamyan et al. assessed two prior vaccines [33] (Pfizer-BioNTech and Moderna) on sperm parameters before and after the first and second vaccination dose in reproductive-age individuals. They reported no significant reduction in sperm analysis parameters. Olana et al. evaluated the effects of COVID-19 vaccination on male fertility parameters. In this study, no significant differences in spermatozoa parameters before and after vaccine inoculations were found. Furthermore, oxidative stress analysis, the activity of the cell membrane, and IL-6, as a marker of inflammation, were not increased in human seminal fluid by the mRNA COVID-19 vaccine [34]. Zhu et al. assessed COVID-19 vaccination impact on sperm count and motility. The study showed that there was no decrease in sperm count and volume post COVID-19 vaccination in comparison with the respective values before vaccination [35]. Barda et al. studied the relation between first and second dose of the vaccination and sperm concentration. Sperm concentration count was significantly increased after the second dose of the vaccine [36]. Chatzimeletiou et al. investigated the effects of the COVID-19 vaccine on sperm parameters in ankylosing spondylitis patients., revealing that the sperm count was significantly increased from the first to the third vaccine dose [37]. Olana et al. evaluated the effects of COVID-19 vaccination on male fertility parameters, reporting that there was no significant difference in oxidative stress level and sperm motility before and after vaccination [34]. In another study, Zhu reported that COVID-19 vaccination has no negative effects on sperm motility [35]. In a study with patients suffering from ankylosing spondylitis, COVID-19 vaccination could improve sperm motility after the second dose [37]. Kumar and his colleagues, in their study, demonstrated that there was no scientific evidence to confirm the impairment effects of COVID-19 vaccination on sperm parameters [38]. Sperm morphology was assessed in ankylosing spondylitis patients by Chatzimeletiou et al. as well. They reported that COVID-19 vaccination had no negative effects on sperm morphology; although the shape of sperm in ankylosing spondylitis individuals showed more defects, reporting that vaccination can exert positive effects on sperm morphology [37]. The effects of COVID-19 vaccination on testis tissue were assessed by Carto et al. [39]. They investigated orchitis and epididymitis in vaccinated and non-vaccinated cases. Their results showed that the rates of orchitis and epididymitis were significantly lower in vaccinated cases compared to non-vaccinated ones, suggesting that a single dose of COVID-19 vaccine can significantly decrease the risk of inflammation in testis.
According to a recent report, COVID-19 vaccinations are safe for the male reproductive system despite the fact that post the second dose about 15% of vaccinated individuals showed mild fever as a clinical symptom [40]. Consequently, vaccination was not able to promote a severe inflammatory response in human reproductive system. To date, no scientific data have been presented to support probability of testis tissue damage and sperm impairment after vaccination. Moreover, in a data analysis from an internet-based pregnancy study that follows U.S. and Canadian couples trying to conceive over time [41], the researchers followed more than 2000 women, 21–45 years old, from December 2020 to November 2021. All were trying to conceive without fertility treatments. Participants answered questions on income, education, lifestyle, and reproductive and medical histories. The latter included questions about COVID-19 vaccine status and positive SARS-CoV-2 tests. The women also provided information about their partners’ vaccine status and positive tests. Male partners aged more than 21 years answered similar questions. Female partners completed follow-up questionnaires every 8 weeks for up to 12 months or until they became pregnant. It was revealed that the chances of conception did not change with either partner’s vaccination status. The researchers adjusted for several factors that could influence the results. These included vaccine type, nationality, occupation, and history of infertility. The final conclusion was that the COVID-19 vaccination in either partner is unrelated to fertility among couples.

5. COVID-19 in Seminal Fluid

Major concerns about the impact of SARS-CoV-2 infection on male fertility have increased, and one of the important questions is the present probability of COVID-19 in the seminal fluid of infected individuals. For the detection of virus co-expression, both angiotensin I-converting enzyme 2 (ACE2) and transmembrane serine protease TMPRSS2 were required in the testis of COVID-19 individuals, raising concerns regarding the virus entrance from the blood–testis barrier and the presence of COVID-19 in testis tissue and seminal fluid. To date, different reports regarding the controversy of the presence of COVID-19 in seminal fluid have been published.
Some studies have reported the significant impact of COVID-19 infection on seminal fluid quality [42,43].
A report by Li et al. demonstrated that semen can shed the virus during the recovery phase, even though COVID-19 does not affect the semen of all men who have the virus. The seminal fluid of 15 cases with severe infection was tested and COVID-19 was detected in about 27% of patients [44].
Gacci et al. [43] showed that after recovery from COVID-19, 25 out of 43 male patients were diagnosed as oligocrypto-azoospermic, while 8 and 3 patients revealed azoospermia and oligospermia, respectively. Furthermore, IL-8 was found in the semen fluid of 76.7% of patients [43].
In a study published in 2020, researchers suggested that COVID-19 may be detectable in acute infected patients after the treatment and recovery period. They also discussed that COVID-19 is detectable in patients who have no infection symptoms [45]. COVID-19-infected patients demonstrated common infection symptoms including fever and inflammation. Fever is an important parameter that has negative effects on seminal fluid and quality of sperm. Low seminal and sperm quality post severe fever seem to continue for at least 90 days [46]. Furthermore, inflammation can activate the immune system and consequently the seminiferous tubules in testis affected by COVID-19, representing a main factor of seminal and sperm quality impairment [47].
In contrast to the above reports, several studies confirmed that there was no COVID-19 virus detection in the seminal fluid of infected patients. For example, Song et al. reported that the presence of COVID-19 in the seminal fluid of male patients post recovery could not be identified [48]. Furthermore, in another study, the seminal fluid of 28 infected individuals was tested and there was no RNA of COVID-19 in the seminal fluid from 8 to 54 days after contamination [49].
In two separate studies, a seminal fluid test and testicular biopsy by real time-polymerase chain reaction (PCR) method reported no presence of COVID-19 in infected patients’ seminal fluid and testis samples [50,51]. Pan et al. demonstrated that there was no COVID-19 in semen fluids 31 days post confirmation of infection with COVID-19 [52]. In another study, Ma and his colleagues evaluated the seminal fluid of 12 patients post COVID-19 infection, and data showed that there was no viral infection in the seminal fluid [53].

5.1. Sperm Count

Recently, Hu et al. assessed the effects of COVID-19 infection on sperm quality in recovered patients compared to healthy cases [54]. The results of their study showed that COVID-19 can significantly decrease sperm count in comparison with the control group. It is also reported that sperm count in recovered patients’ after 150 days was similar to the control group. So, at least 5 months recovery time is needed for sperm count to return to normal. Finally, they concluded that the recovery time in COVID-19 patients plays an important role in increasing sperm quality, especially sperm count. Therefore, COVID-19-infected patients after treatment must receive supplemental care to improve sperm count. In another study, the seminal samples of COVID-19-infected patients after recovery were analyzed and compared with healthy men. The results revealed that sperm count was significantly lower in recovered patients than in the control group [55]. Segars et al. demonstrated that the fertility potential is decreased in COVID-19-infected patients and sperm analysis showed reduced sperm count 3 months post COVID-19 infection [56]. Holtmann et al. in their study, evaluated the effects of COVID-19 on sperm parameters, demonstrating that in COVID-19-infected patients, sperm concentration is significantly decreased [49]. In a study published in 2022, sperm count was assessed in COVID-19-recovered patients and there was a significantly decreased sperm count in patients 1 month post recovery in comparison with 3 months, revealing that recovery time is an important factor for sperm count improvement post COVID-19 infection [57].
Guo et al. in 2021 confirmed that sperm count was significantly decreased in COVID-19-infected patients, suggesting that a direct effect of COVID-19 on spermatogenesis may be responsible for sperm count decrease in COVID-19 patients [58]. A study by Li et al. reported that sperm count was decreased in COVID-19 cases, suggesting that elevation of immunological factors, including interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), might be responsible for differences in sperm count between COVID-19-infected patients and healthy control individuals [59]. Additionally, another study concluded that the elevation of sperm DNA fragmentation is likely the cause of sperm count decrease in COVID-19-infected patients [53].

