Next Article in Journal
Nutrition Genomics
Next Article in Special Issue
How Can We Prevent Mother-to-Child Transmission of HTLV-1?
Previous Article in Journal
Thalidomide Attenuates Epileptogenesis and Seizures by Decreasing Brain Inflammation in Lithium Pilocarpine Rat Model
Previous Article in Special Issue
Feline Calicivirus P39 Inhibits Innate Immune Responses by Autophagic Degradation of Retinoic Acid Inducible Gene I
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives

Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(7), 6489; https://doi.org/10.3390/ijms24076489
Submission received: 4 March 2023 / Revised: 25 March 2023 / Accepted: 28 March 2023 / Published: 30 March 2023
(This article belongs to the Special Issue The Interaction between Cell and Virus)

Abstract

:
Transforming growth factor-beta 1 (TGF-β1) is a pleiotropic growth factor playing various roles in the human body including cell growth and development. More functions of TGF-β1 have been discovered, especially its roles in viral infection. TGF-β1 is abundant at the maternal–fetal interface during pregnancy and plays an important function in immune tolerance, an essential key factor for pregnancy success. It plays some critical roles in viral infection in pregnancy, such as its effects on the infection and replication of human cytomegalovirus in syncytiotrophoblasts. Interestingly, its role in the enhancement of Zika virus (ZIKV) infection and replication in first-trimester trophoblasts has recently been reported. The above up-to-date findings have opened one of the promising approaches to studying the mechanisms of viral infection during pregnancy with links to corresponding congenital syndromes. In this article, we review our current and recent advances in understanding the roles of TGF-β1 in viral infection. Our discussion focuses on viral infection during pregnancy, especially in the first trimester. We highlight the mutual roles of viral infection and TGF-β1 in specific contexts and possible functions of the Smad pathway in viral infection, with a special note on ZIKV infection. In addition, we discuss promising approaches to performing further studies on this topic.

1. Introduction

The human body has to experience significant life changes, including pregnancy status for women. Pregnancy is a particular period not only recognized by various physical changes but also includes immune tolerance, an essential establishment of the immune system to avoid a rejection of the mother against the fetus. The placenta has an indispensable role as a barrier to protect the fetus from any possible vertical infections from the mother, addressing a balance between the tolerance of an allogeneic fetus and the protection against pathogens naturally established at the maternal–fetal interface in a healthy pregnancy [1,2]. With the placenta’s notable physical and immunological roles, pathogens, such as viruses, cannot be easily transmitted to the fetus. However, by utilizing unknown mechanisms, some viruses can still pass through the placenta in some cases [3,4,5,6,7,8].
Viral infection during pregnancy can result in congenital viral syndromes such as congenital rubella syndrome (CRS) or congenital Zika syndrome (CZS), particularly in the first trimester. CRS can occur throughout the pregnancy; however, 90% of the cases of CRS were reported as a result of a rubella virus (RuV) infection in the first trimester [9]. For Zika virus (ZIKV) infection, it is known that the risk of a structural birth defect among infants born to mothers with ZIKV infection during pregnancy ranges from 5 to 10%, with higher incidences when the infection occurs in the first trimester [10,11]. However, the mechanisms for which these viral infections affect trophoblasts, a natural barrier to prevent the fetus from being infected due to a virally infected mother, are not well understood. Trophoblasts are generally well-known for their resistance or low susceptibility to various viruses [12,13].
In a healthy pregnancy, the first trimester is characterized by a balance between the invasion of trophoblast cells and fetal-maternal immune tolerance, with the predominant role of regulatory T cells (Treg cells) being indispensably regulated by TGF-β1 [14,15,16]. TGF-β1 is a pleiotropic growth factor with various functions in cell growth and differentiation leading to the development of the human body. During pregnancy, TGF-β1 plays essential roles in cell growth and differentiation, trophoblast cell invasion, maintenance of fetal-maternal immune tolerance, and uterine spiral artery remodeling [17,18,19].
TGF-β1 also plays some roles in viral infection [20,21,22,23,24,25,26,27,28,29,30,31,32,33]. It is worthy of note that it can promote ZIKV infection in the immortalized first-trimester trophoblast cells via the Smad pathway [34]. This finding provides a potential approach to studying the mechanisms of transplacental viral infections and congenital viral syndromes. To perform more studies to understand further the role of TGF-β1 in ZIKV infection in trophoblasts and in other viral infections at the maternal–fetal interface, this review was conducted to give a comprehensive picture of the current understanding of its roles in viral infection during pregnancy. We briefly focus on the infection of the viruses of the typical ToRCH pathogens and other known potentially transplacental transmission or recently emerging viruses such as human immunodeficiency virus (HIV), influenza A virus (IAV), hepatitis B virus (HBV), ZIKV, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The pathogenesis of typical ToRCH pathogens (refers to toxoplasma, others, RuV, cytomegalovirus (CMV), and herpes simplex virus (HSV)), and ZIKV, which is included in the suggested newest ToRCHZ complex [35,36,37], as well as the above-mentioned other viruses causing vertical transmission, has been reviewed in detail elsewhere [38,39,40,41]. In this review, we also highlight the Smad pathway and its effect on ZIKV binding and replication in trophoblast cells. In addition, promising approaches to performing further studies into this topic have also been discussed.
  • Method:
Besides general information related to pregnancy and placenta, to search for available literature suitable for the title and content of this review, the following keywords were used in PubMed: “TGF-β1” or “TGFbeta1” or “transforming growth factor beta 1”, “virus”, “infection”, and “pregnancy”.
Literature about the roles of TGF-β1 in various viral infections was searched using the keyword “TGF” and one of the following virus names: “HIV”, “influenza”, “cytomegalovirus” or “CMV”, “HBV”, “rubella”, “Zika”, “HSV”, “SARS-CoV-2”. Other keywords, such as “trophoblast” or “Hofbauer,” were added to more specific topics.
For information about TGF-β1, the following keywords were used: “TGF-β1” or “transforming growth factor beta 1”, “function,” “pathway”, “pregnancy”, “placenta”, “first trimester”. Articles pertinent to this review topic were selected for discussion.

2. Trophoblasts and the Placenta Villi in Pregnancy

During pregnancy, the placenta is not only a fetal organ responsible for nutrient and gas exchange between the mother and the fetus but also a barrier to protecting the fetus against various infectious pathogens, including viruses. This review focuses on the development of trophoblasts and the placenta in the first trimester, the critical period for placental formation and fetal organogenesis.
Regarding placental formation, in the first trimester, trophoblasts, the first structure of the placenta, arise from the trophectoderm of the blastocyte, taking place at the end of the first week of conception. With a branching villous structure, the placenta contains many villi that can be divided into two types: anchoring and floating villi. Each villus contains an outer bilayer of trophoblasts functioning in nutrient exchanges and an inner core containing placental blood vessels which link to fetal circulation. The outer trophoblasts comprise two subpopulations, an outer layer of continuous multinucleated syncytiotrophoblasts (STBs) and an inner layer of mononuclear cytotrophoblasts (CTBs), which can fuse into the outer layer. For anchoring villi, the CTBs of the villous tips differentiate into extravillous trophoblasts (EVTs) that migrate out and invade into the decidua. The EVTs not only attach the placenta, anchoring it to the uterus wall, but they also invade the maternal spiral arteries and transform them into wide vessels capable of supplying significant and constant maternal blood to the fetus [2,42,43,44]. Therefore, CTBs, and especially the EVTs, have a chance to come in contact with the mother’s blood since the first trimester, while the STBs are directly exposed to maternal blood from the second trimester of pregnancy onwards. Consequently, trophoblast cells are considered the first barrier in protecting the fetus from potential infections originating from the mother’s side. For viral infection in pregnancy which results in high incident rates of a congenital viral syndrome such as RuV, HCMV, and ZIKV, research into these viral infections of first-trimester trophoblasts is considered key to opening the gate to their transplacental transmission mechanisms.

