Skip to main content

Correlation between viral infections in male semen and infertility: a literature review

Abstract

Infertility affects approximately one-sixth of couples globally, with the incidence of male infertility steadily increasing. However, our understanding of the impact of viral infections on fertility remains limited. This review consolidates findings from previous studies, outlining 40 viruses identified in human semen and summarizing their key characteristics, modes of transmission, and their effects on both the reproductive and endocrine systems. Furthermore, it elucidates potential pathogenic mechanisms and treatment prospects of viruses strongly associated with male infertility. This synthesis will enhance our comprehension of how viral infections influence male reproductive health, offering valuable insights for future research as well as the diagnosis and treatment of infectious infertility.

Graphical Abstract

Abbreviated summary: Semen viral infections can damage the male reproductive system, further affecting semen quality and causing infertility, as well as sexually transmitted infections to partners and vertical transmission resulting in poor pregnancy outcomes.

Introduction

The WHO explicitly defines infertility as the inability to conceive after at least 12 months of regular, unprotected sexual intercourse [1], which may stem from factors related to female, male, or both [2]. It’s reported that about 17% of couples globally grapple with infertility [3], with male infertility constituting half of all cases [4]. In recent years, male infertility has garnered increasing attention. A global burden of disease study encompassing 204 countries and territories revealed that in 2019, the prevalence of male infertility was approximately 56 million, marking a 76.9% increase since 1990 [5].

The reasons for decreased male fertility vary but may be linked to congenital, acquired, or idiopathic factors affecting sperm production [6], among which reproductive tract infections are considered to be one of the most influential [7, 8]. A survey involving over 4000 infertile men showed a prevalence rate of genitourinary tract infections in males as high as 35% [9]. Furthermore, 20% of primary infertile men exhibited asymptomatic semen infections, resulting in varying degrees of abnormal sperm concentration [10].

Previous research has compiled a list of 38 viruses detected in the male reproductive tract and semen, many of which exhibit a strong affinity for the male reproductive organs, particularly the testes [11]. Moreover, amidst the global coronavirus disease 2019 (COVID-19) pandemic, accumulating studies have indicated that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can directly or indirectly impair the male reproductive system, thereby heightening the risk of male infertility [12,13,14]. This resurgence of concern highlights the potential impact of viral infections on male fertility. Within semen, viruses may infect sperm or precursor cells, attaching to molecules on the sperm surface as free viral particles or residing within immune cells, leading to pathology in the reproductive system, abnormalities in semen parameters, and declined sperm quality. This poses a significant threat to individual fertility and overall health. Furthermore, this may also result in virus-induced mutations transmitted to future generations [15].

Despite recent advancements in the study of semen viruses, a definitive correlation between viral infections and fertility remains unclear. This review seeks to summarize the primary discoveries regarding viruses in human semen, delineate the diversity of viruses present, identify the key viral species linked to male infertility, and discuss potential avenues of virus research with clinical relevance and implications for fertility guidance.

Methods

To investigate the presence of viruses in semen, we conducted an unrestricted search on PubMed, Embase and Web of Science using the terms “semen, sperm, virus,” and we obtained 4815 results. Through screening titles, abstracts, and full-text articles, we gathered data describing viral infections in semen. Subsequently, we conducted another search using the term “(virus name) and sexual transmission” to find relevant evidence of sexual transmission.

Results

Our findings revealed the presence of 40 viruses in human semen, with some lacking data on sexual transmission. Among these 40 viruses, many cause chronic or latent infections such as herpes viruses and human immunodeficiency virus (HIV), while others cause acute infections such as zika virus (ZIKV), ebola virus (EBOV), and chikungunya virus (CHIKV). Among the viruses causing acute infections, only ZIKV and EBOV have been systematically studied beyond case reports. Table 1 summarizes the main characteristics, clinical manifestations, and key information related to male fertility for the 40 viruses. In the following sections, we will provide detailed explanations regarding viruses highly relevant to male reproductive health.

Table 1 Summary of virus species detected in semen

Human papillomavirus

Human papillomavirus (HPV) is a virus species with double-stranded circular DNA, and it belongs to the Papillomaviridae family. It is classified into high-risk and low-risk types based on its carcinogenicity [139]. HPV is one of the most common sexually transmitted pathogens worldwide, with approximately 12% of the population infected and over 6.2 million new cases reported annually [140, 141]. Sexual contact is the primary route of HPV transmission. Males not only serve as carriers but also play a crucial role as transmitters in the epidemiological chain of HPV [142]. High-risk HPV is associated with anal cancer, penile cancer, and some head and neck cancers in males [143]. However, HPV may also exist in asymptomatic males [144]. The prevalence of HPV DNA in semen has been reported to range from 0.0% to 46.2%, with an average of 17.1% (95% CI = 14.1 to 20.1%). The prevalence in fertility clinics (20.4%, 95% CI = 16.2 to 24.6%) is significantly higher than that in the general population (11.4%, 95% CI = 7.8 to 15.0%) (P < 0.001), and the prevalence of high-risk HPV (15.5%, 95% CI = 11.4 to 19.7%) is significantly higher than that of low-risk HPV (10.3%, 95% CI = 6.8 to 13.9%) (P < 0.001) [145]. Notably, the detection rate of HPV in donor sperm from sperm banks ranges from 3.1% to 16.7%, significantly impacting the clinical pregnancy rate in assisted reproduction. Therefore, it is recommended that donor sperm should be tested for HPV before being used for insemination [146].

The mechanisms by which sperm function may be impaired after HPV infection are still poorly understood. HR-HPV proteins cause inflammation and increase reactive oxygen species (ROS) levels in host cells, leading to oxidative stress (OS) [147]. Additionally, HPV infection directly inhibits the function of aquaporins (AQPs), making sperm cells more sensitive to OS. This inhibition reduces the AQP-mediated detoxification mechanism, resulting in sperm distress and impaired sperm function [148]. Research indicates that the presence of glycosaminoglycans or other soluble substances on the surface of sperm facilitates HPV attachment to the equatorial region of the sperm head [149]. When the equatorial region fuses with the oocyte plasma membrane [150], HPV may adversely affect fertilization. Certain HPV genotypes are associated with sperm DNA fragmentation [151], decreased motility and fertilization potential [152], abnormal sperm quality [19], and the formation of antisperm antibodies (ASAs) [153]. HPV transinfection may reduce the ability of trophoblasts and embryonic membranes to invade, leading to placental dysfunction and adverse pregnancy outcomes [154]. Viral genes may be transmitted to oocytes and embryo cells, causing DNA fragmentation and apoptosis, ultimately resulting in pregnancy loss [155, 156].

The guidelines issued by the European Society of Human Reproduction and Embryology (ESHRE) state that HPV in semen is a viral factor highly associated with assisted reproduction outcomes. They recommend targeted counseling for couples undergoing assisted reproductive therapies (ART) who are infected [157]. For couples with fertility problems, if they have unexplained infertility, a history of HPV infection, show related clinical manifestations, or if there is the presence of ASA and asthenospermia, HPV DNA testing and genotyping are recommended for the male partner. When HPV is detected, fluorescence in situ hybridization (FISH) analysis is recommended to check for the presence of HPV DNA on the sperm surface, and colposcopy is suggested to rule out subclinical genital lesions [158]. Since some infertile couples cannot afford to delay conception and standard washing procedures in ART are insufficient to reduce semen viral load, two potential strategies are proposed for infertile couples with HPV-infected semen: special sperm washing and HPV adjunctive vaccination. Research indicates that applying a sperm washing procedure based on hyaluronidase (IALu) helps remove HPV viral particles from the sperm surface [159]. Additionally, vaccination of infected patients has been shown to shorten the viral clearance time in semen [160], reduce the risk of recurrence [160], and improve sperm quality [144].

Herpesviruses

Members of the Herpesviridae family are enveloped spherical viruses with linear double-stranded DNA genomes, and their size ranges from 125 to 241 kbp, containing 70 to 170 genes [161]. Nine types of herpesviruses infect humans, categorized into three subfamilies: α-herpesviruses, β-herpesviruses, and γ-herpesviruses, all capable of persisting in host cells and causing recurrent infections [162]. Members of the herpesvirus family have been detected in numerous semen studies and can be transmitted through sexual intercourse or from mother to baby during pregnancy and childbirth [15, 163]. PCR assay is the preferred method for distinguishing type-specific HSV. Antiviral medications can effectively reduce the severity, duration, and recurrence of the disease, as well as prevent transmission to uninfected partners [164].

Herpes simplex virus-1/2

Herpes simplex virus-1/2 (HSV-1/2) belongs to the α-herpesvirus subfamily. HSV-1 primarily causes oral and labial herpes, while HSV-2 mainly causes genital herpes. Both HSV-1 and HSV-2 can be transmitted sexually [163], with a mother-to-child transmission rate of approximately 1:1,400–30,000, sometimes leading to life-threatening widespread infections in newborns [165].

There is significant variation in the reported infection rates of HSV-1 and HSV-2 in semen. One study revealed detection rates of 22.9% for HSV-1 and 14.3% for HSV-2 in semen samples from infertile men, with all HSV-positive samples exhibiting abnormal semen parameters [43]. Another study indicated a detection rate of HSV in 10.7%, with 7.5% positive for HSV-1 and 3.2% positive for HSV-2. HSV-1 infection was associated with decreased sperm count, while HSV-2 infection was associated with hematospermia [44]. Two other studies reported detection rates of HSV at 2.5% and 3.2%, respectively, with no observed impact on semen parameters [41, 57]. Evidence from experiments has shown that components in semen such as prostatic acid phosphatase (PAP), seminalplasmin (SEM), and seminal plasma (SP) can promote the formation of HSV particles and accelerate virus replication, indicating that semen is an important target for HSV [166]. In addition, HSV-2 can internalize into the head of sperm, potentially influencing pregnancy outcomes [167].

Varicella-zoster virus

Varicella-zoster virus (VZV) belongs to the α-herpesvirus subfamily and is responsible for causing chickenpox and shingles. Research on VZV in semen is relatively scarce. One study detected VZV in patients with teratozoospermia but did not explicitly state its correlation with the condition [45]. Other studies reported detection rates of VZV in semen ranging from 1.2% to 4%, but no association with semen quality was found [41, 48]. Additionally, some studies did not detect VZV in semen samples [57, 168, 169].

Epstein-barr virus

Epstein-barr virus (EBV) belongs to the γ-herpesvirus subfamily and is a lymphotropic herpesvirus. Infection can occur in individuals at different times, primarily through saliva transmission, but can also be transmitted through genital secretions or blood. Approximately 3% of EBV-positive mothers may transmit the virus to their babies [170]. Studies have shown that the prevalence of EBV in semen ranges from 0.4% to 45% [45, 50, 51] and may be associated with leukocytospermia [41]. Semen samples from infertile individuals have a significant capacity to induce early antigen (EA) of EBV, which may be related to reproductive damage mediated by immune responses and tumor development [171, 172].

Cytomegalovirus

Cytomegalovirus (CMV) belongs to the β-herpesvirus subfamily and is the largest and most genetically variable virus among human herpesviruses, with a global infection rate of approximately 66% to 90%. CMV can establish latency in long-lived cells and reactivate periodically, posing a risk of severe disease in immunocompromised individuals and being one of the most common causes of congenital disabilities [173]. Transmission mainly occurs through direct contact with body fluids such as saliva, urine, or semen [174].

Since CMV presence in semen was first reported in the United States in 1974 [54], relevant studies have continuously emerged. The prevalence of CMV in semen ranges approximately between 6% and 56.9% [51], and it has been associated with decreased sperm count and motility, as well as an increased failure rate in assisted reproduction [49, 52, 53]. However, some studies suggest that the presence of CMV in semen is not related to male infertility [41, 55]. It has been reported that CMV in semen can be transmitted to partners through sexual intercourse [175] and can infect the endometrium, leading to early miscarriage and fetal birth defects [176].

Human herpesvirus 6A/B

Human herpesvirus 6 (HHV-6) belongs to the β-herpesvirus subfamily and includes HHV-6A and HHV-6B. HHV-6B infects almost 100% of humans, typically before the age of 4, and is the pathogen of exanthema subitum (infant roseola). Little is known about the transmission routes and prevalence of HHV-6A, but recent research suggests a link between high levels of HHV-6A antibodies and multiple sclerosis [177]. HHV-6 typically integrates its genome into the telomeres of host cells to establish latency. Additionally, HHV-6A/B can integrate into the chromosomes of germ cells, leading to offspring carrying copies of the viral genome, a condition known as inherited chromosomally integrated HHV-6 (iciHHV-6) [178].