5.2. Sperm Motility

Several studies investigated the effects of COVID-19 infection on sperm motility and reported decreased sperm motility in patients compared to healthy individuals [56].
Assessing sperm parameters in COVID-19-infected men, Piroozmanesh et al. demonstrated that sperm motility was significantly decreased [60]. Holtman et al. in their study, confirmed a sperm motility decrease in COVID-19 moderately infected patients [49]. Similarly, Li et al. reported that SARS-CoV2 infection can have negative effects on sperm mobility in fertile infected individuals [59]. Recently, Donders et al. reported significantly decreased sperm motility immediately after COVID-19, which was reversed and finally increased 3 months post infection compared to 1 month [57].
Interestingly, in contrast to the above-mentioned papers, Guo et al. analyzed the sperm quality of COVID-19-infected patients and reported that among 23 patients, only in 2 cases was the sperm motility decreased, revealing that in their study population COVID-19 had no negative effect on sperm motility [58]. He et al. in 2021 established that sperm motility was influenced by infection. They reported that sperm motility was significantly decreased during moderate infection in comparison with mild infection or healthy control individuals [61]. The elevation of oxidative stress and damage to sperm DNA are other factors for decreased sperm motility in viral infection cases [62].

5.3. Sperm Morphology

Sperm morphology is a vital parameter in the sperm analysis process. Sperm morphology, concerning the size and shape of sperm, is one factor that is examined as part of a semen analysis to evaluate male infertility. Changes in sperm shape may have negative effect on sperm motility, viability and fertility potential. Several factors can generate defects in sperm morphology. Infection is one of these factors. After the COVID-19 pandemic, an important question about whether COVID-19 is a defective factor for sperm morphology has arisen. A recent study investigated the effects of COVID-19 on sperm morphology in three groups, namely infected patients before and after treatment and healthy control males. In the infected patients before treatment, sperm morphology was significantly decreased in comparison with the control group. Furthermore, in treated individuals, sperm morphology was impaired compared to the control group. They concluded that a continuous fever during COVID-19 was an aggravating factor for sperm morphology [63]. A study by Ma et al. demonstrated that the normal morphology of sperm in COVID-19 patients is significantly decreased [53]. In 2021, Falahieh et al. assessed sperm morphology in treated COVID-19 cases after 3 months. The results of their study showed that the percentage of normal sperm morphology increased 3 months after COVID-19 treatment, but based on WHO guidelines, the rate of normal morphology was lower than standard [64]. Maleki et al. showed that sperm morphology can be decreased after SARS-CoV2 infection [65]. Koc and his colleagues [42] investigated sperm parameters of 21 infertile men who were referred for infertility treatment. The results of COVID-19 tests in these cases were positive without requiring hospitalization. This study showed that normal sperm morphology was significantly decreased.
Since COVID-19 can activate several pathways throughout inflammatory responses, it can induce further oxidative stress. This oxidative stress can cause peroxidative impairment to the plasma membrane of sperm. This process can create defects in sperm chromatin and DNA integrity, which leads to abnormal sperm morphology [66]. On the other hand, fever associated with infection constitutes a defective factor for sperm morphology. Other researchers suggested that the level of fever (mild/medium/severe) and its duration (number of days) are two main parameters that can significantly decrease normal sperm morphology parameters [62]. Two separate studies confirmed that high fever levels demonstrated a negative correlation with normal sperm morphology [67,68]. It is speculated that the decrease in normal sperm morphology during COVID-19 can be attributed to fever levels.

5.4. Sperm Viability

Sperm viability in COVID-19 infection was investigated in some studies. The results of studies confirmed that sperm viability was decreased in COVID-19 patients [55]. Falahieh et al. assessed sperm analysis of 20 COVID-19 patients 2 weeks and 3 months post infection. They found that sperm viability had no significant difference between two investigation time frames [64]. However, Piroozmanesh and his colleagues compared sperm viability in infected patients and healthy control men, showing that sperm viability was significantly decreased in COVID-19 patients [60].
Several mechanisms have been proposed for the negative effects of COVID-19 on sperm viability. Firstly, there is a hypothesis that influenza SARS-CoV-2 viruses cause oxidative stress that negatively affects sperm viability [69]. Secondly, elevation in inflammatory factors, including cytokines, TNF-α, and interleukin-2, 6, can induce lipid peroxidation in sperm, leading tosperm viability damage [70]. Marin et al. have suggested that COVID-19 can induce ACE2 production, and this is a negative factor for sperm viability in COVID-19 patients [71]. Based on Kopper’s study, COVID-19 may induce apoptosis and lead to sperm viability impairment [72].

6. Testicular Changes

Several studies have demonstrated testicular damage and increased orchitis attacks following COVID-19 [73,74]. The function of gonads in SARS-CoV2-infected patients was found to be significantly impaired compared to controls [53]. Bian et al. evaluated testis tissue in COVID-19 patients, revealing considerable damage in seminiferous tubules and a decrease in spermatogenic cell lines [75]. La et al. [76] reported testicular pain in fertile men after COVID-19 infection in both testes, being more severe in the left testis. Li et al. [59] confirmed pathological changes in 6 of 12 patients following COVID-19, where 11 of these patients suffered from orchitis. According to Holtman et al. a mild COVID-19 infection induced the impairment of testicular function [49], whereas a moderate severity infection affected the semen parameters. In contrast to the above-mentioned studies, Song et al. tested testis tissue from men who died from COVID-19 infection, demonstrating no COVID-19-positive cells in testis tissue [48].
Other researchers demonstrated that infections of COVID-19 could have an indirect impact on seminiferous function by affecting the hypothalamic–pituitary–testis axis through immunological and inflammatory responses [77,78]. Xu et al. demonstrated that COVID-19-affected patients showed an increased number of leukocytes infiltrating into the testes, accompanied by an atrophic seminiferous epithelium [73]. The possibility of blood–testis barrier (BTB) breakdown has thus been speculated for COVID-19 patients but still requires experimental validation [79]. Aziz et al. confirmed that COVID-19 can induce immune response and could damage the testis tissue and spermatogenic function due to immune response released mediators [80]. Based on previous observations, COVID-19 can affect CD14 in testes via spermatogonia stem cells [81,82]. A study evaluated that the infiltration and degeneration of CD3+ and CD68+ immune cells in spermatogenic cells can lead to testicular inflammation after COVID-19. Yang et al. showed that viral orchitis was associated with T lymphocyte invasion into the testicular parenchyma in 12 dead patients infected with COVID-19, causing remarkable reproductive tubular damage. They reported that infection with COVID-19 caused histopathological changes in postmortem testicular tissues [83]. This implies that sperm function may be affected by systemic inflammation that leads to increased oxidative stress. During viral infections, fever can impair spermatogenesis, while SARS-CoV2 infection causes testicular apoptosis [84] and disrupts spermatogenesis in general [62]. IgG precipitates were seen in testes infected with COVID-19, and a secondary autoimmune response of extensive IgG production in Sertoli cells and germ cells caused testicular dysfunction [73]. Additionally, IgG has been linked to autoimmune orchitis [85], which might trigger the host’s immune system and cause antibodies to be incorporated into semen fluid [86].
In another study, the researchers hypothesized that an elevation in body temperature is a defective factor for male reproductive tissues [87]. Some data show that afebrile episode due to the viral infection can affect the quality of sperm [88]. This may result in reduced fertility due to testicular damage following COVID-19.
Other mechanisms are linked to ACE2. Changes in ACE2 during infection can damage reproductive tissues. In a paper by Reis et al. [89], ACE2 was reported in reproductive tissues including testis, prostate and epididymis. This is an important factor for male puberty and healthy reproductive function [89]. Multiple reports have demonstrated that COVID-19 causes orchitis, mild lymphocytosis, decreased Leydig cell counts, and the destruction of seminiferous tubules in the testis [90,91,92]. In other words, high ACE2 expression in the testis may facilitate the entrance and colonization of virus, which can have adverse effects on male reproductive functioning [93]. ACE2 is predominantly expressed in the Sertoli and Leydig cells at the transcriptome level in adult human testis [94]. A transcriptome analysis study of human testicular tissues has demonstrated that the TMPRSS2 and ACE2 receptors are prominently expressed in Sertoli cells, Leydig cells, spermatids, and spermatogonia [95].