3. Viral Infections in Pregnancy

The current pandemic of SARS-CoV-2 infection has raised concerns about unfavorable impacts on maternal and fetal health, particularly in light of recent outbreaks of emerging viruses such as ZIKV. Infections in pregnancy can cause various adverse pregnancy outcomes, including premature labor, pregnancy loss, and stillbirth. In addition, once vertical transmission occurs, it is one of the significant causes of morbidity and mortality in pregnancy, leading to severe diseases in the fetus, including birth defects and congenital infection [4,45,46,47] (Table 1).
A healthy pregnancy is a tightly regulated phenomenon derived from the interconnection between the mother and the fetus. During pregnancy, immune cells from the maternal and fetal compartments interact to promote a tolerogenic milieu suitable for fetal development, providing adequate defense against pathogens. Infectious agents, especially ToRCHZ and other viral causative agents, at the maternal–fetal interface are associated with adverse pregnancy outcomes and fetal loss [86,87,88,89]. Mother-to-child transmission (MTCT) of viruses can occur through multiple routes, including direct transplacental infection with placental damage or disruption of the maternal–fetal barrier (such as CMV, RuV, ZIKV, or HIV), ascending transmission from the vaginal cervical area (HSV), transplacental immune transfer of maternal antibodies which enhances viral infection (ZIKV), perinatal transmission (HIV, HBV), postnatal transmission through breastfeeding (HCMV, HIV, HBV, HSV) [55,58,64,76,90,91,92] (Figure 1 and Table 2).

3.1. RuV

This virus belongs to the genus Rubivirus in the family Matonaviridae [111]. It often causes systemic infection in children and young adults with a clinically mild, self-limited illness with fever and a generalized erythematous maculopapular rash. However, RuV is a well-known virus in reproductive health, with a concern for potentially causing dire consequences for the fetus, although the mechanism for maternal–fetal transmission is not well established. Infection with RuV during pregnancy, especially if the infection occurs in early pregnancy, can result in various adverse pregnancy outcomes such as miscarriage, fetal death, stillbirth, or infants born with congenital disabilities including cataracts, sensorineural hearing loss, psychomotor or mental retardation, known as the CRS [48,49,112,113]. Vaccination against RuV has been available for decades, and the eradication of rubella has reached approximately half the total number of countries in the world [48]. RuV infection is still a concern as its epidemics and CRS have still occurred, leading to the recently reinforced RuV vaccination in some regions [50,114,115,116]. It is well established that for most of the CRS cases, the infection and vertical transmission occurred in the first trimester [9]. However, in vitro studies showed that RuV has low infectivity in trophoblasts and suggest that some factors may affect the infection in the first trimester [8,13,92,117].

3.2. Human Cytomegalovirus

Human cytomegalovirus (HCMV) is one member of the herpesvirus family that establishes a lifelong latency following primary infection. It is yet an under-recognized infectious cause of newborn malformation, although endemic worldwide. Over 50% of the world’s population is estimated to be infected with HCMV [118,119,120]. The mother infected with HCMV in pregnancy can transmit the virus to the fetus, causing the congenital cytomegalovirus infection, either asymptomatic or with clinical manifestations. These adverse outcomes include jaundice, hepatosplenomegaly, microcephaly, intrauterine growth restriction, psychomotor and sensorineural disabilities including hearing and vision loss, or death [53,54,55]. In developed countries, congenital cytomegalovirus infection has become the most prevalent infection-related cause of congenital neurological defects since the introduction of the universal rubella vaccination. Vertical HCMV transmission can occur during intrapartum or breastfeeding [121,122]. However, intrauterine transmission through the transplacental crossing of the virus is essential as this infection route results in greater incidences of sequelae compared with intrapartum and postnatal transmission [123,124,125]. In vitro studies reported that cytotrophoblasts were permissive to HCMV replication. Villous syncytiotrophoblasts could be permissively infected by HCMV; however, the infection required high virus titers, and the progeny virus remained predominantly cell-associated [123,126,127].

3.3. HIV

Infection with HIV results in defective cellular immunity and opportunistic infections [59]. HIV can be transmitted from a mother to her child at any time in pregnancy, during the intrauterine, intrapartum, or breastfeeding periods. However, most infants are infected during delivery. There are some factors increasing the MTCT risk, such as the absence of antiretroviral treatment during pregnancy, vaginal delivery, breastfeeding, maternal seroconversion during pregnancy or breastfeeding, high maternal plasma viral RNA load during pregnancy, and advanced maternal HIV disease [57,128]. In the absence of intervention, the rate of transmission of HIV from a mother living with HIV to her child ranges from 15% to over 40% [58,61]. With the inclusion of antiretroviral drugs during pregnancy and the choice of delivery route, the transmission rate amounted to less than 2% or even decreased to almost zero in some settings, including free access to antiretroviral therapy [60,96,129,130,131,132]. Trophoblasts are unlikely to be infected with HIV or with low viral production, and vertical transmission is thought to be through CD4+ endothelial tissues or CD4+ Hofbauer cells [98,133,134,135,136].

3.4. HBV

The Hepatitis B virus (HBV) is well-known for causing chronic hepatitis, potentially leading to cirrhosis or liver cancer. In pregnancy, HBV infection has been associated with the risk of adverse maternal and infant outcomes in a highly endemic setting but not associated with adverse pregnancy outcomes in a low-burden setting such as in the US [65,99]. Vertical transmission to newborn infants of HBV was reported and was positively correlated with the high viral load of pregnant women, especially in the third trimester [100]. It was reported that up to 85% of infants born to HBeAg-positive mothers developed chronic HBV infection [137,138]. It raises concern about the possibility of MTCT of HBV in the fetus during pregnancy and the infants’ chronic infection state in early life thereafter.
Intrauterine transmission of HBV was noted in some studies [101,139,140,141]. HBV is not cytopathogenic, and there is no evidence of placental damage caused by HBV. The findings that acute hepatitis B occurring in the first or second trimester of pregnancy rarely caused HBV infection in infants as well as the lack of anti-HBc IgM in newborn infants of HBV-infected mothers, indicating that the virus does not easily cross the placenta [139,140], suggesting the existence of a placental barrier against HBV infection. However, it was reported that HBV could infect the placental cells and not induce apoptosis, leading to HBV persistence in trophoblasts [142]. Its presence is often in low concentrations in the trophoblast plasma, and tumor necrosis factor-alpha (TNF-α) might enhance HBV replication in these cells [143]. In concordance with the above findings, clinical studies reported that intrauterine HBV infection occurred in approximately 3.7% of infants born to HBsAg-positive pregnant women [101,142]. Consequently, the principal mode of MTCT of HBV is thought to be during the intrapartum due to the rupture of the placental barrier during this period.