One study indicated a detection rate of HHV-6 in semen of 5.6% and a higher prevalence among infertile males with inflammatory diseases of the reproductive tract (19%) [49]. Two other studies reported detection rates of HHV-6 in semen at 4% and 70%, respectively, but found no correlation with semen quality [41, 57].

Human herpesvirus-7

Human herpesvirus-7 (HHV-7) belongs to the β-herpesvirus subfamily, with a global infection rate exceeding 90%. Primary infection often occurs in early childhood, manifesting with different clinical presentations such as rashes, fever, and febrile seizures [179]. There is limited research on HHV-7 in semen. One study found no evidence of HHV-7 infection in 172 semen samples [41], while another study detected only 1 case (0.4%) out of 252 semen samples [57]. Some research has detected HHV-7 in placental samples [180], but its mechanisms of sexual and mother-to-child transmission require further confirmation.

Human herpesvirus-8

Human herpesvirus-8 (HHV-8), also known as Kaposi's Sarcoma-Associated Herpesvirus (KSHV), belongs to the γ-herpesvirus subfamily and is an oncogenic pathogen that causes Kaposi's sarcoma (KS) [181]. HHV-8 primarily spreads through saliva and enters latency after cell infection, with reactivation leading to disease occurrence [182]. In the general population, the prevalence of HHV-8 is low, with most studies unable to detect HHV-8 in semen from healthy donors or infertile males [38, 57]. However, the detection rate of HHV-8 in semen from KS patients is 12% [59], and HHV-8 has also been detected in semen from HIV-1-infected individuals [62]. The prevalence of HHV-8 in semen from healthy males has not yet been determined.

Hepatitis viruses

Hepatitis B virus

Hepatitis B virus (HBV) belongs to the Hepadnaviridae family and affects nearly 400 million people worldwide with chronic hepatitis B, posing a significant global healthcare challenge [183]. It can be transmitted through body fluids such as blood, semen, and vaginal secretions. Most individuals with normal immune function can spontaneously clear the infection, but some may develop acute or chronic hepatitis or even progress to liver cirrhosis and hepatocellular carcinoma [184].

Studies have found HBV in semen [50, 68], suggesting the reproductive tract may serve as an independent reservoir for the virus, transmitting it through sexual intercourse [65]. The HBV genome can also integrate into sperm chromosomes, leading to chromosomal aberrations and even hereditary effects through vertical transmission [185]. HBV infection in semen can induce abnormal cytokine expression [186], trigger cell apoptosis, sperm DNA fragmentation, and reduce fertilization capacity [187]. HBV infection in males has been observed to decrease the success rates of assisted reproduction [188], although some studies suggest HBV positivity in semen has no impact on assisted reproduction and pregnancy outcomes [189]. In male HBV patients, sperm washing procedures can effectively reduce the risk of vertical transmission and prevent HBV from entering the oocyte during intracytoplasmic sperm injection (ICSI). Additionally, in vitro studies have shown that the risk of infected sperm cells acting as carriers in IVF is no different from that in ICSI for male HBV patients. Therefore, when the male partner is a chronic HBV carrier, there is no reason to exclude a couple from undergoing ICSI [190].

Hepatitis C virus

Hepatitis C virus (HCV) belongs to the Flaviviridae family [191] and is a major cause of liver cirrhosis and hepatocellular carcinoma. It is estimated that approximately 56.8 million people worldwide are infected with HCV, with a prevalence rate of 0.7% [192]. HCV is primarily transmitted through blood but can also be transmitted through other body fluids such as saliva, urine, and semen, indicating the possibility of sexual transmission [193]. Several studies have also demonstrated the presence of HCV in semen [72,73,74], correlating with decreased semen quality, abnormal hormone levels, and erectile dysfunction [75, 76]. Medications used to treat HCV fall into two main categories: immunomodulators and antiviral agents. However, interferon therapy can reduce male fertility due to its gonadotoxic effects [194].

Hepatitis E virus

Hepatitis E virus (HEV) belongs to the Hepeviridae family and is a common pathogen causing acute hepatitis and jaundice. Globally, around 200,000 people are infected with HEV annually, with at least five genotypes capable of human infection. HEV-1 and HEV-2 are primarily transmitted through the fecal–oral route, while HEV-3, HEV-4, and HEV-7 are often associated with zoonotic transmission and may spread through food and blood transfusions [195]. Research on HEV in the male reproductive system is limited and controversial. One study found a prevalence of HEV-4 in semen samples from infertile men, with 28.11% (52/185) of samples testing positive for HEV RNA. It was confirmed that HEV-4 infection could disrupt the blood-testis barrier (BTB), infect testicular cells, and reduce sperm quality [78]. However, other studies have failed to detect HEV in semen samples [196,197,198].

Polyomaviruses

Polyomaviruses (PV) belong to the Polyomaviridae family, a group of small, non-enveloped, double-stranded DNA viruses named for their association with various tumors. The Polyomaviridae family is divided into six genera, comprising 117 different viruses, with 14 known to infect humans [199]. The most relevant ones to human disease are the BK virus (BKPyV) and JC virus (JCPyV) [200]. These two viruses are widespread among the global population, with primary infection typically occurring in childhood or adolescence, establishing persistent or latent infections that may reactivate when the immune system is compromised. Blood, oral-fecal, urine, and sexual contact may be routes of transmission for PV [201, 202]. Studies have found the presence of JCPyV, Merkel cell polyomavirus (MCPyV), MW polyomavirus (MWPyV), STL polyomavirus (STLPyV), and Simian virus 40 (SV40) in semen [81, 85, 203]. Comar et al. first reported an infection rate of 24.5% for JCPyV in semen samples from infertile men, significantly associated with decreased sperm motility and morphological changes [82]. In addition, Rotondo et al. also identified JCPyV DNA in semen samples [83].

Mumps virus

The mumps virus (MuV) is a negative-sense RNA virus belonging to the Paramyxoviridae family. MuV is the pathogen of mumps, primarily transmitted through direct contact or respiratory droplets [204]. MuV also demonstrates a high affinity for the testes, leading to orchitis, which is a common cause of viral orchitis and male infertility [205]. In adult mumps patients, approximately 20–30% may develop orchitis, and among affected testes, 30–50% may experience testicular atrophy [206]. During the early stages of MuV infection, the virus can induce parenchymal inflammation, lymphocytic infiltration, and damage to the seminiferous tubules, thereby affecting testicular function and hormone levels [207]. A study found that MuV can be detected in patients' semen, resulting in decreased sperm count, abnormal morphology, and the production of anti-sperm antibodies, which may potentially have long-term adverse effects on fertility [86]. The diagnosis of MuV primarily relies on clinical complications and laboratory tests. The characteristic manifestation of orchitis is testicular swelling and pain. Laboratory diagnosis depends on MuV culture, viral RNA detection, or more commonly, serological confirmation by measuring immunoglobulin antibody levels [206]. MuV infections are mostly self-limiting, and there is currently no specific antiviral therapy. Treatment for mumps orchitis typically includes bed rest, scrotal support, and the use of analgesic and anti-inflammatory medications [208].

Human immunodeficiency virus

Human immunodeficiency virus (HIV) is a virus species with single-stranded positive RNA, and it belongs to the Retroviridae family. Acquired immunodeficiency syndrome (AIDS) is a major global public health concern, with an estimated 39 million people infected with HIV as of 2022. HIV is primarily transmitted through unprotected sexual intercourse, with semen serving as the primary carrier of transmission [209].

HIV infection can contaminate semen at any stage [89], existing in the form of both free viral particles and infected cells [90]. There may be genetic variations in viral RNA and DNA sequences in semen during acute and chronic infection, indicating independent viral replication in semen [91]. The impact of HIV infection on semen parameters mainly includes decreased sperm motility, reduced quantity, increased abnormal morphology, decreased ejaculate volume, elevated semen pH, and increased round cell count [92, 93]. Chronic orchitis may occur in AIDS patients, affecting testicular function and hormone production [210]. Antiretroviral therapy can reduce viral load in semen, thereby lowering the risk of transmission [211]. However, antiviral drugs also pose a risk of reducing sperm quality. This negative impact can be mitigated by using ICSI [212]. The effectiveness of sperm washing in eliminating viral particles from semen remains controversial, and improvements to washing procedures may be needed in the future [212,213,214].

Severe acute respiratory syndrome coronavirus 2

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a single-stranded positive-sense RNA virus belonging to the Coronaviridae family, primarily transmitted through respiratory droplets. The COVID-19 pandemic has led to a massive outbreak worldwide, infecting billions of people and causing millions of deaths [215].

Coronaviruses bind to host cells via the spike (S) protein and the cell receptor angiotensin-converting enzyme 2 (ACE2), and entry into host cells is facilitated by the S protein activated by type II transmembrane serine protease (TMPRSS2) [216]. The high expression of ACE2 and TMPRSS2 in the testes suggests they are target organs for SARS-CoV-2 [217]. Autopsies of individuals infected with SARS-CoV-2 revealed testicular findings such as interstitial edema, congestion, and erythrocyte extravasation. There was an increased proportion of CD3 + and CD68 + leukocytes in the testicular interstitium. Additionally, there were increases in OS, as well as elevated levels of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) [218, 219]. Studies have confirmed severe damage to the testes following SARS-CoV-2 infection, including testicular atrophy, inflammatory cell infiltration, germ cell apoptosis, and microthrombosis in testicular blood vessels [220]. Regarding the presence of SARS-CoV-2 in semen, research results vary. Some studies suggest the virus is present in the semen of infected individuals [95, 97,98,99], while others have failed to detect it [221, 222]. Most studies have shown a decline in sperm quality [100] and impaired endocrine function in the testes following SARS-CoV-2 infection [101]. Due to the limited understanding of these emerging viruses, many drugs are currently being developed and evaluated to combat and mitigate the impact of SARS-CoV-2. Vaccination is considered a crucial pathway to help end this devastating pandemic. Most studies suggest that vaccination does not affect semen parameters [223] and can reduce the incidence of orchitis and/or epididymitis [224]. Overall, the current data seem to indicate that vaccination does not negatively impact fertility in either sex [224,225,226].

Adeno-associated virus

Adeno-associated virus (AAV) belongs to the Parvoviridae family and is a type of defective virus with simple structured single-stranded DNA. AAV requires helper viruses (such as adenovirus, herpesvirus, and HPV) to participate in replication, either establishing latent infections in the absence of helpers [227] or integrating into the chromosome DNA of the host [228].

In 1999, Rohde et al. first detected AAV DNA in semen samples of infertile men, which was associated with decreased sperm vitality [104]. Erles et al. found a higher detection rate of AAV DNA (38%) in semen samples of patients with semen abnormalities compared to those with normal semen (4.6%), and AAV DNA was also detected in testicular tissue, suggesting that AAV may contribute to male infertility by interfering with sperm development [102]. Several other studies have confirmed the presence of AAV DNA in semen samples but have not found a significant correlation with fertility [48, 103].

Zika virus

Zika virus (ZIKV) is a virus species with positive-sense single-stranded RNA and belongs to the Flaviviridae family, first discovered in 1954 [229]. Infection can lead to symptoms such as rash, fever, arthritis, Guillain-Barré syndrome (GBS), myelitis, meningitis, and congenital microcephaly [230]. ZIKV can be transmitted through mosquito bites, blood transfusions, breastfeeding, vertical transmission, and sexual contact [231, 232].