7. Sex Hormones

The negative effects of the viral infection on sex hormones have been validated in several studies [49,96]. In a male hormonal profile, it was confirmed that testosterone is an important hormone in the male reproductive system. Any changes in testosterone level are harmful for male fertility. Decreased levels of testosterone can impair spermatogenesis and fertility potential [97]. Holtmann et al. reported that in COVID-19 patients, the level of sex hormones was decreased significantly [49]. Moreover, Pozzilli et al. [47] revealed that there is a negative correlation between the risk of COVID-19 and the level of the testosterone hormone. The level of testosterone and LH or follicle-stimulating hormone (FSH) is another factor for the diagnosis of male fertility potential. An imbalance in the testosterone/LH/FSH ratio prognosticates defects in male fertility during SARS-CoV2 infection. Furthermore, impaired hypothalamic–pituitary–gonadal axis activity can change hormonal levels and be harmful for healthy reproductive function [98]. Sex hormone concentration was evaluated in 12 patients after infection with COVID-19, reporting a decreased level of testosterone/LH and increased level of LH compared to the controls [53]. Temiz and his colleagues investigated sex hormones in three groups (patients with COVID-19 before and after treatment, and a control group of healthy men). They reported that COVID-19-infected patients before treatment showed a significant decreased level of hormones (testosterone, FSH and LH) in comparison with the control group. Comparison between pre- and post-treatment levels showed that the level of hormones was elevated after treatment, similar to the control group. They concluded that COVID-19 infection has no continuous negative effects on male sex hormones [63]. In the study of Ma et al. the hormonal profile of COVID-19-infected cases was assessed and compared with a healthy control group. The results showed that the level of the LH hormone was significantly higher in COVID-19 cases than controls. Additionally, the testosterone/LH ratio was lower in COVID-19 patients compared to the control group [99]. In another study [100], testosterone levels were decreased, and the LH levels were increased. Consequently, this is an important effect that could impair the balance of the testosterone/LH/FSH ratio. Schroeder et al. [101] found that in COVID-19 men, the levels of FSH and LH were increased, and the level of testosterone decreased. They suggested that these changes in sex hormone levels during the COVID-19 pandemic were due to the activation of the immune system and a down-regulation of hormone production. Interestingly, Cayan S. et al. demonstrated that the average concentration of LH and FSH post COVID-19 infection increases and that the increased levels could be attributed to gonadotropin cell production as a result of early inflammation [102].
The main mechanism that affects hormonal changes during COVID-19 is inflammation in reproductive tissues and the immune response to inflammation. Most androgens are produced by the Leydig cells [103]. Leydig cells were reduced remarkably in 12 patients with COVID-19. It has recently been reported that hypogonadotropic hypogonadism in COVID-19 patients may be related to the replication of COVID-19 in Leydig cells. A recent study by Rastrelli et al. [97] suggests that novel COVID-19 may impair the secretory function of Leydig cells. Moreover, they demonstrated low levels of total dihydrotestosterone in patients with severe COVID-19, speculating that SARS-CoV2 infection may have been related to enhanced inflammation or a cytokine storm, which can lead to decreased testosterone levels [83,97]. A study published in 2019 by Guan X. et al. revealed that testosterone is necessary to preserve male reproductive function, as well as to support the maturation of Sertoli cell and Leydig cell development [104]. It has been reported that TMPRSS2 expression is increased in men upon exposure to androgens [95]. These changes are thought to be the result of Leydig cell damage and the impairment of the steroidogenic pathways [105]. Additionally, psychological stress is an important factor related to elevated levels of oxidative stress in the human body. Increased levels of OS have negative effects on the male reproductive system and can prevent hormone secretion [98]. COVID-19 is characterized as a high-stress disease, and this psychological condition may be responsible for the hormone impairment in COVID-19 men [106]. The key studies of the effects of COVID-19 infection and vaccination on male fertility parameters are shown in Table 1.

8. Conclusions

Recent multi-center research during the outbreak indicates that male fertility might be impaired as a possible consequence of COVID-19. Hence, more attention should be paid to the effect of COVID-19 infection on the male reproductive system. So far, the clinical trials have shown that there is no defective effect of COVID-19 vaccines on male fertility potential. The lack of information regarding the effect of SARS-CoV-2 infection on male fertility represents a weakness for the present understanding of COVID-19. Therefore, we suggest that the medical community should provide assurance to the population about the safety of vaccines for male reproductive function. As COVID-19 infection has demonstrated a clear correlation to male infertility, it is suggested that men should consider preserving their fertility by cryo-preserving their spermatozoa. Vaccination is an important way to prevent the negative effects of COVID-19 on male fertility.