3.5. HSV

The infection caused by HSV is one of the most common sexually transmitted viral diseases among women of reproductive age [144]. The causative agents, HSV viruses, are enveloped double-stranded DNA viruses. HSV-1 is often discovered orally, while HSV-2 is more commonly found in genital tracts. Genital HSV infection in pregnancy was reported to be associated with spontaneous abortion, preterm labor, intrauterine growth retardation, and congenital and neonatal infections [145]. Pregnant women with primary infection may suffer from severe illness. They may likely transmit the virus to their fetus or babies, especially if the infection occurs in the latter half of pregnancy [146,147]. In addition, it is noted that primary maternal infection during the third trimester has the highest percentage of neonatal infection, and neonatal infection occurs when the fetus passes through the infected birth canal [102]. It was proved that the neonatal infection risk was reduced by caesarean section in recurrent maternal HSV infection with clinical symptoms [148,149]. It has been well established that the virus can be transmitted to the fetus in utero and cause congenital malformations such as microcephaly, microphthalmia, or hydranencephaly [150,151]. Although the syncytiotrophoblast layer is considered a barrier to maternal–fetal transmission of HSV in some studies, human trophoblasts were shown to be infected with HSVs, with the complete replicative cycle of these viruses observed in other reports, suggesting that the trophoblast layer may be involved in the mechanisms of this intrauterine HSV infection [152,153,154].

3.6. IAV

Of the three principal types of influenza viruses, influenza A and B viruses can be endemic, while only IAV is the cause of the worldwide influenza pandemic. Based on the two surface proteins, hemagglutinin (H) and neuraminidase (N), IAVs have been further classified. Pregnant women have been well-known for having increased risks for infection with both seasonal and pandemic IAV and influenza complications during the seasonal influenza periods [6,72,155,156,157]. Various adverse pregnancy outcomes such as spontaneous abortion, preterm birth, and death for pregnant women infected with IAV, were reported during the well-known influenza pandemic in 1918 [158,159] and the H1N1 influenza pandemic in 2009 [73,160,161]. In addition, influenza infection in pregnancy also might cause a slight increase in congenital deformities, but this was not consistently reported across studies [155,162]. Using a mouse model, Littauer et al. (2017)’s findings suggested that the disruption of tissue-specific hormonal regulation resulting from H1N1 IAV infection leads to preterm labor, impairment of fetal growth, increased morbidity and mortality, and maternal mortality [72]. Although rare occurrence or no viremia has often been mentioned in influenza [156,163], some studies employing highly sensitive PCR suggested transient viremia before the onset of respiratory infection is common [164,165]. In vitro studies reported that trophoblast cells were susceptible to IAV, both with H1N1 and H3N2 viruses, especially with the H3N2 virus, which could successfully replicate and induce apoptosis in the immortalized human first-trimester trophoblast cells [166,167]. However, vertical transmission of IAV appears to be rare, with no placental transmission noted in a clinical study in the second and third trimesters and in an animal model study using gilts [107,156].

3.7. ZIKV

ZIKV is a Flavivirus, a causative viral agent of a recently known CZS. This mosquito-borne infection was first noted in Yap Island in 2007, later in French Polynesia, and recently in Brazil and other parts of the Americas [168,169,170,171,172]. The infection usually presents with a mild fever, rashes, and joint pain. However, infection in pregnant women often results in severe medical and public health consequences and is likely to cause CRS, especially if the infections occur in the first trimester. The unfavorable outcomes for the fetus include microcephaly and other neurological birth defects, neurological disorders such as Guillain–Barre syndrome, or mental retardation for the fetus [10,173,174]. No vaccine for preventing ZIKV infection is available, and its development is still in progress [175,176,177,178].
Vertical transmission of ZIKV has been confirmed [76]. The virus can infect various cell types at the maternal–fetal interface, such as primary human placental cells, explants-cytotrophoblasts, endothelial cells, fibroblasts, and Hofbauer cells in chorionic villi. Maternal decidual tissues, amniotic epithelial cells, and trophoblast progenitors of amniochorionic membranes are also permissive for this virus [11,108,179,180,181,182].

3.8. SARS-CoV-2

There have been several concerns regarding the pregnancy outcomes if pregnant women are infected with SARS-CoV-2, the virus causing the current pandemic coronavirus disease of 2019 (COVID-19). Although the clinical manifestations of the infected pregnant women were not different from those of the non-pregnant, severe complications for both the mother, including preeclampsia development, and the fetus were noted for SARS-CoV-2 infection in pregnancy [46,85,183,184].
It has been reported that the vertical transmission of SARS-CoV-2 is unlikely; however, its successful transmission was noted in some cases [79,84,110,185,186,187,188]. The vertical transmission rate was estimated to be less than 3.2% [83,189,190]. Regarding mechanisms for the viral entry into susceptible cells, ACE2 is well known as a receptor for this virus. This protein is wildly expressed from the cells present at the maternal–fetal interface [191], implicating that these cells have a high chance of being infected. However, in general, the placenta barrier again works effectively to protect the fetus from this virus, and the majority of the babies born to these mothers were free from this virus infection. Clinical and in vitro evidence have demonstrated that although the placenta showed signs of infections in the invading trophoblasts and placenta, the infection seems not to go further [7,192,193]. The evidence of restricted replication of the virus in trophoblast cells was noted, suggesting that the placental barrier may be present, although not effectively, and the underlying mechanisms remain unclear [7,8].
In summary, during pregnancy, the mothers can be infected with various viral pathogens, through air-borne transmission (RuV, IAV, and SARS-CoV-2), through body fluids in sexual contact (HIV, HBV, and HSV), or in blood-borne transmission (HIV, HBV). Pregnant women can also be infected via direct contact with HSV- or CMV-infected bodily fluids, or by mosquito-borne transmission of ZIKV. Pregnant women infected with one of the above viruses can suffer from increased adverse outcomes such as miscarriage, stillbirth, or premature delivery. RuV, HCMV, and ZIKV are well known for crossing the placenta, especially in the first trimester (RuV and ZIKV), and causing congenital viral syndromes with various birth defects including the well-known microcephaly for ZIKV infection and others such as sensory loss and mental retardation. Vertical transmission during the peripartum period as well as through breastfeeding is prevalent with HIV, HBV, and HSV; especially in HBV-infected pregnant women with a high viral load in the third trimester. These two neonatal infections likely lead to liver chronic infection for HBV, or in immunocompromised status leading to AIDS if no antiretroviral therapy is received as seen in HIV. Primary HSV maternal infection during the third trimester has the highest percentage of neonatal infection, and neonatal infection occurs when the fetus passes through the infected birth canal. Among the two remaining airborne viruses, although IVA can infect trophoblasts in in vitro, a low vertical transmission rate was noted compared to other viruses such as RuV, HCMV, and ZIKV. Signs of infected placenta as well as trophoblast cells were noted with SARS-CoV-2; however, restriction replication of this virus was observed in in vitro. In addition, babies born to a mother infected with SARS-CoV-2 were often free of this virus. These above findings suggest that a placental barrier is present, although limited, to prevent the fetus from intrauterine infection. Of these eight viruses, vaccines against RuV, IVA, HBV, and SARS-CoV-2 have been available in clinical practice leading to improved pregnant outcomes. By the use of antiretroviral therapy with or without avoiding breastfeeding, vertical transmission of HIV and HBV is greatly reduced. HCMV infection is worldwide; however, it is yet an under-recognized infectious cause of newborn malformation. Since the introduction of the universal RuV vaccine, congenital HCMV has become predominant.