ZIKV was the first arthropod-borne virus detected in human semen [109]. Subsequent studies have confirmed the presence of ZIKV in semen [110, 232]. ZIKV RNA can persist in semen for more than six months in some infected males [111]. ZIKV infection can also cause local inflammation and tissue damage in the reproductive organs, resulting in symptoms such as hematospermia, ejaculatory pain, reduced sperm count, and abnormal secretion of reproductive hormones [112, 113]. ZIKV exhibits tropism for various cell types in the reproductive tract [233], and experimental studies have confirmed that ZIKV exhibits a preference for infecting cells within the testes, resulting in cell death and disruption of the seminiferous tubules, ultimately leading to severe testicular damage and infertility [234]. ZIKV infects testicular macrophages, triggering the upregulation of various inflammatory factors. Within seminiferous tubules, sertoli cells express high levels of the Axl receptor, facilitating ZIKV entry and replication. This infection induces the overexpression of antigen presentation genes, pro-inflammatory cytokines, and transcription factors, along with the release of inflammatory cytokines and chemokines. Inhibin-B, crucial for follicle-stimulating hormone (FSH) regulation, is downregulated by ZIKV, while ZIKV increases vascular cell adhesion molecule-1 (VCAM-1) expression, aiding immune cell adhesion. ZIKV can infect various cell types within the tubules but has a lower impact on interstitial cells. Testosterone production, primarily by interstitial cells, is also disrupted by ZIKV, affecting endocrine function [235, 236]. Sexual transmission of Zika virus and persistence of the virus in male reproductive tract (MRT) are the biggest challenges to outbreak control, vaccine and antiviral drug development. DNA-based vaccination and/or ZIKV attenuated live vaccines have shown high efficacy against ZIKV-induced MRT injury in animal trials and may be an important tool for future prevention of ZIKV-induced male infertility [234]. Besides, the best way to prevent transmission is to abstain from sex or use protective measures against suspected infections [237].

Ebola virus

Ebola virus (EBOV) is a linear, non-segmented, single-stranded negative-sense RNA virus belonging to the Filoviridae family. Over 17,000 people survived the 2013–2016 Ebola virus disease outbreak in West Africa, which was the largest outbreak since the virus was first identified in 1976 [238]. This substantial number of survivors has enabled the implementation of several cross-sectional and longitudinal studies, leading to a better understanding of the long-term clinical sequelae in survivors and the persistence of the Ebola virus in biological fluids, particularly in semen [120, 121, 239].Studies have shown that the detection rate of EBOV RNA in the semen of Ebola survivors ranges from 8.1% to 9.8%, with viral shedding persisting for an extended period [240]. One cohort study found that viral RNA could remain in semen for up to 40 months [239]. Dyal et al. suggest that age, disease severity, and immune response may be risk factors for the prolonged presence of EBOV in semen [121]. Significant progress has been made in the treatment and prevention of Ebola virus disease. Currently, many vaccines and specific drugs targeting EBOV have been approved and are in use. However, there are still many issues to be addressed in reducing mortality, preventing viral escape, and improving safety [240].

Discussion

This study conducted a comprehensive and systematic analysis of the correlation between viral infections in male semen and infertility. It summarized 40 viruses found in human semen, with most viruses closely associated with male fertility. The main findings indicate that many viruses can be detected in semen, including those causing acute infections like ZIKV and EBOV, as well as those causing chronic infections like HPV and HIV. Some viruses, especially those showing strong tissue tropism for the male reproductive tract (such as SARS-CoV-2 and MuV), may spread through sexual transmission, which could be an important route of transmission. Viruses can potentially impact semen quality and fertility through various pathways, including direct infection of sperm or germ cells, disruption of testicular functions, induction of reproductive tract inflammation, and triggering immune responses.

Previous studies have investigated the impact of certain viral infections on male infertility. A meta-analysis indicated that individuals infected with HPV had decreased sperm motility, increased abnormal sperm morphology and DNA fragmentation index (DFI) compared to the uninfected group [241]. In contrast, our study included all types of viruses associated with male infertility, conducting a comprehensive analysis of the 40 viruses detected in semen, covering various virus families and potential transmission routes. This provided a comprehensive perspective on the relationship between male reproductive health and viral infections. We found the presence of some viruses, such as ZIKV and ZBOV, in the reproductive system of males and emphasized their potential impact on reproductive health, providing new directions for future research. In viral infections, changes in sperm and semen parameters can arise from (Fig. 1): (1) Genital tract inflammation: leukocyte infiltration and cytokine release. (2) Viral replication: viral replication in MGT cells alters their function and integrity, impacting sperm quality through various means like disrupted endocrine function and sperm DNA damage. (3) Systemic effects: acute infections raise testis temperature, hindering spermatogenesis. Chronic infections increase OS, harming sperm function. Altered hormone release from the pituitary reduces sperm counts by affecting testosterone and inhibin B secretion, crucial for spermatogenesis [11]. Among them, we found that OS is an important way to affect male fertility after virus infection. According to the literature, OS is responsible for 80% of male infertility cases [242]. While small amounts of reactive oxygen species (ROS) play crucial roles in sperm capacitation, acrosome reaction, and hyperactivation, excessive ROS production and OS can severely impair sperm structure and function. The sperm plasma membrane, which is rich in polyunsaturated fatty acids, is particularly susceptible to lipid peroxidation. Additionally, the sperm nuclear and mitochondrial epigenomes are vulnerable to elevated ROS levels, resulting in DNA damage through fragmentation, microdeletions, and mutations [243]. Consequently, OS induced by viral infections can degrade sperm quality, leading to reduced fertilization rates, poor embryo development, recurrent miscarriages, genetic mutations, and overall unfavorable outcomes in ART [242]. This highlights the importance of medical attention to these viruses and emphasizes the need for effective prevention and treatment strategies. Furthermore, protective measures can be developed, such as the use of antioxidants, to reduce the damage caused by oxidative stress in testicular tissues.

Fig. 1
figure 1

The mechanism of seminal parameter change caused by viral infections. A Genital tract inflammation: Leukocyte infiltration leads to fibrosis in testicular tissues; Cytokines released during inflammation trigger germ cell apoptosis and disrupt cell functions. B Viral replication: Leydig cells: viral replication affects leydig cells, altering hormone production; Sertoli cells: viral infections disrupt sertoli cells, affecting the nourishment and development of germ cells; Germ cells: direct viral damage to germ cells impacts sperm quality and DNA integrity; Macrophages: viral interactions with these immune cells induce local inflammatory responses; Others: other testicular cell types may also be affected by viral infections. C Systemic effect: Oxidative stress: chronic infections increase oxidative stress, harming sperm function; Perturbation of the hypothalamo-pituitary–gonadal axis: viral infections disrupt this axis, altering hormone release and affecting testosterone and inhibin B secretion, crucial for spermatogenesis; Fever: acute infections induce fever, raising testis temperature and impairing spermatogenesis. These mechanisms collectively explain the multifaceted impact of viral infections on male fertility, demonstrating how local and systemic effects lead to significant changes in sperm and semen parameters

However, we must recognize that many viral infections in semen (such as HHV-6, JCPyV, and BKPyV, etc.) may remain asymptomatic or latent within the host and may not necessarily lead to impaired reproductive function. Conversely, some viruses (such as influenza viruses) may never be detected in the male reproductive system [240] yet still cause apoptosis and decreased fertility due to fever or direct DNA damage [244]. Influenza virus infection can alter sperm morphology, reducing sperm count, motility, and ability to penetrate the egg [245, 246]. In addition, many infectious diseases are not caused by a single factor but rather by mixed infections involving multiple viruses, virus-bacteria co-infections, virus-fungus co-infections, or even virus-fungus-bacteria co-infections [81, 247,248,249,250]. Studies have indicated that lower viral diversity in semen samples is significantly positively correlated with successful pregnancy rates [81]. The microbiota is also highly associated with viral infections (such as HIV and HPV) [251, 252] and disease states. Therefore, considering viruses as the sole basis for disease is not entirely accurate; attention should be paid to the complex interactions in the microenvironment (including different pathogens, resident microbiota, and immune responses) and how they collectively affect reproductive function [253, 254]. Furthermore, the presence of viruses in semen may negatively impact the efficacy of ART [188, 241]. Viral infections in semen can also lead to congenital malformations and/or chronicity of viruses in fetuses [255,256,257]. Thus, even during natural conception, vertical transmission of viruses is a matter of concern [11].

Our study also has some limitations: it primarily relies on published literature and known data, which may overlook some viral infections or case reports. Although we summarized the presence of viruses in semen and their potential effects, the lack of support from large-scale clinical studies necessitates more experiments and clinical data to validate our findings.

Conclusion

Viral infections in males can potentially have detrimental effects on semen parameters and the efficacy of ART, serving as potential risk factors for male infertility. It is advisable for infertility patients, particularly those with abnormal semen parameters, to consider the potential impact of viral infections. Timely detection and treatment of viral infections are expected to enhance semen parameters and improve the success rate of ART. These insights offer valuable guidance for refining treatment strategies for male infertility patients in the future.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

COVID-19:

Coronavirus disease 2019

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus 2

HIV:

Human immunodeficiency virus

ZIKV:

Zika virus

EBOV:

Ebola virus

CHIKV:

Chikungunya virus

HPV:

Human papillomavirus

HSV-1/2:

Herpes simplex virus-1/2

VZV:

Varicella zoster virus

EBV:

Epstein-barr virus

CMV:

Cytomegalovirus

HHV-6/7:

Human herpesvirus-6/7

KSHV:

Kaposi sarcoma-associated herpesvirus

HBV:

Hepatitis B virus

HCV:

Hepatitis C virus

HDV:

Hepatitis D virus

HEV:

Hepatitis E virus

HGV:

Hepatitis G virus

JCPyV:

JC polyomavirus

MCPyV:

Merkel cell polyomavirus

MWPyV:

MW polyomavirus

BKPyV:

BK polyomavirus

SV40:

Simian virus 40

MuV:

Mumps virus

AAV:

Adeno-associated virus

MPXV:

Monkeypox virus

HTLV:

Human T-lymphotropic virus

WNV:

West-Nile virus

DENV:

Dengue virus

CV:

Coxsackie virus

MARV:

Marburg virus

RVFV:

Rift valley fever virus

Ad:

Adenovirus

CHAPV:

Chapare virus

TOSV:

Toscana virus

LASV:

Lassa virus

SFTSV:

Severe fever with thrombocytopenia syndrome virus

NiV:

Nipah virus

YFV:

Yellow fever virus

PMS:

Progressive motile sperm

MNS:

Morphologically normal sperm

pML:

Progressive multifocal leukoencephalopathy

LF:

Lassa fever

SFTS:

Severe fever with thrombocytopenia syndrome

ASAs:

Antisperm antibodies

PAP:

Prostatic acid phosphatase

SEM:

Seminalplasmin

SP:

Seminal plasma

iciHHV-6:

Inherited chromosomally integrated HHV-6

KS:

Kaposi's sarcoma

BTB:

Blood-testis barrier

PV:

Polyomaviruses

AIDS:

Acquired immunodeficiency syndrome

ACE2:

Angiotensin-converting enzyme 2

TMPRSS2:

Type II transmembrane serine protease

GBS:

Guillain-barré syndrome

DFI:

DNA fragmentation index

ART:

Assisted reproductive therapies

FISH:

Fluorescence in situ hybridization

ICSI:

Intracytoplasmic sperm injection

IALu:

Hyaluronidase

ESHRE:

European Society of Human Reproduction and Embryology

OS:

Oxidative stress

ROS:

Reactive oxygen species

AQPs:

Aquaporins

IL-6:

Interleukin-6

TNF-α:

Tumor necrosis factor-alpha

MCP-1:

Monocyte chemoattractant protein-1

FSH:

Follicle-stimulating hormone

VCAM-1:

Vascular cell adhesion molecule-1

MRT:

Male reproductive tract

References

  1. Chen T, Belladelli F, Del Giudice F, Eisenberg ML. Male fertility as a marker for health. Reprod Biomed Online. 2022;44(1):131–44.

    Article  PubMed  Google Scholar 

  2. Carson SA, Kallen AN. Diagnosis and management of infertility: a review. JAMA. 2021;326(1):65–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pourakbari R, Ahmadi H, Yousefi M, Aghebati-Maleki L. Cell therapy in female infertility-related diseases: Emphasis on recurrent miscarriage and repeated implantation failure. Life Sci. 2020;258:118181.

    Article  CAS  PubMed  Google Scholar 

  4. Agarwal A, Mulgund A, Hamada A, Chyatte MR. A unique view on male infertility around the globe. Reprod Biol Endocrinol. 2015;13:37.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Huang B, Wang Z, Kong Y, Jin M, Ma L. Global, regional and national burden of male infertility in 204 countries and territories between 1990 and 2019: an analysis of global burden of disease study. BMC Public Health. 2023;23(1):2195.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Agarwal A, Baskaran S, Parekh N, et al. Male infertility. Lancet. 2021;397(10271):319–33.