Author Contributions

Conceptualization, S.P., A.N., Z.A., S.N.K., P.R., V.M., P.P., D.V., G.C., M.-Y.H., I.K., S.A. (Servet Altay), P.D., N.G.K.; writing—original draft preparation, N.G.K., C.d.G., P.D., S.A. (Stelios Assimakopoulos), C.G., G.A., P.P., N.M., Z.A. drafting the figure, revision, N.G.K., C.d.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that the review was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Bak, C.W.; Seok, H.H.; Song, S.H.; Kim, E.S.; Her, Y.S.; Yoon, T.K. Hormonal imbalances and psychological scars left behind in infertile men. J. Androl. 2012, 33, 181–189. [Google Scholar] [CrossRef] [PubMed]
  2. Nazari, A.; Ketabchi, A.A.; Pakmanesh, H.; Rahnama, A.; Fard, A.; Kharazmi, F. What is the optimum age of male fertility in infertile couples? J. Kerman Univ. Med. Sci. 2019, 26, 161–168. [Google Scholar]
  3. Wu, A.K.; Elliott, P.; Katz, P.P.; Smith, J.F. Time costs of fertility care: The hidden hardship of building a family. Fertil. Steril. 2013, 99, 2025–2030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Nazari, A.; Mokhtari, M.; Rouhafza, R.; Pourmasumi, S. Pregnancy and live birth rate in idiopathic male infertility treated with Human Menopausal Gonadotropin: A pilot clinical trial. J. Midwifery Reprod. Health 2020, 8, 2016–2021. [Google Scholar]
  5. Zegers-Hochschild, F.; Adamson, G.D.; Dyer, S.; Racowsky, C.; De Mouzon, J.; Sokol, R.; Rienzi, L.; Sunde, A.; Schmidt, L.; Cooke, I.D.; et al. The international glossary on infertility and fertility care, 2017. Hum. Reprod. 2017, 32, 1786–1801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Vander Borght, M.; Wyns, C. Fertility and infertility: Definition and epidemiology. Clin. Biochem. 2018, 62, 2–10. [Google Scholar] [CrossRef]
  7. Inhorn, M.C.; Patrizio, P. Infertility around the globe: New thinking on gender, reproductive technologies and global movements in the 21st century. Hum. Reprod. Update 2015, 21, 411–426. [Google Scholar] [CrossRef] [Green Version]
  8. Sun, H.; Gong, T.-T.; Jiang, Y.-T.; Zhang, S.; Zhao, Y.-H.; Wu, Q.-J. Global, regional, and national prevalence and disability-adjusted life-years for infertility in 195 countries and territories, 1990–2017: Results from a global burden of disease study, 2017. Aging 2019, 11, 10952. [Google Scholar] [CrossRef]
  9. Gianzo, M.; Urizar-Arenaza, I.; Muñoa-Hoyos, I.; Larreategui, Z.; Garrido, N.; Casis, L.; Irazusta, J.; Subirán, N. Human sperm testicular angiotensin-converting enzyme helps determine human embryo quality. Asian J. Androl. 2018, 20, 498. [Google Scholar]
  10. Moein, M.R.; Moein, M.R.; Ghasemzadeh, J.; Pourmasoumi, S. Evaluation of sperm retrieval rate with bilateral testicular sperm extraction in infertile patients with azoospermia. Iran. J. Reprod. Med. 2015, 13, 711–714. [Google Scholar]
  11. Pourmasumi, S.; Nazari, A.; Fagheirelahee, N.; Sabeti, P. Cytochemical tests to investigate sperm DNA damage: Assessment and review. J. Family Med. Prim. Care 2019, 8, 1533–1539. [Google Scholar] [PubMed]
  12. Stanley, K.E.; Thomas, E.; Leaver, M.; Wells, D. Coronavirus disease-19 and fertility: Viral host entry protein expression in male and female reproductive tissues. Fertil. Steril. 2020, 114, 33–43. [Google Scholar] [CrossRef] [PubMed]
  13. Helmy, Y.A.; Fawzy, M.; Elaswad, A.; Sobieh, A.; Kenney, S.P.; Shehata, A.A. The COVID-19 pandemic: A comprehensive review of taxonomy, genetics, epidemiology, diagnosis, treatment, and control. J. Clin. Med. 2020, 9, 1225. [Google Scholar] [CrossRef] [PubMed]
  14. Malle, L. A map of SARS-CoV-2 and host cell interactions. Nat. Rev. Immunol. 2020, 20, 351. [Google Scholar] [CrossRef]
  15. Evenson, D.P.; Jost, L.K.; Corzett, M.; Balhorn, R. Characteristics of human sperm chromatin structure following an episode of influenza and high fever: A case study. J. Androl. 2000, 21, 739–746. [Google Scholar]
  16. Sergerie, M.; Mieusset, R.; Croute, F.; Daudin, M.; Bujan, L. High risk of temporary alteration of semen parameters after recent acute febrile illness. Fertil. Steril. 2007, 88, 970.e1–970.e7. [Google Scholar] [CrossRef]
  17. Jin, J.M.; Bai, P.; He, W.; Wu, F.; Liu, X.F.; Han, D.M.; Liu, S.; Yang, J.K. Gender differences in patients with COVID-19: Focus on severity and mortality. Front. Public Health 2020, 8, 152. [Google Scholar] [CrossRef]
  18. Selvaraj, K.; Ravichandran, S.; Krishnan, S.; Radhakrishnan, R.K.; Manickam, N.; Kandasamy, M. Testicular atrophy and hypothalamic pathology in COVID-19: Possibility of the incidence of male infertility and HPG axis abnormalities. Reprod. Sci. 2021, 28, 2735–2742. [Google Scholar] [CrossRef]
  19. Onyema, E.M.; Eucheria, N.C.; Obafemi, F.A.; Sen, S.; Atonye, F.G.; Sharma, A.; Alsayed, A.O. Impact of Coronavirus pandemic on education. J. Educ. Pract. 2020, 11, 108–121. [Google Scholar]
  20. Khehra, N.; Padda, I.; Jaferi, U.; Atwal, H.; Narain, S.; Parmar, M.S. Tozinameran (BNT162b2) vaccine: The journey from preclinical research to clinical trials and authorization. AAPS PharmSciTech 2021, 22, 172. [Google Scholar] [CrossRef]
  21. Robinson, L.B.; Landman, A.B.; Shenoy, E.S.; Hashimoto, D.; Fu, X.; Camargo, C.A.; Wickner, P.; Blumenthal, K.G. Allergic symptoms after mRNA COVID-19 vaccination and risk of incomplete vaccination. J. Allergy Clin. Immunol. Practice. 2021, 9, 3200–3202. [Google Scholar] [CrossRef] [PubMed]
  22. Borroni, E.; Consonni, D.; Cugno, M.; Lombardi, A.; Mangioni, D.; Bono, P.; Oggioni, M.; Renteria, S.U.; Bordini, L.; Nava, C.D.; et al. Side effects among healthcare workers from a large Milan university hospital after second dose of BNT162b2 mRNA COVID-19 vaccine. Med. Lav. 2021, 112, 477. [Google Scholar]
  23. Markiewicz-Gospodarek, A.; Wdowiak, P.; Czeczelewski, M.; Forma, A.; Flieger, J.; Januszewski, J.; Radzikowska-Büchner, E.; Baj, J. The impact of SARS-CoV-2 infection on fertility and female and male reproductive systems. J. Clin. Med. 2021, 10, 4520. [Google Scholar] [CrossRef] [PubMed]
  24. Reid, J.A.; Mabhala, M.A. Ethnic and minority group differences in engagement with COVID-19 vaccination programmes—At Pandemic Pace; When vaccine confidence in mass rollout meets local vaccine hesitancy. Isr. J. Health Policy Res. 2021, 10, 33. [Google Scholar] [CrossRef] [PubMed]
  25. Chandi, A.; Jain, N. State of assisted reproduction technology in the coronavirus disease 2019 era and consequences on human reproductive system. Biol. Reprod. 2021, 105, 808–821. [Google Scholar] [CrossRef]
  26. Lu, H.; Stratton, C.W.; Tang, Y.W. Outbreak of pneumonia of unknown etiology in Wuhan, China: The mystery and the miracle. J. Med. Virol. 2020, 92, 401. [Google Scholar] [CrossRef] [Green Version]
  27. Jin, Y.; Yang, H.; Ji, W.; Wu, W.; Chen, S.; Zhang, W.; Duan, G. Virology, epidemiology, pathogenesis, and control of COVID-19. Viruses 2020, 12, 372. [Google Scholar] [CrossRef]
  28. Anifandis, G.; Messini, C.I.; Daponte, A.; Messinis, I.E. COVID-19 and fertility: A virtual reality. Reprod. BioMed. Online 2020, 41, 157–159. [Google Scholar] [CrossRef]
  29. Salam, A.P.; Horby, P.W. The breadth of viruses in human semen. Emerg. Infect. Dis. 2017, 23, 1922. [Google Scholar] [CrossRef]
  30. Klimova, R.R.; Chichev, E.V.; Naumenko, V.A.; Gadzhieva, Z.S.; Tsibisov, A.S.; Adieva, A.A.; L’vov, D.K.; Kurilo, L.F.; Shileĭko, L.V.; Ostroumova, T.V.; et al. Herpes simplex virus and cytomegalovirus in male ejaculate: Herpes simplex virus is more frequently encountered in idiopathic infertility and correlates with the reduction in sperm parameters. Vopr. Virusol. 2010, 55, 27–31. [Google Scholar]
  31. Garrido, N.; Meseguer, M.; Remohí, J.; Simón, C.; Pellicer, A. Semen characteristics in human immunodeficiency virus (HIV)-and hepatitis C (HCV)-seropositive males: Predictors of the success of viral removal after sperm washing. Hum. Reprod. 2005, 20, 1028–1034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Tian, Y.; Zhou, L.-Q. Evaluating the impact of COVID-19 on male reproduction. Reproduction 2021, 161, R37–R44. [Google Scholar] [CrossRef] [PubMed]