4. TGF-β1 and Its Essential Roles in Human Body Development and Reproductive Tract

The transforming growth factor (TGF) family, which divides into two main groups, the TGF-β family and the bone morphogenetic proteins, consists of secreted polypeptide growth factors that are involved in a variety of cellular processes such as cell growth and development, differentiation, extracellular matrix synthesis, migration, and apoptosis. The TGF-β family consists of three isoforms sharing approximately 70% sequence homology: TGF-β1, TGF-β2, and TGF-β3. They are secreted as latent complexes and require activation to bind to their receptors. TGF-β isoforms signal through a heteromeric complex of type I and type II serine/threonine kinase receptors, which initiate downstream signaling pathways involving Smad proteins [194,195,196]. TGF-β signaling is complex and plays important roles in regulating cellular processes, including wound healing, chemotaxis, and immune regulation. It is the main modulator of fibrosis upregulating collagen expression [197]. In pregnancy, TGF-β signaling plays a critical role in embryonic development and is essential for proper human fertility and reproduction [194,198,199,200,201]. Its dysregulation can lead to preterm delivery by altering tight junction expression [202,203].
Among the TGF-β family, TGF-β1, a pleiotropic growth factor secreted by many cell types, and the TGF-β receptor 1 (TβRI) serve essential roles in this family [199,204]. In pregnancy, TGF-β1 has been found abundant at the maternal–fetal interface. Immune cells, such as Treg cells, decidual macrophages, Hofbauer cells, and particularly first-trimester trophoblast cells, also secrete TGF-β1 [17,205,206,207,208,209,210]. During pregnancy, the maternal immune system is required to tolerate the presence of the fetus, which has a distinct set of antigens that can potentially trigger an immune response. TGF-β1 promotes an immunosuppressive environment at the maternal–fetal interface, allowing for successful pregnancy outcomes. It inhibits T cell activation, promotes regulatory T cell differentiation and proliferation, suppresses dendritic cell maturation, and induces apoptosis in potentially harmful immune cells. These functions collectively help to maintain the immune tolerance of the developing fetus [194,198,211,212].
Elevated concentrations of TGF-β1 in maternal plasma and placenta were noted in preeclamptic pregnancies [213,214]. TGF-β1 and TGF-β2 are the most abundant isoforms in CTB cell columns, but TGF-β1 is lower in invasive EVTs [200]. TGF-β signaling in the endometrium is active during implantation and has a pivotal role in regulating endometrial receptivity and embryo implantation. It is assumed that TGF-β is the factor that controls both apoptosis and proliferation of endometrial cells during embryo implantation. As being widely recognized as a core component of fibrosis, its potential contribution to the development of intrauterine adhesion has been mentioned [215]. Additionally, it is suggested to be involved in the pathogenesis of endometriosis by favoring the cell survival and proliferation of the ectopic endometrium [216]. Recently, more functions of TGF-β1 have been discovered, including its roles in viral infection, especially viral infection during pregnancy as mentioned earlier.

5. Roles of TGF β in Viral Infection at the Non-Maternal–Fetal Interface

Not only does the TGF-β1 play irreplaceable roles in human cell growth, differentiation, development, and immune regulation, but it also contributes different roles in various human viral infections. Enhanced expression or association with an elevated concentration of TGF-β1 in different viral infections have been thought or reported as a result of system or local immune response to protect against viral pathogens such as IAV, HIV, SARS-CoV-2, hepatitis E virus, chikungunya virus, Rift Valley Fever Virus, etc. [217,218,219,220,221,222,223]. However, its functions related to the viral infection and the viral life cycle have not always been well addressed, including the viruses of the typical ToRCH pathogens and other known potentially transplacental transmissions, or recently emerging viruses such as ZIKV and SARS-CoV-2.

5.1. RuV

Possible roles of TGF-β1 in RuV infection have been studied by our research group. It was found that in human lung cancer epithelial A549 cells, RuV infection was enhanced by TGF-β1. Although the underlying mechanisms have not been clarified, TGF-β1 induced a three- to five-fold increase in RuV binding to the treated A549 cells [20].

5.2. HCMV

It was noted that the secretion and activation of TGF-β1 are promoted in HCMV infection [224]. Although investigating the effect of TGF-β1 on HCMV replication and infection has been mentioned, a study indicated that HCMV-infected renal tubular epithelial cells could undergo EMT after exposure to TGF-β1, similar to uninfected renal epithelial cells but that HCMV infection by inducing active TGF-β1 may potentiate renal fibrosis, which helps to explain the clinical association between HCMV infection, TGF-β1, and adverse renal allograft outcomes [225]. Promotion of the activation of TGF-β1 in human umbilical vein endothelial cells by matrix metalloproteinase 2 (MMP-2) after the endothelial mesenchymal transition was addressed by Chen et al. (2019). Treatment with TGF-β1 on human umbilical vein endothelial cells infected with HCMV, can activate the extracellular potential TGF-β1 by activating MMP-2 [226]. In this research trend, another study reported that, under TGF-β1 treatment, HCMV and TGF-β1 promoted cell invasion and migration in glioma cells by the JNK pathway [227].

5.3. HIV

Elevated TGF-β1 in chronic HIV infection was often noted and suggested to contribute to immunosuppression in HIV-infected individuals [217,228,229,230,231]. It was proved that TGF-β1 could induce CXCR4 expression, a co-receptor for HIV binding, and HIV-1 entry in human monocyte-derived macrophages [24]. As an elevated TGF-β1 has often been found in patients with chronic lung diseases such as chronic obstructive pulmonary disease and asthma, its roles in HIV infection in primary differentiated human bronchial epithelial cells have recently been investigated. Chinnapaiyan et al. (2017) reported that ex vivo cultured primary bronchial epithelial cells and the bronchial brushings from human subjects which express canonical HIV receptors CD4, CCR5, and CXCR4 can be infected with HIV [232]. In addition, TGF-β1 promoting HIV latency by upregulating a transcriptional repressor BLIMP-1 (B lymphocyte-induced maturation protein-1) was further reported. These authors suggested that in patients with chronic airway diseases, TGF-β1 can elevate the HIV viral reservoir load that could further exacerbate the HIV-associated lung comorbidities [25]. Increased production of TGF-β which promotes immunosuppression was noted in HIV and also in simian immunodeficiency virus (SIV) infection. An enhanced intestinal TGF-β/Smad-dependent signaling in SIV-infected rhesus macaques was reported by Boby et al. (2021) [233].