    Article  PubMed  Google Scholar 

  7. Schuppe HC, Meinhardt A, Allam JP, Bergmann M, Weidner W, Haidl G. Chronic orchitis: a neglected cause of male infertility? Andrologia. 2008;40(2):84–91.

    Article  PubMed  Google Scholar 

  8. Haidl G, Allam JP, Schuppe HC. Chronic epididymitis: impact on semen parameters and therapeutic options. Andrologia. 2008;40(2):92–6.

    Article  CAS  PubMed  Google Scholar 

  9. Henkel R, Maass G, Jung A, Haidl G, Schill WB, Schuppe HC. Age-related changes in seminal polymorphonuclear elastase in men with asymptomatic inflammation of the genital tract. Asian J Androl. 2007;9(3):299–304.

    Article  CAS  PubMed  Google Scholar 

  10. Boeri L, Pederzoli F, Capogrosso P, et al. Semen infections in men with primary infertility in the real-life setting. Fertil Steril. 2020;113(6):1174–82.

    Article  CAS  PubMed  Google Scholar 

  11. Le Tortorec A, Matusali G, Mahé D, et al. From ancient to emerging infections: the odyssey of viruses in the male genital tract. Physiol Rev. 2020;100(3):1349–414.

    Article  PubMed  Google Scholar 

  12. Rago V, Perri A. SARS-CoV-2 infection and the male reproductive system: a brief review. Life (Basel). 2023;13(2):586.

    CAS  PubMed  Google Scholar 

  13. Mukherjee AG, Wanjari UR, Gopalakrishnan AV, et al. Insights into the scenario of SARS-CoV-2 infection in male reproductive toxicity. Vaccines (Basel). 2023;11(3):510.

    Article  CAS  PubMed  Google Scholar 

  14. Shcherbitskaia AD, Komarova EM, Milyutina YP, et al. Oxidative stress markers and sperm DNA fragmentation in men recovered from COVID-19. Int J Mol Sci. 2022;23(17):10060.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dejucq N, Jégou B. Viruses in the mammalian male genital tract and their effects on the reproductive system. Microbiol Mol Biol Rev. 2001;65(2):208–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Green J, Monteiro E, Bolton VN, Sanders P, Gibson PE. Detection of human papillomavirus DNA by PCR in semen from patients with and without penile warts. Genitourin Med. 1991;67(3):207–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lai YM, Lee JF, Huang HY, Soong YK, Yang FP, Pao CC. The effect of human papillomavirus infection on sperm cell motility. Fertil Steril. 1997;67(6):1152–5.

    Article  CAS  PubMed  Google Scholar 

  18. Didelot-Rousseau MN, Diafouka F, Yayo E, Kouadio LP, Monnet D, Segondy M. HPV seminal shedding among men seeking fertility evaluation in Abidjan. Ivory Coast J Clin Virol. 2007;39(2):153–5.

    Article  PubMed  Google Scholar 

  19. Foresta C, Pizzol D, Moretti A, Barzon L, Palù G, Garolla A. Clinical and prognostic significance of human papillomavirus DNA in the sperm or exfoliated cells of infertile patients and subjects with risk factors. Fertil Steril. 2010;94(5):1723–7.

    Article  CAS  PubMed  Google Scholar 

  20. Garolla A, Pizzol D, Bertoldo A, De Toni L, Barzon L, Foresta C. Association, prevalence, and clearance of human papillomavirus and antisperm antibodies in infected semen samples from infertile patients. Fertil Steril. 2013;99(1):125-131.e2.

    Article  PubMed  Google Scholar 

  21. Yang Y, Jia CW, Ma YM, Zhou LY, Wang SY. Correlation between HPV sperm infection and male infertility. Asian J Androl. 2013;15(4):529–32.

    Article  PubMed  PubMed Central  Google Scholar 

  22. La Vignera S, Vicari E, Condorelli RA, et al. Prevalence of human papilloma virus infection in patients with male accessory gland infection. Reprod Biomed Online. 2015;30(4):385–91.

    Article  PubMed  Google Scholar 

  23. Garolla A, Engl B, Pizzol D, et al. Spontaneous fertility and in vitro fertilization outcome: new evidence of human papillomavirus sperm infection. Fertil Steril. 2016;105(1):65-72.e1.

    Article  PubMed  Google Scholar 

  24. Luttmer R, Dijkstra MG, Snijders PJ, et al. Presence of human papillomavirus in semen in relation to semen quality. Hum Reprod. 2016;31(2):280–6.

    PubMed  Google Scholar 

  25. Cortés-Gutiérrez EI, Dávila-Rodríguez MI, Fernández JL, et al. The presence of human papillomavirus in semen does not affect the integrity of sperm DNA. Andrologia. 2017;49(10). https://doi.org/10.1111/and.12774.

  26. Damke E, Kurscheidt FA, Balani VA, et al. Male partners of infertile couples with seminal infections of human papillomavirus have impaired fertility parameters. Biomed Res Int. 2017;2017:4684629.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Tangal S, Taşçı Y, Pabuçcu EG, Çağlar GS, Haliloğlu AH, Yararbaş K. DNA fragmentation index and human papilloma virus in males with previous assisted reproductive technology failures. Turk J Urol. 2018;45(1):12–6.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Bossi RL, Valadares JBF, Puerto HLD, et al. Prevalence of human papillomavirus (HPV) in the semen of patients submitted to assisted reproductive technology treatment in a private clinic in Brazil. JBRA Assist Reprod. 2019;23(3):205–9.

    PubMed  PubMed Central  Google Scholar 

  29. Depuydt CE, Donders GGG, Verstraete L, et al. Infectious human papillomavirus virions in semen reduce clinical pregnancy rates in women undergoing intrauterine insemination. Fertil Steril. 2019;111(6):1135–44.

    Article  PubMed  Google Scholar 

  30. Piroozmand A, Mousavi Nasab SD, Erami M, et al. Distribution of human papillomavirus and antisperm antibody in semen and its association with semen parameters among infertile men. J Reprod Infertil. 2020;21(3):183–8.

    PubMed  PubMed Central  Google Scholar 

  31. Depuydt C, Donders G, Verstraete L, et al. Negative impact of elevated DNA fragmentation and Human Papillomavirus (HPV) presence in sperm on the outcome of Intra-Uterine Insemination (IUI). J Clin Med. 2021;10(4):717.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kato Y, Shigehara K, Nakagawa T, et al. Human papillomavirus detected in sperm of Japanese infertile males affects reproductive parameters. Int J Infect Dis. 2021;112:294–9.

    Article  PubMed  Google Scholar 

  33. Jaworek H, Koudelakova V, Oborna I, et al. Impact of human papillomavirus infection on semen parameters and reproductive outcomes. Reprod Biol Endocrinol. 2021;19(1):156.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Capra G, Notari T, Buttà M, Serra N, Rizzo G, Bosco L. Human Papillomavirus (HPV) infection and its impact on male infertility. Life (Basel). 2022;12(11):1919.

    CAS  PubMed  Google Scholar 

  35. Borai N, Inoue M, Lefèvre C, Naumova EN, Sato B, Yamamura M. Detection of herpes simplex DNA in semen and menstrual blood of individuals attending an infertility clinic. J Obstet Gynaecol Res. 1997;23(1):17–24.

    Article  PubMed  Google Scholar 

  36. Kotronias D, Kapranos N. Detection of herpes simplex virus DNA in human spermatozoa by in situ hybridization technique. In Vivo. 1998;12(4):391–4.

    CAS  PubMed  Google Scholar 

  37. El Borai N, LeFèvre C, Inoue M, et al. Presence of HSV-1 DNA in semen and menstrual blood. J Reprod Immunol. 1998;41(1–2):137–47.

    Article  PubMed  Google Scholar 

  38. Kapranos N, Petrakou E, Anastasiadou C, Kotronias D. Detection of herpes simplex virus, cytomegalovirus, and Epstein-Barr virus in the semen of men attending an infertility clinic. Fertil Steril. 2003;79(Suppl 3):1566–70.

    Article  PubMed  Google Scholar 

  39. Bezold G, Politch JA, Kiviat NB, Kuypers JM, Wolff H, Anderson DJ. Prevalence of sexually transmissible pathogens in semen from asymptomatic male infertility patients with and without leukocytospermia. Fertil Steril. 2007;87(5):1087–97.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Wu KH, Zhou QK, Huang JH, et al. Infection of cytomegalovirus and herpes simplex virus and morphology of the infected spermatogenic cells in infertile men. Zhonghua Nan Ke Xue. 2007;13(12):1075–9.

    PubMed  Google Scholar 

  41. Neofytou E, Sourvinos G, Asmarianaki M, Spandidos DA, Makrigiannakis A. Prevalence of human herpes virus types 1–7 in the semen of men attending an infertility clinic and correlation with semen parameters. Fertil Steril. 2009;91(6):2487–94.

    Article  CAS  PubMed  Google Scholar 

  42. Klimova RR, Chichev EV, Naumenko VA, 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(1):27–31.

    CAS  PubMed  Google Scholar 

  43. Monavari SH, Vaziri MS, Khalili M, et al. Asymptomatic seminal infection of herpes simplex virus: impact on male infertility. J Biomed Res. 2013;27(1):56–61.

    Article  PubMed  Google Scholar 

  44. Kurscheidt FA, Damke E, Bento JC, et al. Effects of herpes simplex virus infections on seminal parameters in male partners of infertile couples. Urology. 2018;113:52–8.

    Article  PubMed  Google Scholar 

  45. Tavakolian S, Goudarzi H, Nazarian H, Raee P, Niakan S, Faghihloo E. The evaluation of Human papilloma virus and human herpes viruses (EBV, CMV, VZV HSV-1 and HSV-2) in semen samples. Andrologia. 2021;53(6):e14051.

    Article  CAS  PubMed  Google Scholar 

  46. Komijani M, Momeni HR, Shaykh-Baygloo N, Ghafarizadeh AA, Maleki P, Tahsili MR. Association of herpes simplex virus I&II infections with rs187084 SNP of TLR9 and male infertility. Andrologia. 2021;53(9):e14163.

    Article  CAS  PubMed  Google Scholar 

  47. Aynaud O, Poveda JD, Huynh B, Guillemotonia A, Barrasso R. Frequency of herpes simplex virus, cytomegalovirus and human papillomavirus DNA in semen. Int J STD AIDS. 2002;13(8):547–50.

    Article  PubMed  Google Scholar 

  48. Behboudi E, Mokhtari-Azad T, Yavarian J, et al. Molecular detection of HHV1-5, AAV and HPV in semen specimens and their impact on male fertility. Hum Fertil (Camb). 2019;22(2):133–8.

    Article  CAS  PubMed  Google Scholar 

  49. Naumenko V, Tyulenev Y, Kurilo L, et al. Detection and quantification of human herpes viruses types 4–6 in sperm samples of patients with fertility disorders and chronic inflammatory urogenital tract diseases. Andrology. 2014;2(5):687–94.

    Article  CAS  PubMed  Google Scholar 

  50. Afrakhteh H, Joharinia N, Momen A, et al. Relative frequency of hepatitis B virus, human papilloma virus, Epstein-Barr virus, and herpes simplex viruses in the semen of fertile and infertile men in Shiraz, Iran: A cross-sectional study. Int J Reprod Biomed. 2021;19(8):699–706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kaspersen MD, Höllsberg P. Seminal shedding of human herpesviruses. Virol J. 2013;10:226 Published 2013 Jul 8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Jahromi BN, Yaghobi R, Matlub N, et al. Prevalence of cytomegalovirus in semen of male partners of infertile couples and the virus impact on sperm parameters. J Reprod Infertil. 2020;21(2):124–9.

    PubMed  PubMed Central  Google Scholar 

  53. Mohseni M, Mollaei HR, Arabzadeh SA, Mirshekari TR, Ghorbani P. Frequency of cytomegalovirus in fertile and infertile men, referring to Afzalipour Hospital IVF Research Center, Kerman, IRAN: A case-control study. Int J Reprod Biomed. 2018;16(7):443–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lang DJ, Kummer JF, Hartley DP. Cytomegalovirus in semen. Persistence and demonstration in extracellular fluids. N Engl J Med. 1974;291(3):121–3.