  33. Adamyan, L.; Elagin, V.; Vechorko, V.; Stepanian, A.; Dashko, A.; Doroshenko, D.; Aznaurova, Y.; Sorokin, M.; Garazha, A.; Buzdin, A. A Review of Recent Studies on the Effects of SARS-CoV-2 Infection and SARS-CoV-2 Vaccines on Male Reproductive Health. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2022, 28, e935879. [Google Scholar] [CrossRef] [PubMed]
  34. Olana, S.; Mazzilli, R.; Salerno, G.; Zamponi, V.; Tarsitano, M.G.; Simmaco, M.; Paoli, D.; Faggiano, A. 4BNT162b2 mRNA COVID-19 Vaccine and semen: What do we know? Andrology 2022, 10, 1023–1029. [Google Scholar] [CrossRef] [PubMed]
  35. Zhu, H.; Wang, X.; Zhang, F.; Zhu, Y.; Du, M.-R.; Tao, Z.-W.; Sun, C.; Ma, H.-T.; Li, Y.-D.; Liang, G.-Q.; et al. Evaluation of inactivated COVID-19 vaccine on semen parameters in reproductive-age males: A retrospective cohort study. Asian J. Androl. 2022, 24, 441–444. [Google Scholar] [PubMed]
  36. Barda, S.; Laskov, I.; Grisaru, D.; Lehavi, O.; Kleiman, S.; Wenkert, A.; Azem, F.; Hauser, R.; Michaan, N. The impact of COVID-19 vaccine on sperm quality. Int. J. Gynaecol. Obstet. 2022, 158, 116–120. [Google Scholar] [CrossRef] [PubMed]
  37. Chatzimeletiou, K.; Fleva, A.; Sioga, A.; Georgiou, I.; Nikolopoulos, T.T.; Markopoulou, M.; Petrogiannis, N.; Anifandis, G.; Patrikiou, A.; Kolibianakis, E.; et al. Effects of Different Drug Therapies and COVID-19 mRNA Vaccination on Semen Quality in a Man with Ankylosing Spondylitis: A Case Report. Medicina 2022, 58, 173. [Google Scholar] [CrossRef] [PubMed]
  38. Kumar, V.; Kaur, M. COVID-19 vaccine and male fertility. Urol. J. 2021, 4, 6987. [Google Scholar] [CrossRef]
  39. Carto, C.; Nackeeran, S.; Ramasamy, R. COVID-19 vaccination is associated with a decreased risk of orchitis and/or epididymitis in men. Andrologia 2022, 54, e14281. [Google Scholar] [CrossRef]
  40. Kaur, R.J.; Dutta, S.; Bhardwaj, P.; Charan, J.; Dhingra, S.; Mitra, P.; Singh, K.; Yadav, D.; Sharma, P.; Misra, S. Adverse events reported from COVID-19 vaccine trials: A systematic review. Indian J. Clin. Biochem. 2021, 36, 427–439. [Google Scholar] [CrossRef]
  41. Wesselink, A.K.; Hatch, E.E.; Rothman, K.J.; Wang, T.R.; Willis, M.D.; Yland, J.; Crowe, H.M.; Geller, R.J.; Willis, S.K.; Perkins, R.B.; et al. A Prospective Cohort Study of COVID-19 Vaccination, SARS-CoV-2 Infection, and Fertility. Am. J. Epidemiol. 2022, 191, 1383–1395. [Google Scholar] [CrossRef] [PubMed]
  42. Koc, E.; Keseroğlu, B.B. Does COVID-19 worsen the semen parameters? Early results of a tertiary healthcare center. Urol. Int. 2021, 105, 743–748. [Google Scholar] [CrossRef] [PubMed]
  43. Gacci, M.; Coppi, M.; Baldi, E.; Sebastianelli, A.; Zaccaro, C.; Morselli, S.; Pecoraro, A.; Manera, A.; Nicoletti, R.; Liaci, A.; et al. Semen impairment and occurrence of SARS-CoV-2 virus in semen after recovery from COVID-19. Hum. Reprod. 2021, 36, 1520–1529. [Google Scholar] [CrossRef] [PubMed]
  44. Li, D.; Jin, M.; Bao, P.; Zhao, W.; Zhang, S. Clinical characteristics and results of semen tests among men with coronavirus disease 2019. JAMA Netw. Open 2020, 3, e208292. [Google Scholar] [CrossRef] [PubMed]
  45. Maya, W.D.C.; Du Plessis, S.S.; Velilla, P.A. SARS-CoV-2 and the testis: Similarity with other viruses and routes of infection. Reprod. Biomed. Online 2020, 40, 763–764. [Google Scholar] [CrossRef]
  46. Ivell, R. Lifestyle impact and the biology of the human scrotum. Reprod. Biol. Endocrinol. 2007, 5, 15. [Google Scholar] [CrossRef] [Green Version]
  47. Pozzilli, P.; Lenzi, A. Commentary: Testosterone, a key hormone in the context of COVID-19 pandemic. Metabolism 2020, 108, 154252. [Google Scholar] [CrossRef]
  48. Song, C.; Wang, Y.; Li, W.; Hu, B.; Chen, G.; Xia, P.; Wang, W.; Li, C.; Diao, F.; Hu, Z.; et al. Absence of 2019 novel coronavirus in semen and testes of COVID-19 patients. Biol. Reprod. 2020, 103, 4–6. [Google Scholar] [CrossRef] [Green Version]
  49. Holtmann, N.; Edimiris, P.; Andree, M.; Doehmen, C.; Baston-Buest, D.; Adams, O.; Kruessel, J.S.; Bielfeld, A.P. Assessment of SARS-CoV-2 in human semen—A cohort study. Fertil. Steril. 2020, 114, 233–238. [Google Scholar] [CrossRef]
  50. Song, C.; Wang, Y.; Li, W.; Hu, B.; Chen, G.; Xia, P.; Wang, W.; Li, C.; Hu, Z.; Yang, X.; et al. Detection of 2019 novel coronavirus in semen and testicular biopsy specimen of COVID-19 patients. medRxiv 2020. [Google Scholar] [CrossRef]
  51. Paoli, D.; Pallotti, F.; Colangelo, S.; Basilico, F.; Mazzuti, L.; Turriziani, O.; Antonelli, G.; Lenzi, A.; Lombardo, F. Study of SARS-CoV-2 in semen and urine samples of a volunteer with positive naso-pharyngeal swab. J. Endocrinol. Investig. 2020, 43, 1819–1822. [Google Scholar] [CrossRef] [PubMed]
  52. Pan, F.; Xiao, X.; Guo, J.; Song, Y.; Li, H.; Patel, D.P.; Spivak, A.M.; Alukal, J.P.; Zhang, X.; Xiong, C.; et al. No evidence of severe acute respiratory syndrome–coronavirus 2 in semen of males recovering from coronavirus disease 2019. Fertil. Steril. 2020, 113, 1135–1139. [Google Scholar] [CrossRef] [PubMed]
  53. Ma, L.; Xie, W.; Li, D.; Shi, L.; Ye, G.; Mao, Y.; Xiong, Y.; Sun, H.; Zheng, F.; Chen, Z.; et al. Evaluation of sex-related hormones and semen characteristics in reproductive-aged male COVID-19 patients. J. Med. Virol. 2021, 93, 456–462. [Google Scholar] [CrossRef] [PubMed]
  54. Hu, B.; Liu, K.; Ruan, Y.; Wei, X.; Wu, Y.; Feng, H.; Deng, Z.; Liu, J.; Wang, T. Evaluation of mid-and long-term impact of COVID-19 on male fertility through evaluating semen parameters. Transl. Androl. Urol. 2022, 11, 159. [Google Scholar] [CrossRef] [PubMed]
  55. Ruan, Y.; Hu, B.; Liu, Z.; Liu, K.; Jiang, H.; Li, H.; Li, R.; Luan, Y.; Liu, X.; Yu, G.; et al. No detection of SARS-CoV-2 from urine, expressed prostatic secretions, and semen in 74 recovered COVID-19 male patients: A perspective and urogenital evaluation. Andrology 2021, 9, 99–106. [Google Scholar] [CrossRef]
  56. Segars, J.; Katler, Q.; McQueen, D.B.; Kotlyar, A.; Glenn, T.; Knight, Z.; Feinberg, E.C.; Taylor, H.S.; Toner, J.P.; Kawwass, J.F. Prior and novel coronaviruses, Coronavirus Disease 2019 (COVID-19), and human reproduction: What is known? Fertil. Steril. 2020, 113, 1140–1149. [Google Scholar] [CrossRef]
  57. Donders, G.G.; Bosmans, E.; Reumers, J.; Donders, F.; Jonckheere, J.; Salembier, G.; Stern, N.; Jacquemyn, Y.; Ombelet, W.; Depuydt, C.E. Sperm quality and absence of SARS-CoV-2 RNA in semen after COVID-19 infection: A prospective, observational study and validation of the SpermCOVID test. Fertil. Steril. 2022, 117, 287–296. [Google Scholar] [CrossRef]
  58. Guo, L.; Zhao, S.; Li, W.; Wang, Y.; Li, L.; Jiang, S.; Ren, W.; Yuan, Q.; Zhang, F.; Kong, F.; et al. Absence of SARS-CoV-2 in semen of a COVID-19 patient cohort. Andrology 2021, 9, 42–47. [Google Scholar] [CrossRef]
  59. Li, H.; Xiao, X.; Zhang, J.; Zafar, M.I.; Wu, C.; Long, Y.; Lu, W.; Pan, F.; Meng, T.; Zhao, K.; et al. Impaired spermatogenesis in COVID-19 patients. EClinicalMedicine 2020, 28, 100604. [Google Scholar] [CrossRef]
  60. Piroozmanesh, H.; Cheraghi, E.; Naserpoor, L.; Aghashahi, M.; Jannatifar, R. The Effect of COVID-19 Infection on Sperm Quality and Male Fertility. Jentashapir J. Cell. Mol. Biol. 2021, 12, e115390. [Google Scholar] [CrossRef]
  61. He, Y.; Wang, J.; Ren, J.; Zhao, Y.; Chen, J.; Chen, X. Effect of COVID-19 on male reproductive system—A systematic review. Front. Endocrinol. 2021, 12, 677701. [Google Scholar] [CrossRef] [PubMed]
  62. Carlsen, E.; Andersson, A.M.; Petersen, J.H.; Skakkebæk, N.E. History of febrile illness and variation in semen quality. Hum. Reprod. 2003, 18, 2089–2092. [Google Scholar] [CrossRef] [PubMed]