5.4. HBV

Like other viral infections, elevated concentration and TGF-β1 production upon HBV infection have been reported in clinical and in vitro studies [27,234,235,236,237,238]. Plasma concentration of TGF-β1 was high in patients with HBV infection, especially in the first week of acute viral B hepatitis [27,235]. Subsequently, many studies investigated the role of TGF-β1 in developing hepatocellular carcinoma and its connection with liver fibrosis. Guo et al. (2009) suggested that hepatitis B virus X protein (HBx) may facilitate liver fibrosis by promoting hepatic stellate cell proliferation and upregulating the expression of fibrosis-related molecules including the TGF-β1 [236]. Although TGF-β level is thought to be an independent factor related to the occurrence of chronic HBV infection (CHB) [239], serum TGF-β1 and IL-31 were markedly higher in HBV-related liver cirrhosis (LC) patients and correlated with the severity of HBV-LC, suggesting possible roles of the TGF-β1/IL-31 pathway in the pathogenesis of liver fibrosis during CHB [237,240]. The miR-15a/Smad-7/TGF-β pathway and the TGF-β1/miR-21-5p pathway were reported to play an important role in HBV-associated liver cancer and HBV-induced liver fibrosis, respectively [29,30]. In a recent study, liver fibrogenesis promoted by HBV infection through the TGF-β1-induced OCT4/Nanog pathway has also been demonstrated [241]. Of the rare studies investigating the roles of TGF-β1 in HBV infection and replication, a study reported that TGF-β1 does not affect HBV duplication in human hepatocellular carcinoma cells HepG2.2.15 and can inhibit the expression of HBsAg and HBeA [242]. TGF-β1 was shown to suppress HBV replication effectively, and this effect was primarily through transcriptional inhibition of pregenomic RNA. The authors suggested that TGF-β1 may play a dual role in HBV infection, in the suppression of immune responses against viral infection and the direct inhibition of viral replication [28].

5.5. HSV

Early in this century, induction of the release of TGF-β1 protein was noted in in vitro infection of human mononuclear cells with HSV type 1 (HSV-1). This TGF-β1 production was highly significant after 48 h [243]. High concentrations of TGF-β1 in peripheral blood was also confirmed in patients infected with HSV-1 or HCMV and other viruses such as Varicella-zoster virus, Epstein–Barr virus, and mumps virus [244]. However, the above increased TGF-β1 production seems to be cell type-dependent, as its suppressive expression was found in HSV-1-infected human corneal epithelial cells and HSV-1-infected human trabecular meshwork cells [245,246]. On the other hand, TGF-β signaling results in increased HSV-1 latency in a mouse model [22]. TGF-β1 exposure enhances HSV-1 replication along with a significant reduction in CXCL10 expression in 3-dimensional human corneal keratocyte cultures [247].

5.6. IAV

An elevated concentration of TGF-β1 was noted in severe cases of influenza A H1N1 infection in an early report [248]. A reciprocal TGF-β1–integrin crosstalk regulated by the immune adapter ADAP (Adhesion and Degranulation-promoting Adapter Protein) is suggested to play a protective role against influenza infection [249]. An increase in TGF-β1 was also noted in nasal mucosal lining fluid collected from neonates of mothers receiving A (H1N1) pnd09 vaccination during pregnancy [250]. In the mucosal immune response, the role of epithelial-derived TGF-β1 in suppressing early interferon β responses leading to increased viral burden and pathology was noted [21]. On the other hand, pre-treatment of TGF-β1 significantly inhibited apoptosis and the presence of proapoptotic factors of H1N1-infected A549 cells in an in vitro study [251].

5.7. ZIKV

There has been a limited number of published articles pertaining to this review topic. It has been suggested that TGF-β1 may play a role in the immune response and pathogenesis of ZIKV infection. ZIKV infection induced increased expression of TGF-β along with other proinflammatory and anti-inflammatory cytokines in the neural parenchyma in fetal cases of microcephaly [252]. In recent reports, TGF-β1 does not affect the replication of ZIKV in Setoli cells in an in vitro study using a multiplicity of infection (MOI) of one [253]. However, as mentioned earlier, it does increase ZIKV replication in the first-trimester trophoblast cells at the maternal–fetal interface.

5.8. SARS-CoV-2

In the current COVID-19 pandemic, which has caused many severe death cases for people worldwide since the beginning, changes in TGF-β1 expression at SARS-CoV-2 targeted tissues and its concentration in serum samples have been investigated. Studies have reported a low mRNA expression of TGF-β1 at the mRNA level in the early inflammatory response in upper airway samples [33] or no elevated concentration of TGF-β1 in blood samples at diagnosis of COVID-19 by PCR suggesting no help to anticipate long-term prognosis [254]. However, upregulation of TGF-β1 was often reported, especially in COVID-19 cases associated with lung injury. Serum levels of TGF-β1 were significantly increased at the early and middle stages of COVID-19 and correlated with the levels of SARS-CoV-2-specific IgA [32]. In an immunohistochemical analysis study, using paraffin lung samples from patients who died of COVID-19, a significant increase in the immunoexpression of TGF-β1 was observed compared to control groups. Recently, Laloglu and Alay (2022) reported that in patients with confirmed COVID-19 and pulmonary involvement, along with elevated connective tissue growth factor levels, significantly high concentrations of TGF-β1 in serum samples were noted, especially in more severe pneumonia groups [255]. The authors suggested that TGF-β1 is one of the potential markers that can distinguish COVID-19 patients with pulmonary involvement and indicate disease severity. TGF-β1 has been suggested to play some crucial roles in SARS-CoV-2 infection. Subsequently, the use of some TGF-β1 inhibitors has been proposed to mitigate the current COVID-19 pandemic [31,256].
The reported possible roles of TGF-β1 in various viral infections including those mentioned above, have been summarized in Table 3 and Table 4.

6. TGF-β1 and Viral Infection at the Maternal–Fetal Interface

Although there have been several studies on the topic of TGF-β1 and viral infection as mentioned above, reports addressing the possible roles of TGF-β1 in viral infection at the maternal–fetal interface are quite limited. Quite a few published studies conducted in in vitro with relevant contents reported the TGF-β1 roles in viral infection in trophoblast cells, the critical barrier in protecting the fetus from maternal viral infection (Figure 2 and Table 4).

6.1. HCMV

At the end of the last century, Bacsi et al. (1999) reported a necessary contact of placental macrophage for HCMV replication in STBs [26]. TGF-β1 and interleukin-8 (IL-8), which are released from placental macrophages, promote the complete replicative cycle of HCMV in the studied STBs. The findings indicated an interactive role for the STB layer and placental macrophages in the dissemination of HCMV among placental tissue, contributing to the transmission of HCMV from the mother to the fetus. On the other hand, a recent study using EVTs isolated from early placentae reported an elevated level of TGF-β1 protein in HCMV-infected EVT cells at 48 h postinfection, suggesting a role of HCMV in the proliferation and invasion of these EVT cells [259].

6.2. HIV

Early in this century, Zachar et al. (2002) tested the roles of various cytokines and growth factors (including TGF-β1) typically present in the placental micro-environment in HIV infection in trophoblasts. Unlike the following four cytokines, epidermal growth factor (EGF), granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-1β, and TNF-α, which showed stimulation of promoters of tested HIV viruses, no effect on the transcriptional expression of the promoter constructs was noted with the TGF-β1 [257].

6.3. HBV

In an in vitro model, Cui et al. (2015) induced a multifunctional viral regulator of HBV gene products, HBx, and its different fragments to overexpress in a trophoblast cell line, HTR-8/SVneo. The authors reported that TGF-β1 decreases HTR-8/SVneo cell proliferation and invasion while increasing HBx-transfected HTR-8/SVneo cell proliferation and invasion [258].

6.4. ZIKV

Recently, our research group presented an in vitro study testing possible roles of TGF-β1 in ZIKV infection in the immortalized human first-trimester trophoblast cells Swan. 71. By using enough MOI to assure every single cell has a chance to come in contact with at least one virus particle theoretically. The results showed an enhancement in ZIKV binding and replication in these trophoblast cells. In addition, such enhancement effects were abolished using an inhibitor of the Smad pathway, SB431542 or SB525334 [34].