    Article  CAS  PubMed  Google Scholar 

  55. Cito G, Coccia ME, Picone R, et al. Can cytomegalovirus infection affect male reproductive function? Results of a retrospective single-centre analysis. Andrologia. 2020;52(9):e13699.

    Article  CAS  PubMed  Google Scholar 

  56. Godet AN, Soignon G, Koubi H, et al. Presence of HHV-6 genome in spermatozoa in a context of couples with low fertility: what type of infection? Andrologia. 2015;47(5):531–5.

    Article  CAS  PubMed  Google Scholar 

  57. Bezold G, Schuster-Grusser A, Lange M, Gall H, Wolff H, Peter RU. Prevalence of human herpesvirus types 1–8 in the semen of infertility patients and correlation with semen parameters. Fertil Steril. 2001;76(2):416–8.

    Article  CAS  PubMed  Google Scholar 

  58. Monini P, de Lellis L, Fabris M, Rigolin F, Cassai E. Kaposi’s sarcoma-associated herpesvirus DNA sequences in prostate tissue and human semen. N Engl J Med. 1996;334(18):1168–72.

    Article  CAS  PubMed  Google Scholar 

  59. LaDuca JR, Love JL, Abbott LZ, Dube S, Freidman-Kien AE, Poiesz BJ. Detection of human herpesvirus 8 DNA sequences in tissues and bodily fluids. J Infect Dis. 1998;178(6):1610–5.

    Article  CAS  PubMed  Google Scholar 

  60. Pellett PE, Spira TJ, Bagasra O, et al. Multicenter comparison of PCR assays for detection of human herpesvirus 8 DNA in semen. J Clin Microbiol. 1999;37(5):1298–301. https://doi.org/10.1128/JCM.37.5.1298-1301.1999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Bagasra O, Patel D, Bobroski L, et al. Localization of human herpesvirus type 8 in human sperms by in situ PCR. J Mol Histol. 2005;36(6–7):401–12.

    CAS  PubMed  Google Scholar 

  62. Bobroski L, Bagasra AU, Patel D, et al. Localization of human herpesvirus type 8 (HHV-8) in the Kaposi’s sarcoma tissues and the semen specimens of HIV-1 infected and uninfected individuals by utilizing in situ polymerase chain reaction. J Reprod Immunol. 1998;41(1–2):149–60.

    Article  CAS  PubMed  Google Scholar 

  63. Hadchouel M, Scotto J, Huret JL, et al. Presence of HBV DNA in spermatozoa: a possible vertical transmission of HBV via the germ line. J Med Virol. 1985;16(1):61–6.

    Article  CAS  PubMed  Google Scholar 

  64. Karayiannis P, Novick DM, Lok AS, Fowler MJ, Monjardino J, Thomas HC. Hepatitis B virus DNA in saliva, urine, and seminal fluid of carriers of hepatitis B e antigen. Br Med J (Clin Res Ed). 1985;290(6485):1853–5.

    Article  CAS  PubMed  Google Scholar 

  65. Davison F, Alexander GJ, Trowbridge R, Fagan EA, Williams R. Detection of hepatitis B virus DNA in spermatozoa, urine, saliva and leucocytes, of chronic HBsAg carriers. A lack of relationship with serum markers of replication. J Hepatol. 1987;4(1):37–44.

    Article  CAS  PubMed  Google Scholar 

  66. Liu H, Geng CH, Wang W, et al. Effects of hepatitis B virus on human semen parameters and sperm DNA integrity. Zhonghua Nan Ke Xue. 2013;19(10):896–8.

    CAS  PubMed  Google Scholar 

  67. Fei QJ, Yang XD, Ni WH, Pan CS, Huang XF. Can hepatitis B virus DNA in semen be predicted by serum levels of hepatitis B virus DNA, HBeAg, and HBsAg in chronically infected men from infertile couples? Andrology. 2015;3(3):506–11.

    Article  CAS  PubMed  Google Scholar 

  68. Yasaghi M, Hosseini SD, Moradi A, Hassanpour M, Tabarraei A. Molecular detection of HHV-1, HHV-2, HHV-5 and HBV in semen of fertile and infertile men by multiplex PCR method. Iran J Microbiol. 2022;14(6):921–7.

    PubMed  PubMed Central  Google Scholar 

  69. Turner SS, Gianella S, Yip MJ, et al. Shedding of hepatitis C virus in semen of human immunodeficiency virus-infected men. Open Forum Infect Dis. 2016;3(2):ofw057.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Cavalheiro Nde P, Santos AC, Melo CE, Morimitsu SR, Barone AA. Hepatitis C virus detection in the semen of infected patients. Braz J Infect Dis. 2008;12(5):358–61.

    PubMed  Google Scholar 

  71. Briat A, Dulioust E, Galimand J, et al. Hepatitis C virus in the semen of men coinfected with HIV-1: prevalence and origin. AIDS. 2005;19(16):1827–35.

    Article  PubMed  Google Scholar 

  72. Bourlet T, Levy R, Maertens A, et al. Detection and characterization of hepatitis C virus RNA in seminal plasma and spermatozoon fractions of semen from patients attempting medically assisted conception. J Clin Microbiol. 2002;40(9):3252–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Cassuto NG, Sifer C, Feldmann G, et al. A modified RT-PCR technique to screen for viral RNA in the semen of hepatitis C virus-positive men. Hum Reprod. 2002;17(12):3153–6.

    Article  CAS  PubMed  Google Scholar 

  74. Bradshaw D, Lamoury F, Catlett B, et al. A comparison of seminal hepatitis C virus (HCV) RNA levels during recent and chronic HCV infection in HIV-infected and HIV-uninfected individuals. J Infect Dis. 2015;211(5):736–43.

    Article  CAS  PubMed  Google Scholar 

  75. Hofny ER, Ali ME, Taha EA, et al. Semen and hormonal parameters in men with chronic hepatitis C infection. Fertil Steril. 2011;95(8):2557–9.

    Article  CAS  PubMed  Google Scholar 

  76. Hunter SS, Gadallah A, Azawi MK, Doss W. Erectile dysfunction in patients with chronic hepatitis C virus infection. Arab J Gastroenterol. 2014;15(1):16–20.

    Article  PubMed  Google Scholar 

  77. Mansour W, Lemoine M, Neri Pinto F, et al. Markers of hepatitis delta virus infection can be detected in follicular fluid and semen. J Clin Virol. 2014;61(2):279–81.

    Article  CAS  PubMed  Google Scholar 

  78. Huang F, Long F, Yu W, et al. High prevalence of hepatitis E virus in semen of infertile male and causes testis damage. Gut. 2018;67(6):1199–201.

    Article  CAS  PubMed  Google Scholar 

  79. Hollingsworth RC, Jameson CL, Minton JE, et al. GBV-C/HGV coinfection in HIV-1-positive men: frequent detection of viral RNA in blood plasma but absence from seminal fluid plasma. J Med Virol. 1998;56(4):321–6.

    Article  CAS  PubMed  Google Scholar 

  80. Semprini AE, Persico T, Thiers V, et al. Absence of hepatitis C virus and detection of hepatitis G virus/GB virus C RNAsequences in the semen of infected men. J Infect Dis. 1998;177(4):848–54.

    Article  CAS  PubMed  Google Scholar 

  81. Gunderson S, Eskew AM, Stoutenburg D, et al. Association of the human semen DNA virome with successful in vitro fertilization. F S Sci. 2022;3(1):2–9.

    PubMed  Google Scholar 

  82. Comar M, Zanotta N, Croci E, et al. Association between the JC polyomavirus infection and male infertility. PLoS One. 2012;7(8):e42880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Rotondo JC, Candian T, Selvatici R, et al. Tracing males from different continents by genotyping JC polyomavirus in DNA from semen samples. J Cell Physiol. 2017;232(5):982–5.

    Article  CAS  PubMed  Google Scholar 

  84. Zanotta N, Delbue S, Signorini L, et al. Merkel Cell Polyomavirus Is Associated with Anal Infections in Men Who Have Sex with Men. Microorganisms. 2019;7(2):54 Published 2019 Feb 19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Barbanti-Brodano G, Trabanelli C, Lazzarin L, et al. SV40 as a possible cofactor in the etiopathogenesis of mesothelioma and other human tumors. G Ital Med Lav Ergon. 1998;20(4):218–24.

    CAS  PubMed  Google Scholar 

  86. Jalal H, Bahadur G, Knowles W, Jin L, Brink N. Mumps epididymo-orchitis with prolonged detection of virus in semen and the development of anti-sperm antibodies. J Med Virol. 2004;73(1):147–50.

    Article  CAS  PubMed  Google Scholar 

  87. Pasquier C, Walschaerts M, Raymond S, et al. Patterns of residual HIV-1 RNA shedding in the seminal plasma of patients on effective antiretroviral therapy. Basic Clin Androl. 2017;27:17.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Umapathy E, Simbini T, Chipata T, Mbizvo M. Sperm characteristics and accessory sex gland functions in HIV-infected men. Arch Androl. 2001;46(2):153–8.

    Article  CAS  PubMed  Google Scholar 

  89. López Zúñiga MÁ, Chueca N, de Salazar A, et al. Genetic diversity of HIV in seminal plasma remains higher than in blood after short-term antiretroviral therapy. Sex Transm Infect. 2020;96(5):337–41.

    Article  PubMed  Google Scholar 

  90. Anderson DJ, Politch JA, Nadolski AM, Blaskewicz CD, Pudney J, Mayer KH. Targeting Trojan Horse leukocytes for HIV prevention. AIDS. 2010;24(2):163–87.

    Article  PubMed  Google Scholar 

  91. Ghosn J, Viard JP, Katlama C, et al. Evidence of genotypic resistance diversity of archived and circulating viral strains in blood and semen of pre-treated HIV-infected men. AIDS. 2004;18(3):447–57.

    Article  CAS  PubMed  Google Scholar 

  92. Dondero F, Rossi T, D’Offizi G, et al. Semen analysis in HIV seropositive men and in subjects at high risk for HIV infection. Hum Reprod. 1996;11(4):765–8.

    Article  CAS  PubMed  Google Scholar 

  93. Nicopoullos JD, Almeida PA, Ramsay JW, Gilling-Smith C. The effect of human immunodeficiency virus on sperm parameters and the outcome of intrauterine insemination following sperm washing. Hum Reprod. 2004;19(10):2289–97.

    Article  PubMed  Google Scholar 

  94. Zhou SF, Huang C, Li SK, et al. CBPH assay for the highest sensitive detection of SARS-COV-2 in the semen. Clin Chim Acta. 2023;547:117415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Purpura LJ, Alukal J, Chong AM, et al. SARS-CoV-2 RNA shedding in semen and oligozoospermia of patient with severe coronavirus disease 11 weeks after infection. Emerg Infect Dis. 2022;28(1):196–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Saylam B, Uguz M, Yarpuzlu M, Efesoy O, Akbay E, Çayan S. The presence of SARS-CoV-2 virus in semen samples of patients with COVID-19 pneumonia. Andrologia. 2021;53(8):e14145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Machado B, Barcelos Barra G, Scherzer N, et al. Presence of SARS-CoV-2 RNA in semen-cohort study in the United States COVID-19 positive patients. Infect Dis Rep. 2021;13(1):96–101.

    Article  PubMed  PubMed Central  Google Scholar 

  98. 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(6):e2010845.

    Google Scholar 

  99. Delaroche L, Bertine M, Oger P, et al. Evaluation of SARS-CoV-2 in semen, seminal plasma, and spermatozoa pellet of COVID-19 patients in the acute stage of infection. PLoS One. 2021;16(12):e0260187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Hamarat MB, Ozkent MS, Yilmaz B, Aksanyar SY, Karabacak K. Effect of SARS-CoV-2 infection on semen parameters. Can Urol Assoc J. 2022;16(3):E173–7.

    PubMed  Google Scholar 

  101. Kadihasanoglu M, Aktas S, Yardimci E, Aral H, Kadioglu A. SARS-CoV-2 pneumonia affects male reproductive hormone levels: a prospective, Cohort Study. J Sex Med. 2021;18(2):256–64.