  63. Temiz, M.Z.; Dincer, M.M.; Hacibey, I.; Yazar, R.O.; Celik, C.; Kucuk, S.H.; Alkurt, G.; Doganay, L.; Yuruk, E.; Muslumanoglu, A.Y. Investigation of SARS-CoV-2 in semen samples and the effects of COVID-19 on male sexual health by using semen analysis and serum male hormone profile: A cross-sectional, pilot study. Andrologia 2021, 53, e13912. [Google Scholar] [CrossRef] [PubMed]
  64. Falahieh, F.M.; Zarabadipour, M.; Mirani, M.; Abdiyan, M.; Dinparvar, M.; Alizadeh, H.; Paktinat, S.; Hosseinirad, H. Effects of moderate COVID-19 infection on semen oxidative status and parameters 14 and 120 days after diagnosis. Reprod. Fertil. Dev. 2021, 33, 683–690. [Google Scholar] [CrossRef]
  65. Maleki, B.H.; Tartibian, B. COVID-19 and male reproductive function: A prospective, longitudinal cohort study. Reproduction 2021, 161, 319–331. [Google Scholar] [CrossRef]
  66. Smith, R.; Kaune, H.; Parodi, D.; Madariaga, M.; Morales, I.; Ríos, R.; Castro, A. Extent of sperm DNA damage in spermatozoa from men examined for infertility. Relationship with oxidative stress. Rev. Med. Chile 2007, 135, 279–286. [Google Scholar]
  67. Andrade-Rocha, F.T. Temporary impairment of semen quality following recent acute fever. Ann. Clin. Lab. Sci. 2013, 43, 94–97. [Google Scholar]
  68. Jung, A.; Schuppe, H.; Schill, W. Fever as etiology of temporary infertility in the man. Hautarzt Z. Dermatol. Venerol. VerwandteGeb. 2001, 52, 1090–1093. [Google Scholar] [CrossRef]
  69. Khomich, O.A.; Kochetkov, S.N.; Bartosch, B.; Ivanov, A.V. Redox biology of respiratory viral infections. Viruses 2018, 10, 392. [Google Scholar] [CrossRef] [Green Version]
  70. Dutta, S.; Majzoub, A.; Agarwal, A. Oxidative stress and sperm function: A systematic review on evaluation and management. Arab J. Urol. 2019, 17, 87–97. [Google Scholar] [CrossRef]
  71. Marin, L.; Ambrosini, G.; Nuzzi, L.; Bordin, L.; Andrisani, A. A suggestion to overcome potential risks for infertility due to COVID-19: A response to Aitken. Andrology 2021, 10, 11. [Google Scholar] [CrossRef] [PubMed]
  72. Koppers, A.J.; Mitchell, L.A.; Wang, P.; Lin, M.; Aitken, R.J. Phosphoinositide 3-kinase signalling pathway involvement in a truncated apoptotic cascade associated with motility loss and oxidative DNA damage in human spermatozoa. Biochem. J. 2011, 436, 687–698. [Google Scholar] [CrossRef] [PubMed]
  73. Xu, J.; Qi, L.; Chi, X.; Yang, J.; Wei, X.; Gong, E.; Peh, S.; Gu, J. Orchitis: A complication of severe acute respiratory syndrome (SARS). Biol. Reprod. 2006, 74, 410–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Stranieri, A.; Probo, M.; Pisu, M.C.; Fioletti, A.; Meazzi, S.; Gelain, M.E.; Bonsembiante, F.; Lauzi, S.; Paltrinieri, S. Preliminary investigation on feline coronavirus presence in the reproductive tract of the tom cat as a potential route of viral transmission. J. Feline Med. Surg. 2020, 22, 178–185. [Google Scholar] [CrossRef]
  75. Bian, X.-W. Autopsy of COVID-19 patients in China. Natl. Sci. Rev. 2020, 7, 1414–1418. [Google Scholar] [CrossRef]
  76. La Marca, A.; Busani, S.; Donno, V.; Guaraldi, G.; Ligabue, G.; Girardis, M. Testicular pain as an unusual presentation of COVID-19: A brief review of SARS-CoV-2 and the testis. Reprod. Biomed. Online 2020, 41, 903–906. [Google Scholar] [CrossRef]
  77. Abobaker, A.; Raba, A.A. Does COVID-19 affect male fertility? World J. Urol. 2021, 39, 975–976. [Google Scholar] [CrossRef] [Green Version]
  78. Kelly, J.C.; Dombrowksi, M.; O’Neil-Callahan, M.; Kernberg, A.S.; Frolova, A.I.; Stout, M.J. False-negative testing for severe acute respiratory syndrome coronavirus 2: Consideration in obstetrical care. Am. J. Obstet. Gynecol. MFM 2020, 2, 100130. [Google Scholar] [CrossRef]
  79. Olaniyan, O.T.; Dare, A.; Okotie, G.E.; Adetunji, C.O.; Ibitoye, B.O.; Bamidele, O.J.; Eweoya, O.O. Testis and blood-testis barrier in COVID-19 infestation: Role of angiotensin-converting enzyme 2 in male infertility. J. Basic Clin. Physiol. Pharmacol. 2020, 31, 20200156. [Google Scholar] [CrossRef]
  80. Aziz, N.; Agarwal, A.; Lewis-Jones, I.; Sharma, R.K.; Thomas, A.J., Jr. Novel associations between specific sperm morphological defects and leukocytospermia. Fertil. Steril. 2004, 82, 621–627. [Google Scholar] [CrossRef]
  81. Ulrich, H.; Pillat, M.M. CD147 as a target for COVID-19 treatment: Suggested effects of azithromycin and stem cell engagement. Stem Cell Rev. Rep. 2020, 16, 434–440. [Google Scholar] [CrossRef] [PubMed]
  82. López-Romero, R.; Nambo-Lucio, M.d.J.; Salcedo-Carrillo, E.; Hernández-Cueto, M.d.l.Á.; Salcedo-Vargas, M. The big challenge of SARS-CoV-2 latency: Testes as reservoir. Gac. Médica México 2020, 156, 324–329. [Google Scholar] [CrossRef]
  83. Yang, M.; Chen, S.; Huang, B.O.; Zhong, J.M.; Su, H.; Chen, Y.J.; Cao, Q.; Ma, L.; He, J.; Li, X.F.; et al. Pathological findings in the testes of COVID-19 patients: Clinical implications. Eur. Urol. Focus 2020, 6, 1124–1129. [Google Scholar] [CrossRef] [PubMed]
  84. Jung, A.; Schuppe, H.C. Influence of genital heat stress on semen quality in humans. Andrologia 2007, 39, 203–215. [Google Scholar] [CrossRef] [PubMed]
  85. Itoh, M.; Hiramine, C.; Tokunaga, Y.; Mukasa, A.; Hojo, K. A New Murine Model of Autoimmune Orchitis Induced by Immunization With Viable Syngeneic Testicular Germ Cells Alone.: II. Immunohistochemical Findings of Fully-Developed Inflammatory Lesion. Autoimmunity 1991, 10, 89–97. [Google Scholar] [CrossRef] [PubMed]
  86. Moldoveanu, Z.; Huang, W.-Q.; Kulhavy, R.; Pate, M.S.; Mestecky, J. Human male genital tract secretions: Both mucosal and systemic immune compartments contribute to the humoral immunity. J. Immunol. 2005, 175, 4127–4136. [Google Scholar] [CrossRef]
  87. Rockett, J.C.; Mapp, F.L.; Garges, J.B.; Luft, J.C.; Mori, C.; Dix, D.J. Effects of hyperthermia on spermatogenesis, apoptosis, gene expression, and fertility in adult male mice. Biol. Reprod. 2001, 65, 229–239. [Google Scholar] [CrossRef]
  88. Wiwanitkit, V. Influenza, swine flu, sperm quality and infertility: A story. J. Hum. Reprod. Sci. 2010, 3, 116. [Google Scholar] [CrossRef]
  89. Reis, A.B.; Araújo, F.C.; Pereira, V.M.; Dos Reis, A.M.; Santos, R.A.; Reis, F.M. Angiotensin (1–7) and its receptor Mas are expressed in the human testis: Implications for male infertility. J. Mol. Histol. 2010, 41, 75–80. [Google Scholar] [CrossRef]
  90. Duarte-Neto, A.N.; Monteiro, R.A.; da Silva, L.F.; Malheiros, D.M.; de Oliveira, E.P.; Theodoro-Filho, J.; Pinho, J.R.R.; Gomes-Gouvêa, M.S.; Salles, A.P.M.; de Oliveira, I.R.S.; et al. Pulmonary and systemic involvement in COVID-19 patients assessed with ultrasound-guided minimally invasive autopsy. Histopathology 2020, 77, 186–197. [Google Scholar] [CrossRef]
  91. Teixeira, T.A.; Bernardes, F.S.; Oliveira, Y.C.; Hsieh, M.K.; Esteves, S.C.; Duarte Neto, A.N.; Kallas, E.G.; Hallak, J. SARS-CoV-2 and Multi-Organ damage—What men’s health specialists should know about the COVID-19 pathophysiology. Int. Braz. J. Urol. 2021, 47, 637–646. [Google Scholar] [CrossRef] [PubMed]
  92. Teixeira, T.A.; Oliveira, Y.C.; Bernardes, F.S.; Kallas, E.G.; Duarte-Neto, A.N.; Esteves, S.C.; Drevet, J.R.; Hallak, J. Viral infections and implications for male reproductive health. Asian J. Androl. 2021, 23, 335. [Google Scholar] [PubMed]
  93. Lukassen, S.; Chua, R.L.; Trefzer, T.; Kahn, N.C.; Schneider, M.A.; Muley, T.; Winter, H.; Meister, M.; Veith, C.; Boots, A.W.; et al. SARS-CoV-2 receptor ACE2 and TMPRSS2 are predominantly expressed in a transient secretory cell type in subsegmental bronchial branches. EMBO J. 2020, 39, e105114. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, Z.; Xu, X. scRNA-seq profiling of human testes reveals the presence of the ACE2 receptor, a target for SARS-CoV-2 infection in spermatogonia, Leydig and Sertoli cells. Cells 2020, 9, 920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lucas, J.M.; Heinlein, C.; Kim, T.; Hernandez, S.A.; Malik, M.S.; True, L.D.; Morrissey, C.; Corey, E.; Montgomery, B.; Mostaghel, E.; et al. The androgen-regulated protease TMPRSS2 activates a proteolytic cascade involving components of the tumor microenvironment and promotes prostate cancer metastasis. Cancer Discov. 2014, 4, 1310–1325. [Google Scholar] [CrossRef] [Green Version]