7. The Smad Pathway and Promising Future Approaches

One of the major downstream signaling pathways of TGF-β1 is the Smad pathway. Smads are intracellular signaling proteins that transduce TGF-β signaling from the cell surface to the nucleus, where they regulate gene expression. The Smad pathway is initiated by activating and binding TGF-β1 to its receptors, TβRI and TβRII, resulting in the phosphorylation of receptor-regulated Smad2/3 proteins [260]. This forms complexes with other Smad proteins (Smad4) and translocate to the nucleus where they regulate gene transcription in a cell-specific manner (Figure 3).
To date, besides studies reporting the elevated concentration or an enhancement of TGF-β1 as a systemic response or locally from infected tissues, several studies investigated the roles of TGF-β1 in viral infection and replication. It has been shown that TGF-β1 could promote the infection and replication of some viruses as mentioned earlier, and in some cases their underlying mechanisms are still being investigated. It is thought that TGF-β1 might enhance the non-specific binding of a given virus to extracellular matrix proteins, which play a cofactor supporting the viral entry. There is also a possibility that TGF-β1 promotes viral infection via effects on the expression of moonlighting proteins. A moonlighting protein is a protein that has multiple functions in addition to its primary role. Heat-shock proteins (HSP) such as HSP90, glucose-regulated protein 94 (GRP94), and GRP78 may have other functions such as signal transduction, immunoregulation, and especially, working as cellular receptors for some viruses, in addition to their primary roles as chaperone proteins in assisting in protein folding and refolding within the endoplasmic reticulum [261,262,263,264,265,266,267,268]. Other moonlighting proteins, such as annexin A2 and cyclophilin A, have been shown to play roles in the entry of several viruses, including HIV, IAV, and SARS-CoV-2 [269,270,271,272,273]. These proteins can interact with the viral envelope proteins and serve as cellular cofactors supporting the viral entry. Further studies are necessary to clarify these mechanisms in viral infection during pregnancy.
It is noteworthy that TGF-β1 could induce enhanced expression of cellular receptors for viral entry via the Smad pathway. In recent SARS-CoV-2 research, Mezger and colleagues reported that activation of the Smad pathway via TGF-β1 or ALK5 agonists led to increased expression of furin, a protease that cleaves the spike protein of SARS-CoV-2, in a broad spectrum of cells including Huh-7 (a permanent cell line established from male hepatoma tissue), and Calu-3 cells (epithelial cells isolated from lung tissue derived from a male patient with lung adenocarcinoma), which enhanced susceptibility to SARS-CoV-2 infection in these cells [256]. Expanding the above findings to further studies to investigate possible mechanisms of transplacental infection of the SARS-CoV-2 is highly recommended.
One of the reported studies addressing the role of TGF-β1 in viral infection in pregnancy is our recent work on Zika virus infection in trophoblast cells with the predominant role of the Smad pathway. The enhancement effect of ZIKV induced by TGF-β1 might be attributed to an increase in cellular receptors of ZIKV, AXL and Tyro3 [34]. In addition, increasing extracellular matrix synthesis resulting from the Smad pathway leading to an enhancement of non-specific binding of ZIKV to the trophoblast cells may not be excluded. Studying possible activated or moonlighting proteins as downstream results of the Smad pathway turning into cellular receptors for ZIKV can be paid more attention to. Possible downstream factors of the Smad pathway leading to an increase in ZIKV replication in trophoblast cells should also be investigated. Importantly, the findings on the effects of TGF-β1 in viral infection and replication were extracted from in vitro experiments, which may differ from those of actual intrauterine in vivo experiments. Therefore, expanding the experiments to other trophoblast cells, ex vivo using explant cultures or mouse model, is indeed necessary. Lastly, it has been known that, in pregnancy, a complex cytokine network is present at the maternal–fetal interface to support normal growth and development of the placenta and fetus. Therefore, not only the TGF-β1 but the possible roles of other cytokines and factors present at the maternal–fetal interface should also be studied.

Author Contributions

Conceptualization, Q.D.T., S.K.-A., H.U. and S.H.; methodology, Q.D.T. and K.T.; writing—original draft preparation, Q.D.T. and N.T.K.P.; writing—review and editing, Q.D.T., N.T.K.P., K.T.; H.U., S.K.-A. and S.H.; supervision, S.H. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Grants-in-Aid for Scientific Research under the Japan Society for the Promotion of Science (JSPS KAKENHI), grant numbers 17H04341 (to S.H.), 20K08829 (to Q.D.T.), and is partially supported by the Nihon University Research Grant for 2022 (to S.H.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

TGF-β1Transforming growth factor-beta 1
CRSCongenital rubella syndrome
CZSCongenital Zika syndrome
MTCTMother-to-child transmission
RuVRubella virus
HCMVHuman cytomegalovirus
HIVHuman immunodeficiency virus
HBVHepatitis B virus
HSVHerpes simplex virus
IAVInfluenza A virus
ZIKVZika virus
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
SIVSimian immunodeficiency virus
STBSyncytiotrophoblasts
CTBCytotrophoblasts
EVTExtravillous trophoblasts
MOIMultiplicity of infection
HBxHepatitis B virus X protein
HSPHeat-shock proteins
GRPGlucose-regulated protein
COVID-19Coronavirus disease of 2019
ToRCHToxoplasma, others, rubella, cytomegalovirus, herpes simplex virus