    Article  CAS  PubMed  Google Scholar 

  102. Erles K, Rohde V, Thaele M, Roth S, Edler L, Schlehofer JR. DNA of adeno-associated virus (AAV) in testicular tissue and in abnormal semen samples. Hum Reprod. 2001;16(11):2333–7.

    Article  CAS  PubMed  Google Scholar 

  103. Schlehofer JR, Boeke C, Reuland M, Eggert-Kruse W. Presence of DNA of adeno-associated virus in subfertile couples, but no association with fertility factors. Hum Reprod. 2012;27(3):770–8.

    Article  CAS  PubMed  Google Scholar 

  104. Rohde V, Erles K, Sattler HP, Derouet H, Wullich B, Schlehofer JR. Detection of adeno-associated virus in human semen: does viral infection play a role in the pathogenesis of male infertility? Fertil Steril. 1999;72(5):814–6.

    Article  CAS  PubMed  Google Scholar 

  105. Vogt MB, McDonald EM, Delorey M, et al. Association between prolonged shedding of Zika virus in human semen and male reproductive tract inflammation. J Infect Dis. 2022;226(7):1140–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Major CG, Paz-Bailey G, Hills SL, Rodriguez DM, Biggerstaff BJ, Johansson M. Risk estimation of sexual transmission of Zika virus-United States, 2016–2017. J Infect Dis. 2021;224(10):1756–64.

    CAS  PubMed  Google Scholar 

  107. Vanegas H, González F, Reyes Y, et al. Zika RNA and flavivirus-like antigens in the sperm cells of symptomatic and asymptomatic subjects. Viruses. 2021;13(2):152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Mahé D, Bourgeau S, Frouard J, et al. Long-term Zika virus infection of non-sperm cells in semen. Lancet Infect Dis. 2020;20(12):1371.

    Article  PubMed  Google Scholar 

  109. Guedes DR, Paiva MH, Donato MM, et al. Zika virus replication in the mosquito Culex quinquefasciatus in Brazil. Emerg Microbes Infect. 2017;6(8):e69.

    PubMed  PubMed Central  Google Scholar 

  110. Kurscheidt FA, Mesquita CSS, Damke GMZF, et al. Persistence and clinical relevance of Zika virus in the male genital tract. Nat Rev Urol. 2019;16(4):211–30.

    Article  PubMed  Google Scholar 

  111. Mead PS, Duggal NK, Hook SA, et al. Zika virus shedding in semen of symptomatic infected men. N Engl J Med. 2018;378(15):1377–85.

    Article  PubMed  Google Scholar 

  112. Paz-Bailey G, Rosenberg ES, Doyle K, et al. Persistence of Zika virus in body fluids - final report. N Engl J Med. 2017;379(13):1234–43.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Joguet G, Mansuy JM, Matusali G, et al. Effect of acute Zika virus infection on sperm and virus clearance in body fluids: a prospective observational study. Lancet Infect Dis. 2017;17(11):1200–8.

    Article  PubMed  Google Scholar 

  114. Palich R, Burrel S, Monsel G, et al. Viral loads in clinical samples of men with monkeypox virus infection: a French case series. Lancet Infect Dis. 2023;23(1):74–80.

    Article  PubMed  Google Scholar 

  115. Colavita F, Mazzotta V, Rozera G, et al. Kinetics of viral DNA in body fluids and antibody response in patients with acute Monkeypox virus infection. iScience. 2023;26(3):106102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Tan DHS, Pico Espinosa O, Matelski J, et al. Longitudinal analysis of Mpox Virus dna detectability from multiple specimen types during acute illness: a cohort study. Open Forum Infect Dis. 2024;11(2):ofae073.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Zagury D, Bernard J, Leibowitch J, et al. HTLV-III in cells cultured from semen of two patients with AIDS. Science. 1984;226(4673):449–51.

    Article  CAS  PubMed  Google Scholar 

  118. Ho DD, Schooley RT, Rota TR, et al. HTLV-III in the semen and blood of a healthy homosexual man. Science. 1984;226(4673):451–3.

    Article  CAS  PubMed  Google Scholar 

  119. Stewart GJ, Tyler JP, Cunningham AL, et al. Transmission of human T-cell lymphotropic virus type III (HTLV-III) by artificial insemination by donor. Lancet. 1985;2(8455):581–5.

    Article  CAS  PubMed  Google Scholar 

  120. Habib N, Hughes MD, Broutet N, et al. Statistical methodologies for evaluation of the rate of persistence of Ebola virus in semen of male survivors in Sierra Leone. PLoS ONE. 2022;17(10):e0274755.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Dyal J, Kofman A, Kollie JZ, et al. Risk factors for ebola virus persistence in semen of survivors in Liberia. Clin Infect Dis. 2023;76(3):e849–56.

    Article  CAS  PubMed  Google Scholar 

  122. Thorson AE, Deen GF, Bernstein KT, et al. Persistence of Ebola virus in semen among Ebola virus disease survivors in Sierra Leone: A cohort study of frequency, duration, and risk factors. PLoS Med. 2021;18(2):e1003273.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Gorchakov R, Gulas-Wroblewski BE, Ronca SE, et al. Optimizing PCR detection of west nile virus from body fluid specimens to delineate natural history in an infected human cohort. Int J Mol Sci. 2019;20(8):1934.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Mons J, Mahé-Poiron D, Mansuy JM, et al. Effects of acute dengue infection on sperm and virus clearance in body fluids of men. Emerg Infect Dis. 2022;28(6):1146–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Martins EB, Silva MFB, Tassinari WS, et al. Detection of Chikungunya virus in bodily fluids: The INOVACHIK cohort study. PLoS Negl Trop Dis. 2022;16(3):e0010242.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Bandeira AC, Campos GS, Rocha VF, et al. Prolonged shedding of Chikungunya virus in semen and urine: A new perspective for diagnosis and implications for transmission. IDCases. 2016;6:100–3.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Pilatz A, Arneth B, Kaiser R, et al. Acute orchitis deciphered: Coxsackievirus B strains are the main etiology and their presence in semen is associated with acute inflammation and risk of persistent oligozoospermia. J Med Virol. 2023;95(7):e28970.

    Article  CAS  PubMed  Google Scholar 

  128. Martini GA, Schmidt HA. Spermatogenic transmission of the “Marburg virus”. (Causes of “Marburg simian disease”). Klin Wochenschr. 1968;46(7):398–400.

    Article  CAS  PubMed  Google Scholar 

  129. Haneche F, Leparc-Goffart I, Simon F, et al. Rift Valley fever in kidney transplant recipient returning from Mali with viral RNA detected in semen up to four months from symptom onset, France, autumn 2015. Euro Surveill. 2016;21(18). https://doi.org/10.2807/1560-7917.ES.2016.21.18.30222.

  130. Csata S, Kulcsár G. Virus-host studies in human seminal and mouse testicular cells. Acta Chir Hung. 1991;32(1):83–90.

    CAS  PubMed  Google Scholar 

  131. Loayza Mafayle R, Morales-Betoulle ME, Romero C, et al. Chapare hemorrhagic fever and virus detection in rodents in Bolivia in 2019. N Engl J Med. 2022;386(24):2283–94.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Matusali G, D’Abramo A, Terrosi C, et al. Infectious Toscana virus in seminal fluid of young man returning from Elba Island. Italy Emerg Infect Dis. 2022;28(4):865–9.

    Article  PubMed  Google Scholar 

  133. Salu OB, Amoo OS, Shaibu JO, et al. Monitoring of Lassa virus infection in suspected and confirmed cases in Ondo State. Nigeria Pan Afr Med J. 2020;36:253.

    PubMed  Google Scholar 

  134. Raabe VN, Kann G, Ribner BS, et al. Favipiravir and ribavirin treatment of epidemiologically linked cases of Lassa fever. Clin Infect Dis. 2017;65(5):855–9.

    Article  PubMed  Google Scholar 

  135. McElroy AK, Akondy RS, Harmon JR, et al. A case of human lassa virus infection with robust acute T-cell activation and long-term virus-specific T-cell responses. J Infect Dis. 2017;215(12):1862–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Koga S, Takazono T, Ando T, et al. Severe fever with thrombocytopenia syndrome virus RNA in semen. Japan Emerg Infect Dis. 2019;25(11):2127–8.

    Article  PubMed  Google Scholar 

  137. Arunkumar G, Abdulmajeed J, Santhosha D, et al. Persistence of Nipah virus RNA in semen of survivor. Clin Infect Dis. 2019;69(2):377–8.

    Article  PubMed  Google Scholar 

  138. Barbosa CM, Di Paola N, Cunha MP, et al. Yellow fever virus RNA in urine and semen of convalescent patient. Brazil Emerg Infect Dis. 2018;24(1):176–8.

    Article  PubMed  Google Scholar 

  139. Zou K, Huang Y, Li Z. Prevention and treatment of human papillomavirus in men benefits both men and women. Front Cell Infect Microbiol. 2022;12:1077651.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Doorbar J, Quint W, Banks L, et al. The biology and life-cycle of human papillomaviruses. Vaccine. 2012;30(Suppl 5):F55–70.

    Article  CAS  PubMed  Google Scholar 

  141. Nicolau SM, Camargo CG, Stávale JN, et al. Human papillomavirus DNA detection in male sexual partners of women with genital human papillomavirus infection. Urology. 2005;65(2):251–5.

    Article  PubMed  Google Scholar 

  142. Castellsagué X, Bosch FX, Muñoz N. The male role in cervical cancer. Salud Publica Mex. 2003;45(Suppl 3):S345–53.

    Article  PubMed  Google Scholar 

  143. Tsai SC, Huang JY, Lin C, Liaw YP, Lin FC. The association between human papillomavirus infection and head and neck cancer: A population-based cohort study. Medicine (Baltimore). 2019;98(7):e14436.

    Article  PubMed  Google Scholar 

  144. Foresta C, Garolla A, Parisi S, et al. HPV prophylactic vaccination in males improves the clearance of semen infection. EBioMedicine. 2015;2(10):1487–93.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Lyu Z, Feng X, Li N, et al. Human papillomavirus in semen and the risk for male infertility: a systematic review and meta-analysis. BMC Infect Dis. 2017;17(1):714.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Depuydt CE, Donders G, Verstraete L, et al. Time has come to include Human Papillomavirus (HPV) testing in sperm donor banks. Facts Views Vis Obgyn. 2018;10(4):201–5.

    CAS  PubMed  Google Scholar 

  147. Pérez-Soto E, Medel-Flores MO, Fernández-Martínez E, Oros-Pantoja R, Miranda-Covarrubias JC, Sánchez-Monroy V. High-risk HPV with multiple infections promotes CYP2E1, lipoperoxidation and pro-inflammatory cytokines in semen of asymptomatic infertile men. Antioxidants (Basel). 2022;11(6):1051.

    Article  PubMed  Google Scholar 

  148. Pellavio G, Todaro F, Alberizzi P, et al. HPV infection affects human sperm functionality by inhibition of aquaporin-8. Cells. 2020;9(5):1241.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Pérez-Andino J, Buck CB, Ribbeck K. Adsorption of human papillomavirus 16 to live human sperm. PLoS One. 2009;4(6):e5847.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Anifandis G, Messini C, Dafopoulos K, Sotiriou S, Messinis I. Molecular and cellular mechanisms of sperm-oocyte interactions opinions relative to in vitro fertilization (IVF). Int J Mol Sci. 2014;15(7):12972–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Connelly DA, Chan PJ, Patton WC, King A. Human sperm deoxyribonucleic acid fragmentation by specific types of papillomavirus. Am J Obstet Gynecol. 2001;184(6):1068–70.

    Article  CAS  PubMed  Google Scholar 

  152. Lee CA, Huang CT, King A, Chan PJ. Differential effects of human papillomavirus DNA types on p53 tumor-suppressor gene apoptosis in sperm. Gynecol Oncol. 2002;85(3):511–6.

    Article  CAS  PubMed  Google Scholar 

  153. Rusz A, Pilatz A, Wagenlehner F, et al. Influence of urogenital infections and inflammation on semen quality and male fertility. World J Urol. 2012;30(1):23–30.