  96. Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Ou, C.Q.; He, J.X.; Liu, L.; Shan, H.; Lei, C.L.; Hui, D.S.; et al. China medical treatment expert group for COVID-19. Clin. Charact. Coronavirus Dis. 2019, 382, 1708–1720. [Google Scholar]
  97. Rastrelli, G.; Di Stasi, V.; Inglese, F.; Beccaria, M.; Garuti, M.; Di Costanzo, D.; Spreafico, F.; Greco, G.F.; Cervi, G.; Pecoriello, A.; et al. Low testosterone levels predict clinical adverse outcomes in SARS-CoV-2 pneumonia patients. Andrology 2021, 9, 88–98. [Google Scholar] [CrossRef]
  98. Li, R.; Yin, T.; Fang, F.; Li, Q.; Chen, J.; Wang, Y.; Hao, Y.; Wu, G.; Duan, P.; Wang, Y.; et al. Potential risks of SARS-CoV-2 infection on reproductive health. Reprod. Biomed. Online 2020, 41, 89–95. [Google Scholar] [CrossRef]
  99. Ma, L.; Xie, W.; Li, D.; Shi, L.; Mao, Y.; Xiong, Y.; Zhang, Y.; Zhang, M. Effect of SARS-CoV-2 infection upon male gonadal function: A single center-based study. medRxiv 2020. Preprint. [Google Scholar] [CrossRef] [Green Version]
  100. José, F.G.; González, J.G.Á.; Molina, J.M.C.; Arnau, L.B.; Iribarren, I.M.; Jabaloyas, J.M.M.; Rico, F.M.; García-Baquero, R.; Gayá, M.R.; García, E.L.; et al. SARS-CoV-2 infection: Implications for sexual and reproductive health. A position statement of the Asociación Española de Andrología, Medicina Sexual y Reproductiva (ASESA). Rev. Int. Androl. 2020, 18, 117–123. [Google Scholar]
  101. Schroeder, M.; Schaumburg, B.; Mueller, Z.; Parplys, A.; Jarczak, D.; Nierhaus, A.; Kloetgen, A.; Schneider, B.; Peschka, M.; Stoll, F.; et al. The majority of male patients with COVID-19 present low testosterone levels on admission to Intensive Care in Hamburg, Germany: A retrospective cohort study. medRxiv 2020. [Google Scholar] [CrossRef]
  102. Çayan, S.; Uğuz, M.; Saylam, B.; Akbay, E. Effect of serum total testosterone and its relationship with other laboratory parameters on the prognosis of coronavirus disease 2019 (COVID-19) in SARS-CoV-2 infected male patients: A cohort study. Aging Male 2020, 23, 1493–1503. [Google Scholar] [CrossRef] [PubMed]
  103. Neto, F.T.L.; Bach, P.V.; Najari, B.B.; Li, P.S.; Goldstein, M. Spermatogenesis in humans and its affecting factors. In Seminars in Cell & Developmental Biology; Academic Press: Cambridge, MA, USA, 2016; Volume 59, pp. 10–26. [Google Scholar]
  104. Guan, X.; Chen, F.; Chen, P.; Zhao, X.; Mei, H.; Liu, J.; Lian, Q.; Zirkin, B.R.; Chen, H. Effects of spermatogenic cycle on Stem Leydig cell proliferation and differentiation. Mol. Cell. Endocrinol. 2019, 481, 35–43. [Google Scholar] [CrossRef] [PubMed]
  105. Salonia, A.; Corona, G.; Giwercman, A.; Maggi, M.; Minhas, S.; Nappi, R.E.; Sofikitis, N.; Vignozzi, L. SARS-CoV-2, Testosterone and frailty in males (PROTEGGIMI): A multidimensional research project. Andrology 2021, 9, 19–22. [Google Scholar] [CrossRef]
  106. Amiri, I.; Najafi, R.; Sheykh, N. Nitric oxide level in seminal plasma and its relation with sperm DNA damages. Iran. Biomed. J. 2007, 11, 259–264. [Google Scholar]
Figure 1. Once infection begins, the virus can affect the testes and prostate through the interaction with angiotensin I-converting enzyme 2 (ACE2) receptors and transmembrane serine protease. However, this complication does not occur in every patient with COVID-19 disease, being more likely in predisposed individuals with lengthy infection or fever. As the virus moves into their reproductive organs, seminal fluid and sperm characteristics are modified. Infection may also cause direct seminal fluid inflammation and sperm damage, with polymorphonucleate and lymphocyte activation, interleukins (IL-6, IL-8, others) and cytokine release, tumor necrosis factor (TNF-α), etc. Most authors reported these as transient complications (see text).
Figure 1. Once infection begins, the virus can affect the testes and prostate through the interaction with angiotensin I-converting enzyme 2 (ACE2) receptors and transmembrane serine protease. However, this complication does not occur in every patient with COVID-19 disease, being more likely in predisposed individuals with lengthy infection or fever. As the virus moves into their reproductive organs, seminal fluid and sperm characteristics are modified. Infection may also cause direct seminal fluid inflammation and sperm damage, with polymorphonucleate and lymphocyte activation, interleukins (IL-6, IL-8, others) and cytokine release, tumor necrosis factor (TNF-α), etc. Most authors reported these as transient complications (see text).
Vaccines 10 01982 g001
Table 1. COVID-19 vaccination and male reproductive potential. Summarization of some key studies of the effects of COVID-19 infection and vaccination on male fertility parameters.
Table 1. COVID-19 vaccination and male reproductive potential. Summarization of some key studies of the effects of COVID-19 infection and vaccination on male fertility parameters.
First NameType of StudyFinding and Conclusion
Seminal Fluid
Olana et al. (2022) [34]Prospective studyInflammatory factors including IL-6 did not increase after vaccination in human seminal fluid.
Gacci et al. (2021) [43]Cohort studyIL-8 was detected in seminal fluid of COVID-19 patients.
Li et al. (2020) [44]Cohort studySemen can shed the virus during the recovery phase, even though COVID-19 does not affect the semen of all men who have the virus.
Maya et al. (2020) [45]CommentaryCoronavirus is detectable in acute infected patients after treatment and in the recovery period. They also discussed that COVID-19 is detectable in patients who have no infection symptoms.
Song et al. (2020) [48]Letter to the EditorThey cannot identify the presence of COVID-19 in the seminal fluid of male patients after recovery.
Holtman et al. (2020) [49]Cohort studyThere was no RNA of COVID-19 in seminal fluid 8 to 54 days after contamination.
Paoli et al. (2020) [51]Case reportNegative presence of COVID-19 in infected patient’s seminal fluid and testis sample.
Pan et al. (2020) [52]Observational, cross-sectional studyNo SARS-CoV2 was found in semen fluids 31 days after confirming their infection with COVID-19.
Sperm Count
Zhu et al. (2022) [35]Cohort studyThere was no decrease in sperm count and volume after COVID-19 vaccination in comparison with before vaccination.
Barda et al. (2022) [36]Cohort studySperm concentration count significantly increased after the second vaccine.
Chatzimeletiou et al. (2022) [37]Case reportThe sperm count significantly increased from first to third vaccine dose.
Holtman et al. (2020) [49]Cohort studyIn COVID-19 patients, sperm concentration significantly decreased.
Ma et al. (2021) [53]Case control studyElevation of sperm DNA fragmentation is the cause of sperm count decrease in COVID-19 men.
Hu et al. (2022) [54]Case control studyCOVID-19 can significantly decrease sperm count in comparison with control group. They also reported sperm count in recovered patients after 150 days was similar to control group.
Ruan et al. (2021) [55]Cohort studySperm count was significantly lower in recovered patients than control group.
Segars et al. (2020) [56]Review studySperm analysis showed reduced sperm count 3 months after COVID-19.
Donders et al. (2020) [57]Cohort studyThere was a significant decreased sperm count in patients 1month after recovery in comparison with 3 months after recovery.
Guo et al. (2021) [58]Cohort studyCOVID-19 direct attack on sperm is responsible for sperm count decrease in COVID-19 patients.