References

  1. Mor, G. Introduction to the immunology of pregnancy. Immunol. Rev. 2022, 308, 5–8. [Google Scholar] [CrossRef] [PubMed]
  2. Boss, A.L.; Chamley, L.W.; James, J.L. Placental formation in early pregnancy: How is the centre of the placenta made? Hum. Reprod. Update 2018, 24, 750–760. [Google Scholar] [CrossRef] [PubMed]
  3. Cornish, E.F.; Filipovic, I.; Asenius, F.; Williams, D.J.; McDonnell, T. Innate immune responses to acute viral infection during pregnancy. Front. Immunol. 2020, 11, 572567. [Google Scholar] [CrossRef] [PubMed]
  4. Alberca, R.W.; Pereira, N.Z.; Oliveira, L.; Gozzi-Silva, S.C.; Sato, M.N. Pregnancy, viral infection, and COVID-19. Front. Immunol. 2020, 11, 1672. [Google Scholar] [CrossRef]
  5. Racicot, K.; Mor, G. Risks associated with viral infections during pregnancy. J. Clin. Investig. 2017, 127, 1591–1599. [Google Scholar] [CrossRef] [Green Version]
  6. Silasi, M.; Cardenas, I.; Kwon, J.Y.; Racicot, K.; Aldo, P.; Mor, G. Viral infections during pregnancy. Am. J. Reprod. Immunol. 2015, 73, 199–213. [Google Scholar] [CrossRef] [Green Version]
  7. Takada, K.; Shimodai-Yamada, S.; Suzuki, M.; Trinh, Q.D.; Takano, C.; Kawakami, K.; Asai-Sato, M.; Komatsu, A.; Okahashi, A.; Nagano, N.; et al. Restriction of SARS-CoV-2 replication in the human placenta. Placenta 2022, 127, 73–76. [Google Scholar] [CrossRef]
  8. Komine-Aizawa, S.; Takada, K.; Hayakawa, S. Placental barrier against COVID-19. Placenta 2020, 99, 45–49. [Google Scholar] [CrossRef]
  9. Hobman, T.C. Rubella virus. In Fields Virology, 6th ed.; Knipe, D.M., Howley, P., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2013; Volume 1, pp. 687–711. [Google Scholar]
  10. Rasmussen, S.A.; Jamieson, D.J. Teratogen update: Zika virus and pregnancy. Birth Defects Res. 2020, 112, 1139–1149. [Google Scholar] [CrossRef]
  11. Sheridan, M.A.; Yunusov, D.; Balaraman, V.; Alexenko, A.P.; Yabe, S.; Verjovski-Almeida, S.; Schust, D.J.; Franz, A.W.; Sadovsky, Y.; Ezashi, T.; et al. Vulnerability of primitive human placental trophoblast to zika virus. Proc. Natl. Acad. Sci. USA 2017, 114, E1587–E1596. [Google Scholar] [CrossRef] [Green Version]
  12. Bayer, A.; Delorme-Axford, E.; Sleigher, C.; Frey, T.K.; Trobaugh, D.W.; Klimstra, W.B.; Emert-Sedlak, L.A.; Smithgall, T.E.; Kinchington, P.R.; Vadia, S.; et al. Human trophoblasts confer resistance to viruses implicated in perinatal infection. Am. J. Obstet. Gynecol. 2015, 212, 71.e1–71.e8. [Google Scholar] [CrossRef] [Green Version]
  13. Pham, N.T.K.; Trinh, Q.D.; Takada, K.; Komine-Aizawa, S.; Hayakawa, S. Low susceptibility of rubella virus in first-trimester trophoblast cell lines. Viruses 2022, 14, 1169. [Google Scholar] [CrossRef]
  14. Du, M.R.; Guo, P.F.; Piao, H.L.; Wang, S.C.; Sun, C.; Jin, L.P.; Tao, Y.; Li, Y.H.; Zhang, D.; Zhu, R.; et al. Embryonic trophoblasts induce decidual regulatory t cell differentiation and maternal–fetal tolerance through thymic stromal lymphopoietin instructing dendritic cells. J. Immunol. 2014, 192, 1502–1511. [Google Scholar] [CrossRef] [Green Version]
  15. Ramhorst, R.; Fraccaroli, L.; Aldo, P.; Alvero, A.B.; Cardenas, I.; Leiros, C.P.; Mor, G. Modulation and recruitment of inducible regulatory t cells by first trimester trophoblast cells. Am. J. Reprod. Immunol. 2012, 67, 17–27. [Google Scholar] [CrossRef] [Green Version]
  16. Yang, D.; Dai, F.; Yuan, M.; Zheng, Y.; Liu, S.; Deng, Z.; Tan, W.; Chen, L.; Zhang, Q.; Zhao, X.; et al. Role of transforming growth factor-beta1 in regulating fetal-maternal immune tolerance in normal and pathological pregnancy. Front. Immunol. 2021, 12, 689181. [Google Scholar] [CrossRef]
  17. Graham, C.H.; Lysiak, J.J.; McCrae, K.R.; Lala, P.K. Localization of transforming growth factor-beta at the human fetal-maternal interface: Role in trophoblast growth and differentiation. Biol. Reprod. 1992, 46, 561–572. [Google Scholar] [CrossRef] [Green Version]
  18. Ingman, W.V.; Robertson, S.A. The essential roles of tgfb1 in reproduction. Cytokine Growth Factor Rev. 2009, 20, 233–239. [Google Scholar] [CrossRef]
  19. Zhao, M.R.; Qiu, W.; Li, Y.X.; Zhang, Z.B.; Li, D.; Wang, Y.L. Dual effect of transforming growth factor beta1 on cell adhesion and invasion in human placenta trophoblast cells. Reproduction 2006, 132, 333–341. [Google Scholar] [CrossRef] [Green Version]
  20. Pham, N.T.K.; Trinh, Q.D.; Takada, K.; Takano, C.; Sasano, M.; Okitsu, S.; Ushijima, H.; Komine-Aizawa, S.; Hayakawa, S. The epithelial-to-mesenchymal transition-like process induced by tgf-beta1 enhances rubella virus binding and infection in a549 cells via the smad pathway. Microorganisms 2021, 9, 662. [Google Scholar] [CrossRef]
  21. Denney, L.; Branchett, W.; Gregory, L.G.; Oliver, R.A.; Lloyd, C.M. Epithelial-derived tgf-beta1 acts as a pro-viral factor in the lung during influenza a infection. Mucosal. Immunol. 2018, 11, 523–535. [Google Scholar] [CrossRef] [Green Version]
  22. Allen, S.J.; Mott, K.R.; Wechsler, S.L.; Flavell, R.A.; Town, T.; Ghiasi, H. Adaptive and innate transforming growth factor beta signaling impact herpes simplex virus 1 latency and reactivation. J. Virol. 2011, 85, 11448–11456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Esaki, S.; Nigim, F.; Moon, E.; Luk, S.; Kiyokawa, J.; Curry, W., Jr.; Cahill, D.P.; Chi, A.S.; Iafrate, A.J.; Martuza, R.L.; et al. Blockade of transforming growth factor-beta signaling enhances oncolytic herpes simplex virus efficacy in patient-derived recurrent glioblastoma models. Int. J. Cancer 2017, 141, 2348–2358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Chen, S.; Tuttle, D.L.; Oshier, J.T.; Knot, H.J.; Streit, W.J.; Goodenow, M.M.; Harrison, J.K. Transforming growth factor-beta1 increases cxcr4 expression, stromal-derived factor-1alpha-stimulated signalling and human immunodeficiency virus-1 entry in human monocyte-derived macrophages. Immunology 2005, 114, 565–574. [Google Scholar] [CrossRef]
  25. Chinnapaiyan, S.; Dutta, R.K.; Nair, M.; Chand, H.S.; Rahman, I.; Unwalla, H.J. Tgf-beta1 increases viral burden and promotes HIV-1 latency in primary differentiated human bronchial epithelial cells. Sci. Rep. 2019, 9, 12552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bacsi, A.; Aranyosi, J.; Beck, Z.; Ebbesen, P.; Andirko, I.; Szabo, J.; Lampe, L.; Kiss, J.; Gergely, L.; Toth, F.D. Placental macrophage contact potentiates the complete replicative cycle of human cytomegalovirus in syncytiotrophoblast cells: Role of interleukin-8 and transforming growth factor-beta1. J. Interf. Cytokine Res. 1999, 19, 1153–1160. [Google Scholar] [CrossRef] [PubMed]