    Article  CAS  PubMed  Google Scholar 

  154. Gomez LM, Ma Y, Ho C, McGrath CM, Nelson DB, Parry S. Placental infection with human papillomavirus is associated with spontaneous preterm delivery. Hum Reprod. 2008;23(3):709–15.

    Article  CAS  PubMed  Google Scholar 

  155. Henneberg AA, Patton WC, Jacobson JD, Chan PJ. Human papilloma virus DNA exposure and embryo survival is stage-specific. J Assist Reprod Genet. 2006;23(6):255–9.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Noventa M, Andrisani A, Gizzo S, Nardelli GB, Ambrosini G. Is it time to shift the attention on early stages embryo development to avoid inconclusive evidence on HPV-related infertility: debate and proposal. Reprod Biol Endocrinol. 2014;12:48.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Mocanu E, Drakeley A, et al. ESHRE guideline: medically assisted reproduction in patients with a viral infection/disease. Hum Reprod Open. 2021;2021(4):hoab037.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Muscianisi F, De Toni L, Giorato G, Carosso A, Foresta C, Garolla A. Is HPV the novel target in male idiopathic infertility? A Systematic review of the literature. Front Endocrinol (Lausanne). 2021;12:643539.

    Article  PubMed  Google Scholar 

  159. De Toni L, Cosci I, Carosso A, et al. Hyaluronidase-based swim-up for semen selection in patients with human papillomavirus semen infection. Biol Reprod. 2021;104(1):211–22.

    Article  PubMed  Google Scholar 

  160. Muscianisi F, Foresta C, Garolla A. Role of HPV vaccination for prevention of male infertility. Minerva Endocrinol (Torino). 2022;47(1):70–6.

    PubMed  Google Scholar 

  161. Gatherer D, Depledge DP, Hartley CA, et al. ICTV virus taxonomy profile: herpesviridae 2021. J Gen Virol. 2021;102(10):001673.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Carneiro VCS, Pereira JG, de Paula VS. Family Herpesviridae and neuroinfections: current status and research in progress. Mem Inst Oswaldo Cruz. 2022;117:e220200.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Moore DE, Ashley RL, Zarutskie PW, Coombs RW, Soules MR, Corey L. Transmission of genital herpes by donor insemination. JAMA. 1989;261(23):3441–3.

    Article  CAS  PubMed  Google Scholar 

  164. Workowski KA, Bachmann LH, Chan PA, et al. Sexually Transmitted Infections Treatment Guidelines, 2021. MMWR Recomm Rep. 2021;70(4):1–187.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Enright AM, Prober CG. Neonatal herpes infection: diagnosis, treatment and prevention. Semin Neonatol. 2002;7(4):283–91.

    Article  PubMed  Google Scholar 

  166. Torres L, Ortiz T, Tang Q. Enhancement of herpes simplex virus (HSV) infection by seminal plasma and semen amyloids implicates a new target for the prevention of HSV infection. Viruses. 2015;7(4):2057–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Bocharova EN, Zavalishina LE, Bragina EE, et al. Detection of herpes simplex virus genomic DNA in spermatozoa of patients with fertility disorders by in situ hybridization. Dokl Biol Sci. 2007;412:82–6.

    Article  CAS  PubMed  Google Scholar 

  168. Michou V, Liarmakopoulou S, Thomas D, et al. Herpes virus infected spermatozoa following density gradient centrifugation for IVF purposes. Andrologia. 2012;44(3):174–80.

    Article  CAS  PubMed  Google Scholar 

  169. Chan DYL, Lam KKW, Lau EYL, Yeung WSB, Ng EHY. Human varicella zoster virus is not present in the semen of a man affected by chickenpox during the in vitro fertilisation of his wife. Andrologia. 2017;49(10).

  170. Meyohas MC, Maréchal V, Desire N, Bouillie J, Frottier J, Nicolas JC. Study of mother-to-child Epstein-Barr virus transmission by means of nested PCRs. J Virol. 1996;70(10):6816–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Ito Y, Tokuda H, Morigaki T, et al. Epstein-Barr virus-activating principle in human semen. Cancer Lett. 1984;23(2):129–34.

    Article  CAS  PubMed  Google Scholar 

  172. Ida K, Tokuda H, Kanaoka T, et al. Epstein-Barr virus activating principle in husbands’ semen of cervical cancer patients. Am J Reprod Immunol. 1991;26(2):89–92.

    Article  CAS  PubMed  Google Scholar 

  173. Charles OJ, Venturini C, Gantt S, et al. Genomic and geographical structure of human cytomegalovirus. Proc Natl Acad Sci U S A. 2023;120(30):e2221797120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Navti OB, Al-Belushi M, Konje JC, FRCOG. Cytomegalovirus infection in pregnancy - An update. Eur J Obstet Gynecol Reprod Biol. 2021;258:216–22.

    Article  CAS  PubMed  Google Scholar 

  175. Lisco A, Munawwar A, Introini A, et al. Semen of HIV-1-infected individuals: local shedding of herpesviruses and reprogrammed cytokine network. J Infect Dis. 2012;205(1):97–105.

    Article  CAS  PubMed  Google Scholar 

  176. Frank TS, Himebaugh KS, Wilson MD. Granulomatous endometritis associated with histologically occult cytomegalovirus in a healthy patient. Am J Surg Pathol. 1992;16(7):716–20.

    Article  CAS  PubMed  Google Scholar 

  177. Wu J, Engdahl E, Gustafsson R, et al. High antibody levels against human herpesvirus-6A interact with lifestyle factors in multiple sclerosis development. Mult Scler. 2022;28(3):383–92.

    Article  CAS  PubMed  Google Scholar 

  178. Aimola G, Beythien G, Aswad A, Kaufer BB. Current understanding of human herpesvirus 6 (HHV-6) chromosomal integration. Antiviral Res. 2020;176:104720.

    Article  CAS  PubMed  Google Scholar 

  179. Foiadelli T, Rossi V, Paolucci S, et al. Human Herpes virus 7-related encephalopathy in children. Acta Biomed. 2022;92(S4):e2021415.

    PubMed  Google Scholar 

  180. Al-Buhtori M, Moore L, Benbow EW, Cooper RJ. Viral detection in hydrops fetalis, spontaneous abortion, and unexplained fetal death in utero. J Med Virol. 2011;83(4):679–84.

    Article  PubMed  Google Scholar 

  181. Andrei G, Snoeck R. Kaposi’s sarcoma-associated herpesvirus: the role of lytic replication in targeted therapy. Curr Opin Infect Dis. 2015;28(6):611–24.

    Article  CAS  PubMed  Google Scholar 

  182. Santiago JC, Westfall DH, Adams SV, Okuku F, Phipps W, Mullins JI. Variation within major internal repeats of KSHV in vivo. Virus Evol. 2023;9(1):vead034.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Nebbia G, Peppa D, Maini MK. Hepatitis B infection: current concepts and future challenges. QJM. 2012;105(2):109–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Tripathi N, Mousa OY. Hepatitis B. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2023.

  185. Huang JM, Huang TH, Qiu HY, et al. Effects of hepatitis B virus infection on human sperm chromosomes. World J Gastroenterol. 2003;9(4):736–40.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Qian L, Li Q, Li H. Effect of hepatitis B virus infection on sperm quality and oxidative stress state of the semen of infertile males. Am J Reprod Immunol. 2016;76(3):183–5.

    Article  CAS  PubMed  Google Scholar 

  187. Han TT, Huang JH, Gu J, Xie QD, Zhong Y, Huang TH. Hepatitis B virus surface protein induces sperm dysfunction through the activation of a Bcl2/Bax signaling cascade triggering AIF/Endo G-mediated apoptosis. Andrology. 2021;9(3):944–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Oger P, Yazbeck C, Gervais A, et al. Adverse effects of hepatitis B virus on sperm motility and fertilization ability during IVF. Reprod Biomed Online. 2011;23(2):207–12.

    Article  PubMed  Google Scholar 

  189. Lee VC, Ng EH, Yeung WS, Ho PC. Impact of positive hepatitis B surface antigen on the outcome of IVF treatment. Reprod Biomed Online. 2010;21(5):712–7.

    Article  CAS  PubMed  Google Scholar 

  190. Lutgens SP, Nelissen EC, van Loo IH, Koek GH, Derhaag JG, Dunselman GA. To do or not to do: IVF and ICSI in chronic hepatitis B virus carriers. Hum Reprod. 2009;24(11):2676–8.

    Article  PubMed  Google Scholar 

  191. Li YQ, Ghafari M, Holbrook AJ, et al. The evolutionary history of hepaciviruses. Preprint. bioRxiv. 2023;2023.06.30.547218.

  192. Tang Q, Chen Z, Li H, et al. Molecular epidemiology of hepatitis C virus genotypes in different geographical regions of Chinese mainland and a phylogenetic analysis. Infect Dis Poverty. 2023;12(1):66.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Liou TC, Chang TT, Young KC, Lin XZ, Lin CY, Wu HL. Detection of HCV RNA in saliva, urine, seminal fluid, and ascites. J Med Virol. 1992;37(3):197–202.

    Article  CAS  PubMed  Google Scholar 

  194. Bukhari SA, Ahmed MM, Anjum F, et al. Post interferon therapy decreases male fertility through gonadotoxic effect. Pak J Pharm Sci. 2018;31(4(Supplementary)):1565–70.

    CAS  PubMed  Google Scholar 

  195. Songtanin B, Molehin AJ, Brittan K, Manatsathit W, Nugent K. Hepatitis E virus infections: epidemiology, genetic diversity, and clinical considerations. Viruses. 2023;15(6):1389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Wang L, Zhang Z, Shu J, et al. Absence of hepatitis E virus RNA in semen samples of infertile male in China. Gut. 2020;69(7):1363–4.

    Article  PubMed  Google Scholar 

  197. El-Mokhtar MA, Seddik MI, Osman AOB, et al. No evidence of HEV genotype 1 infections harming the male reproductive system. Virology. 2021;554:37–41.

    Article  CAS  PubMed  Google Scholar 

  198. Horvatits T, Varwig-Janssen D, Schulze Zur Wiesch J, et al. No link between male infertility and HEV genotype 3 infection. Gut. 2020;69(6):1150–1.

    Article  PubMed  Google Scholar 

  199. Bellott TR, Luz FB, Silva AKFD, Varella RB, Rochael MC, Pantaleão L. Merkel cell polyomavirus and its etiological relationship with skin tumors. An Bras Dermatol. 2023;98(6):737–49.

    Article  PubMed  PubMed Central  Google Scholar 

  200. De Gascun CF, Carr MJ. Human polyomavirus reactivation: disease pathogenesis and treatment approaches. Clin Dev Immunol. 2013;2013:373579.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Comar M, Zanotta N, Bovenzi M, Campello C. JCV/BKV and SV40 viral load in lymphoid tissues of young immunocompetent children from an area of north-east Italy. J Med Virol. 2010;82(7):1236–40.

    Article  PubMed  Google Scholar 

  202. Barbanti-Brodano G, Sabbioni S, Martini F, Negrini M, Corallini A, Tognon M. BK virus, JC virus and Simian Virus 40 infection in humans, and association with human tumors. Adv Exp Med Biol. 2006;577:319–41.

    Article  CAS  PubMed  Google Scholar 

  203. Bu X, Zhang X, Zhang X, Yi S. A study of simian virus 40 infection and its origin in human brain tumors. Zhonghua Liu Xing Bing Xue Za Zhi. 2000;21(1):19–21.

    CAS  PubMed  Google Scholar 

  204. Gavilán AM, van de Nes-Reijnen L, Castellanos A, et al. Comparison of circulation patterns of mumps virus in the Netherlands and Spain (2015–2020). Front Microbiol. 2023;14:1207500.

    Article  PubMed  PubMed Central  Google Scholar 

  205. Masarani M, Wazait H, Dinneen M. Mumps orchitis. J R Soc Med. 2006;99(11):573–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Wu H, Wang F, Tang D, Han D. Mumps orchitis: clinical aspects and mechanisms. Front Immunol. 2021;12:582946.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Wu H, Jiang X, Gao Y, et al. Mumps virus infection disrupts blood-testis barrier through the induction of TNF-α in Sertoli cells. FASEB J. 2019;33(11):12528–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Casella R, Leibundgut B, Lehmann K, Gasser TC. Mumps orchitis: report of a mini-epidemic. J Urol. 1997;158(6):2158–61.