Li et al. (2020) [59]Observational studyElevation of immunological factors including interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) are responsible for the difference in sperm count between COVID-19 patients and healthy control men.
Sperm Motility
Olana et al. (2022) [34]Prospective studyThere was no significant difference in OS level and sperm motility before and after vaccination.
Zhu et al. (2022) [35]Cohort studyCOVID-19 vaccination no had negative effects on sperm motility.
Chatzimeletiou et al. (2022) [37]Case reportIn ankylosing spondylitis patients, COVID-19 vaccination can improve sperm motility after second dose.
Holtman et al. (2020) [49]Cohort studySperm motility decreased in COVID-19 moderately infected patients.
Donders et al. (2020) [57]Cohort studySperm motility significantly decreased immediately after COVID-19, and over time (3 months after infection), sperm motility was repaired and increased compared to 1 month after infection.
Guo et al. (2021) [58]Cohort studyCOVID-19 no had negative effect on sperm motility.
Li et al. (2021) [59]Observational study COVID-19 had negative effects on sperm motility in fertile infected men.
Piroozmanesh et al. (2022) [60]Case control studySperm motility significantly decreased in COVID-19 patients.
He et al. (2021) [61]Systematic reviewSperm motility significantly decreased in moderate infection in comparison with mild infection or healthy control cases.
Sperm Morphology
Chatzimeletiou et al. (2022) [37]Case reportCOVID-19 vaccination had no negative effects on sperm morphology, although the shape of sperm in head and neck after ankylosing spondylitis showed more defects. However, vaccination had positive effects on sperm morphology.
Kumar et al. (2021) [38]Letter to the EditorThere was no scientific evidence to confirm the damaging effects of COVID-19 vaccination on sperm parameters.
Koc et al. (2021) [42]Research studyNormal sperm morphology in COVID-19 cases significantly decreased.
Ma et al. (2021) [53]Case control studyNormal morphology of sperm in COVID-19 patients significantly decreased.
Temiz et al. (2021) [63]A cross-sectional, pilot studyInfected patients before sperm morphology treatment significantly decreased in comparison with control group.
Falahieh et al. (2021) [64]Prospective studyPercentage of normal sperm morphology increased 3 months after COVID-19 treatment but based on WHO guideline, the rate of normal morphology was lower than standard.
Maleki et al. (2021) [65]Cohort studySperm morphology can decrease after COVID-19.
Sperm Viability
Ruan et al. (2021) [55]Cohort studySperm viability decreased in COVID-19 patients.
Piroozmanesh et al. (2022) [60]Case control studySperm viability significantly decreased in COVID-19 patients.
Falahieh et al. (2021) [64]Prospective studySperm viability showed no significant difference between 2 weeks and 3 months after infection.
Marin et al. (2021) [71]Letter to the EditorCoronavirus can induce ACE2 production, and this is a negative factor on sperm viability in COVID-19 patients.
Testis Tissue
Stanley et al. (2020) [12]Descriptive studyCOVID-19 used Basigin (BSG) and protease Cathepsin L (CTSL) receptors to enter Leydig cells.
Kumar et al. (2021) [38]Letter to the EditorRate of orchitis and epididymitis were significantly lower in vaccinated cases compared to non-vaccinated ones. They also concluded a single dose of COVID-19 vaccine can significantly decrease the risk of inflammation in testis.
Song et al. (2020) [48]Letter to the EditorThere were no COVID-19-positive cells in testis tissue.
Holtman et al. (2020) [49]Cohort studyMild infection of COVID-19 impairs testicular function.
Ma et al. (2021) [53]Case control studyThe function of gonads in COVID-19-infected patients was significantly lower than control group.
Li et al. (2020) [59]Observational study Pathological changes observed in 6 of 12 expired patients after COVID-19 infection.
Bian et al. (2020) [75]National science reviewConsiderable impairment was detected in seminiferous tubules and a decrease in spermatogenic cell lines of COVID-19 patients.
LaMarca et al. (2020) [76]Review studyIn COVID-19-infected cases, testicular pain in both sides by severity in left testis was observed.
Abobaker et al. (2021) [77]Letter to the EditorCOVID-19 could have an indirect impact on seminiferous function by affecting the hypothalamic–pituitary–testis axis through immunological and inflammatory responses.
Olaniyan et al. (2021) [79]Review studyBTB breakdown has thus been speculated for COVID-19 patients but still requires experimental validation.
Ulrish et al. (2020) [81]Review studyCOVID-19 infection can affect CD14 in testes via spermatogonia stem cells.
Yang et al. (2020) [83]Research studyInfection with COVID-19 has been shown to cause histopathological changes in postmortem testicular tissues.
Teixeira et al. (2021) [91]Mini reviewCOVID-19 causes orchitis, mild lymphocytosis, decreased Leydig cell counts, and destruction of seminiferous tubules in the testis.
Lukassen et al. (2020) [93]Research studyHigh ACE2 expression in the testis may facilitate entrance and colonization of virus, which can have adverse effects on male reproductive function.
Sex Hormones
Adamyan et al. (2022) [33]Review studyNo negative effects of the COVID-19 vaccine on the level of testosterone, FSH and LH hormone after vaccination.
Pozzilli et al. (2020) [47]Mini reviewThere was a negative correlation between the risk of COVID-19 and level of testosterone hormone.
Holtman et al. (2020) [49]Cohort studyIn COVID-19 patients, the level of sex hormones decreased significantly.
Ma et al. (2021) [53]Case control studyDecreased level of testosterone/LH and increased level of LH in COVID-19 patients compared to the controls.
Temiz et al. (2021) [63]A cross-sectional, pilot studyInfected patients with COVID-19 before treatment showed significant decreased level of hormones (testosterone, FSH and LH) in comparison with control group. Comparison between before and after treatment showed the level of hormones elevated after treatment and was similar to control group.
Rastrelli et al. (2021) [97]Research studyPatients with severe COVID-19 had low levels of total dihydrotestosterone.
Li et al. (2020) [98]Review studyImbalance of testosterone/LH/FSH ratio is prognostication of defects in male fertility during COVID-19.
Ma et al. (2020) [99]Research studyThe level of LH hormone was significantly higher in COVID-19 cases than control. Additionally, the testosterone/LH ratio was lower in COVID-19 patients than control.
Jose et al. (2020) [100]Systematic reviewLevel of sex hormones during COVID-19 showed decrease in the level of testosterone and increased in the level of LH.
Cayan et al. (2020) [102]Cohort studyConcentration of LH and FSH after COVID-19 increases.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pourmasumi, S.; Nazari, A.; Ahmadi, Z.; Kouni, S.N.; de Gregorio, C.; Koniari, I.; Dousdampanis, P.; Mplani, V.; Plotas, P.; Assimakopoulos, S.; et al. The Effect of Long COVID-19 Infection and Vaccination on Male Fertility; A Narrative Review. Vaccines 2022, 10, 1982. https://doi.org/10.3390/vaccines10121982

AMA Style

Pourmasumi S, Nazari A, Ahmadi Z, Kouni SN, de Gregorio C, Koniari I, Dousdampanis P, Mplani V, Plotas P, Assimakopoulos S, et al. The Effect of Long COVID-19 Infection and Vaccination on Male Fertility; A Narrative Review. Vaccines. 2022; 10(12):1982. https://doi.org/10.3390/vaccines10121982

Chicago/Turabian Style

Pourmasumi, Soheila, Alireza Nazari, Zahra Ahmadi, Sophia N. Kouni, Cesare de Gregorio, Ioanna Koniari, Periklis Dousdampanis, Virginia Mplani, Panagiotis Plotas, Stelios Assimakopoulos, and et al. 2022. "The Effect of Long COVID-19 Infection and Vaccination on Male Fertility; A Narrative Review" Vaccines 10, no. 12: 1982. https://doi.org/10.3390/vaccines10121982

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