    Article  CAS  PubMed  Google Scholar 

  209. World Health Organization. HIV - Global[EB/OL]. /2023–08–07. https://www.who.int/health-topics/hiv-aids.

  210. Teixeira TA, Oliveira YC, Bernardes FS, et al. Viral infections and implications for male reproductive health. Asian J Androl. 2021;23(4):335–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Bernard-Stoecklin S, Gommet C, Corneau AB, et al. Semen CD4+ T cells and macrophages are productively infected at all stages of SIV infection in macaques. PLoS Pathog. 2013;9(12):e1003810.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Sergouniotis F, Olofsson JI, Westling K, Rodriguez-Wallberg KA. First 15 years of assisted reproductive technology using washed sperm in HIV-positive individuals under antiretroviral therapy: Sweden’s nationwide outcomes. AIDS Patient Care STDS. 2023;37(12):566–73.

    Article  PubMed  Google Scholar 

  213. Zamora MJ, Obradors A, Woodward B, Vernaeve V, Vassena R. Semen residual viral load and reproductive outcomes in HIV-infected men undergoing ICSI after extended semen preparation. Reprod Biomed Online. 2016;32(6):584–90.

    Article  PubMed  Google Scholar 

  214. 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(4):1028–34.

    Article  PubMed  Google Scholar 

  215. WHO Coronavirus (COVID-19) Dashboard[EB/OL]. /2023–08–07. https://covid19.who.int.

  216. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and Is blocked by a clinically proven protease inhibitor. Cell. 2020;181(2):271-280.e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Qi J, Zhou Y, Hua J, et al. The scRNA-seq expression profiling of the receptor ACE2 and the cellular protease TMPRSS2 reveals human organs susceptible to SARS-CoV-2 infection. Int J Environ Res Public Health. 2021;18(1):284.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Li H, Xiao X, Zhang J, et al. Impaired spermatogenesis in COVID-19 patients. EClinicalMedicine. 2020;28:100604.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Iakymenko OA, Ramasamy R, Kryvenko ON. Testicular changes associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Arch Pathol Lab Med. 2021;145(7):781a–781.

    Article  PubMed  Google Scholar 

  220. Duarte-Neto AN, Teixeira TA, Caldini EG, et al. Testicular pathology in fatal COVID-19: A descriptive autopsy study. Andrology. 2022;10(1):13–23.

    Article  CAS  PubMed  Google Scholar 

  221. Fraietta R, de Carvalho RC, Camillo J, et al. SARS-CoV-2 is not found in human semen during mild COVID-19 acute stage. Andrologia. 2022;54(1):e14286.

    Article  CAS  PubMed  Google Scholar 

  222. Huang Z, Do DV, Beh D, et al. Effects of acute severe acute respiratory syndrome coronavirus 2 infection on male hormone profile, ACE2 and TMPRSS2 expression, and potential for transmission of severe acute respiratory syndrome coronavirus 2 in semen of Asian men. F S Sci. 2022;3(1):29–34.

    PubMed  Google Scholar 

  223. Gonzalez DC, Nassau DE, Khodamoradi K, et al. Sperm parameters before and after COVID-19 mRNA vaccination. JAMA. 2021;326(3):273–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. 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(2):e14281.

    Article  CAS  PubMed  Google Scholar 

  225. Fradkin MA, Alexandroni H, Eldar-Geva T, Ben-Ami I. IVF outcomes pre- and post-anti-COVID-19 vaccination - Are there any differences? Reprod Biol. 2024;24(2):100879.

    Article  CAS  PubMed  Google Scholar 

  226. Berkowitz HE, Vann JCJ. Strategies to address COVID-19 vaccine and pregnancy myths. MCN Am J Matern Child Nurs. 2023;48(4):215–23.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Duan D, Sharma P, Yang J, et al. Circular intermediates of recombinant adeno-associated virus have defined structural characteristics responsible for long-term episomal persistence in muscle tissue. J Virol. 1998;72(11):8568–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Kotin RM, Siniscalco M, Samulski RJ, et al. Site-specific integration by adeno-associated virus. Proc Natl Acad Sci U S A. 1990;87(6):2211–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Macnamara FN. Zika virus: a report on three cases of human infection during an epidemic of jaundice in Nigeria. Trans R Soc Trop Med Hyg. 1954;48(2):139–45.

    Article  CAS  PubMed  Google Scholar 

  230. do Rosário MS, de Jesus PA, Vasilakis N, et al. Guillain-Barré Syndrome After Zika Virus Infection in Brazil. Am J Trop Med Hyg. 2016;95(5):1157–60.

    Article  PubMed  PubMed Central  Google Scholar 

  231. Musso D, Stramer SL, AABB Transfusion-Transmitted Diseases Committee, Busch MP, International Society of Blood Transfusion Working Party on Transfusion-Transmitted Infectious Diseases. Zika virus: a new challenge for blood transfusion. Lancet. 2016;387(10032):1993–4.

    Article  PubMed  Google Scholar 

  232. Counotte MJ, Kim CR, Wang J, et al. Sexual transmission of Zika virus and other flaviviruses: A living systematic review. PLoS Med. 2018;15(7):e1002611.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Mlera L, Bloom ME. Differential Zika virus infection of testicular cell lines. Viruses. 2019;11(1):42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Almeida RDN, Braz-de-Melo HA, Santos IO, Corrêa R, Kobinger GP, Magalhaes KG. The cellular impact of the ZIKA virus on male reproductive tract immunology and physiology. Cells. 2020;9(4):1006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Govero J, Esakky P, Scheaffer SM, et al. Zika virus infection damages the testes in mice. Nature. 2016;540(7633):438–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Ma W, Li S, Ma S, et al. Zika virus causes testis damage and leads to male infertility in mice. Cell. 2017;168(3):542.

    Article  CAS  PubMed  Google Scholar 

  237. Musso D, Ko AI, Baud D. Zika virus infection - after the pandemic. N Engl J Med. 2019;381(15):1444–57.

    Article  PubMed  Google Scholar 

  238. Etard JF, Sow MS, Leroy S, et al. Multidisciplinary assessment of post-Ebola sequelae in Guinea (Postebogui): an observational cohort study. Lancet Infect Dis. 2017;17(5):545–52.

    Article  PubMed  Google Scholar 

  239. PREVAIL III Study Group, Sneller MC, Reilly C, et al. A longitudinal study of ebola sequelae in Liberia. N Engl J Med. 2019;380(10):924–34.

    Article  Google Scholar 

  240. Payne K, Kenny P, Scovell JM, Khodamoradi K, Ramasamy R. Twenty-first century viral pandemics: a literature review of sexual transmission and fertility implications in men. Sex Med Rev. 2020;8(4):518–30.

    Article  PubMed  PubMed Central  Google Scholar 

  241. Moreno-Sepulveda J, Rajmil O. Seminal human papillomavirus infection and reproduction: a systematic review and meta-analysis. Andrology. 2021;9(2):478–502.

    Article  PubMed  Google Scholar 

  242. Symeonidis EN, Evgeni E, Palapelas V, et al. Redox Balance in Male Infertility: Excellence through Moderation-"Μέτρον ἄριστον". Antioxidants (Basel). 2021;10(10):1534.

    Article  CAS  PubMed  Google Scholar 

  243. Dimitriadis F, Symeonidis EN, Tsounapi P, et al. Administration of antioxidants in infertile male: when it may have a detrimental effect? Curr Pharm Des. 2021;27(24):2796–801.

    Article  CAS  PubMed  Google Scholar 

  244. 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(4):970.e1-970.e9707.

    Article  PubMed  Google Scholar 

  245. Buch JP, Havlovec SK. Variation in sperm penetration assay related to viral illness. Fertil Steril. 1991;55(4):844–6.

    Article  CAS  PubMed  Google Scholar 

  246. MacLEOD J. Effect of chickenpox and of pneumonia on semen quality. Fertil Steril. 1951;2(6):523–33.

    Article  CAS  PubMed  Google Scholar 

  247. Jakobsen RR, Haahr T, Humaidan P, et al. Characterization of the vaginal DNA virome in health and dysbiosis. Viruses. 2020;12(10):1143.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Eskew AM, Stout MJ, Bedrick BS, et al. Association of the eukaryotic vaginal virome with prophylactic antibiotic exposure and reproductive outcomes in a subfertile population undergoing in vitro fertilisation: a prospective exploratory study. BJOG. 2020;127(2):208–16.

    Article  CAS  PubMed  Google Scholar 

  249. Flynn M, Lyall Z, Shepherd G, et al. Interactions of the bacteriome, virome, and immune system in the nose. FEMS Microbes. 2022;3:xtac020.

    Article  PubMed  PubMed Central  Google Scholar 

  250. van de Wijgert JH, Morrison CS, Cornelisse PG, et al. Bacterial vaginosis and vaginal yeast, but not vaginal cleansing, increase HIV-1 acquisition in African women. J Acquir Immune Defic Syndr. 2008;48(2):203–10.

    Article  PubMed  Google Scholar 

  251. Liu CM, Osborne BJ, Hungate BA, et al. The semen microbiome and its relationship with local immunology and viral load in HIV infection. PLoS Pathog. 2014;10(7):e1004262.

    Article  PubMed  PubMed Central  Google Scholar 

  252. Tuominen H, Rautava J, Kero K, Syrjänen S, Collado MC, Rautava S. HPV infection and bacterial microbiota in the semen from healthy men. BMC Infect Dis. 2021;21(1):373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Domínguez-Díaz C, García-Orozco A, Riera-Leal A, Padilla-Arellano JR, Fafutis-Morris M. Microbiota and its role on viral evasion: is it with us or against us? Front Cell Infect Microbiol. 2019;9:256.

    Article  PubMed  PubMed Central  Google Scholar 

  254. Stern J, Miller G, Li X, Saxena D. Virome and bacteriome: two sides of the same coin. Curr Opin Virol. 2019;37:37–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Gentile I, Borgia G. Vertical transmission of hepatitis B virus: challenges and solutions. Int J Womens Health. 2014;6:605–11.

    PubMed  PubMed Central  Google Scholar 

  256. Khodakaram-Tafti A, Farjanikish GH. Persistent bovine viral diarrhea virus (BVDV) infection in cattle herds. Iran J Vet Res. 2017;18(3):154–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  257. Duggal NK, McDonald EM, Ritter JM, Brault AC. Sexual transmission of Zika virus enhances in utero transmission in a mouse model. Sci Rep. 2018;8(1):4510.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This study was supported by the Yunnan Major Scientific and Technological Projects (202302AA310044), the National Key Research and Development Program of China (2021YFA0805703), the Kunming University of Science and Technology & the First People's Hospital of Yunnan Province Joint Special Project on Medical Research (KUST-KH2022003Z), the Yunnan Provincial Young and Middle-aged Academic and Technical Leaders Reserve Talents Program (202305AC160022), the Open project of Clinical Medical Research Center of the First People's Hospital of Yunnan Province (2022YJZX-FC16), the National Natural Science Foundation of China (32270981, 82160294,  82360304), the Joint Special Funds for the Department of Science and Technology of Yunnan Province‑Kunming Medical University (202401AY070001-301, 202301AY070001-003, 202101AY070001-260, 202301AY070001-298), Open project of Yunnan Provincial Reproductive and Obstetrics and Gynecology Clinical Medicine Center (2022LCZXKF-SZ04), Key Projects of Yunnan Province Science and Technology Department (202302AA310044).

Author information

Authors and Affiliations

Authors

Contributions

YG and YYD performed the literature search, prepared the figures, and wrote the first draft; RZZ and JCY performed the literature search and data analysis; WYL and YX wrote the first draft; XLY, YMK and YTL supervised and reviewed the manuscript; LFX: developed the idea for the article, review and editing of the final manuscript. All authors contributed to the final version of the manuscript.

Corresponding author

Correspondence to Lifeng Xiang.

Ethics declarations

Ethics approval and consent to participate

The data used in this article were all sourced from published literature. Therefore, it is not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, Y., Dong, Y., Zheng, R. et al. Correlation between viral infections in male semen and infertility: a literature review. Virol J 21, 167 (2024). https://doi.org/10.1186/s12985-024-02431-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12985-024-02431-w

Keywords