  27. Flisiak, R.; Prokopowicz, D.; Jaroszewicz, J.; Flisiak, I. Plasma transforming growth factor-beta(1) in acute viral hepatitis. Med. Sci. Monit. 2005, 11, CR304–CR308. [Google Scholar]
  28. Chou, Y.C.; Chen, M.L.; Hu, C.P.; Chen, Y.L.; Chong, C.L.; Tsai, Y.L.; Liu, T.L.; Jeng, K.S.; Chang, C. Transforming growth factor-beta1 suppresses hepatitis b virus replication primarily through transcriptional inhibition of pregenomic rna. Hepatology 2007, 46, 672–681. [Google Scholar] [CrossRef]
  29. Li, W.; Yu, X.; Chen, X.; Wang, Z.; Yin, M.; Zhao, Z.; Zhu, C. Hbv induces liver fibrosis via the tgf-beta1/mir-21-5p pathway. Exp. Ther. Med. 2021, 21, 169. [Google Scholar] [CrossRef]
  30. Wang, Y. The inhibition of microrna-15a suppresses hepatitis b virus-associated liver cancer cell growth through the smad/tgf-beta pathway. Oncol. Rep. 2017, 37, 3520–3526. [Google Scholar] [CrossRef] [Green Version]
  31. Huntington, K.E.; Carlsen, L.; So, E.Y.; Piesche, M.; Liang, O.; El-Deiry, W.S. Integrin/tgf-beta1 inhibitor glpg-0187 blocks SARS-CoV-2 delta and omicron pseudovirus infection of airway epithelial cells in vitro, which could attenuate disease severity. Pharmaceuticals 2022, 15, 618. [Google Scholar] [CrossRef]
  32. Wang, E.Y.; Chen, H.; Sun, B.Q.; Wang, H.; Qu, H.Q.; Liu, Y.; Sun, X.Z.; Qu, J.; Fang, Z.F.; Tian, L.; et al. Serum levels of the iga isotype switch factor tgf-beta1 are elevated in patients with COVID-19. FEBS Lett. 2021, 595, 1819–1824. [Google Scholar] [CrossRef]
  33. Montalvo Villalba, M.C.; Valdes Ramirez, O.; Mune Jimenez, M.; Arencibia Garcia, A.; Martinez Alfonso, J.; Gonzalez Baez, G.; Roque Arrieta, R.; Rosell Simon, D.; Alvarez Gainza, D.; Sierra Vazquez, B.; et al. Interferon gamma, tgf-beta1 and rantes expression in upper airway samples from SARS-CoV-2 infected patients. Clin. Immunol. 2020, 220, 108576. [Google Scholar] [CrossRef]
  34. Trinh, Q.D.; Pham, N.T.K.; Takada, K.; Takano, C.; Komine-Aizawa, S.; Hayakawa, S. Tgf-beta1 promotes zika virus infection in immortalized human first-trimester trophoblasts via the smad pathway. Cells 2022, 11, 3026. [Google Scholar] [CrossRef]
  35. Kovacs, A.A.Z. Zika, the newest torch infectious disease in the americas. Clin. Infect. Dis. 2020, 70, 2673–2674. [Google Scholar] [CrossRef]
  36. Morand, A.; Zandotti, C.; Charrel, R.; Minodier, P.; Fabre, A.; Chabrol, B.; De Lamballerie, X. From torch to torchz: Zika virus infection highlights infectious fetopathies. Arch. Pediatr. 2017, 24, 911–913. [Google Scholar] [CrossRef]
  37. Warnecke, J.M.; Pollmann, M.; Borchardt-Loholter, V.; Moreira-Soto, A.; Kaya, S.; Sener, A.G.; Gomez-Guzman, E.; Figueroa-Hernandez, L.; Li, W.; Li, F.; et al. Seroprevalences of antibodies against torch infectious pathogens in women of childbearing age residing in brazil, mexico, germany, poland, turkey and china. Epidemiol. Infect. 2020, 148, e271. [Google Scholar] [CrossRef]
  38. Yu, W.; Hu, X.; Cao, B. Viral infections during pregnancy: The big challenge threatening maternal and fetal health. Matern.-Fetal Med. 2022, 4, 72–86. [Google Scholar] [CrossRef]
  39. Semmes, E.C.; Coyne, C.B. Innate immune defenses at the maternal-fetal interface. Curr. Opin. Immunol. 2022, 74, 60–67. [Google Scholar] [CrossRef]
  40. Megli, C.J.; Coyne, C.B. Infections at the maternal–fetal interface: An overview of pathogenesis and defence. Nat. Rev. Microbiol. 2022, 20, 67–82. [Google Scholar] [CrossRef]
  41. Kumar, M.; Saadaoui, M.; Al Khodor, S. Infections and pregnancy: Effects on maternal and child health. Front. Cell. Infect. Microbiol. 2022, 12, 873253. [Google Scholar] [CrossRef]
  42. Red-Horse, K.; Zhou, Y.; Genbacev, O.; Prakobphol, A.; Foulk, R.; McMaster, M.; Fisher, S.J. Trophoblast differentiation during embryo implantation and formation of the maternal-fetal interface. J. Clin. Investig. 2004, 114, 744–754. [Google Scholar] [CrossRef] [PubMed]
  43. Pijnenborg, R.; Vercruysse, L.; Hanssens, M. The uterine spiral arteries in human pregnancy: Facts and controversies. Placenta 2006, 27, 939–958. [Google Scholar] [CrossRef] [PubMed]
  44. Renaud, S.J.; Jeyarajah, M.J. How trophoblasts fuse: An in-depth look into placental syncytiotrophoblast formation. Cell. Mol. Life Sci. 2022, 79, 433. [Google Scholar] [CrossRef] [PubMed]
  45. Maudhoo, A.; Khalil, A. Viral pulmonary infection in pregnancy—Including COVID-19, sars, influenza a, and varicella. Best Pract. Res. Clin. Obstet. Gynaecol. 2022, 85, 17–25. [Google Scholar] [CrossRef]
  46. Hayakawa, S.; Komine-Aizawa, S.; Mor, G.G. COVID-19 pandemic and pregnancy. J. Obstet. Gynaecol. Res. 2020, 46, 1958–1966. [Google Scholar] [CrossRef]
  47. Alouini, S.; Guinard, J.; Belin, O.; Mesnard, L.; Werner, E.; Prazuck, T.; Pichon, C. Maternal-fetal implications of SARS-CoV-2 infection during pregnancy, viral, serological analyses of placenta and cord blood. Int. J. Environ. Res. Public Health 2022, 19, 2105. [Google Scholar] [CrossRef]
  48. Zimmerman, L.A.; Knapp, J.K.; Antoni, S.; Grant, G.B.; Reef, S.E. Progress toward rubella and congenital rubella syndrome control and elimination—Worldwide, 2012–2020. MMWR Morb. Mortal. Wkly. Rep. 2022, 71, 196–201. [Google Scholar] [CrossRef]
  49. Namiki, T.; Takano, C.; Aoki, R.; Trinh, Q.D.; Morioka, I.; Hayakawa, S. Parenchymal calcification is associated with the neurological prognosis in patients with congenital rubella syndrome. Congenit. Anom. 2022, 62, 38–41. [Google Scholar] [CrossRef]
  50. Ekuma, U.O.; Ogbu, O.; Oli, A.N.; Okolo, M.O.; Edeh, P.A.; Al-Dahmoshi, H.O.M.; Akrami, S.; Saki, M. The burden of likely rubella infection among healthy pregnant women in abakaliki, ebonyi state, nigeria. Interdiscip. Perspect. Infect. Dis. 2022, 2022, 5743106. [Google Scholar] [CrossRef]
  51. Shukla, S.; Maraqa, N.F. Congenital rubella. In Statpearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  52. Gudeloglu, E.; Akillioglu, M.; Demirdag, T.B.; Unal, N.A.; Tapisiz, A.A. Congenital rubella syndrome: A short report and literature review. Trop. Dr. 2023, 53, 171–175. [Google Scholar] [CrossRef]
  53. Singh, G.; Gaidhane, A. A review of sensorineural hearing loss in congenital cytomegalovirus infection. Cureus 2022, 14, e30703. [Google Scholar] [CrossRef]
  54. Xia, W.; Yan, H.; Zhang, Y.; Wang, C.; Gao, W.; Lv, C.; Wang, W.; Liu, Z. Congenital human cytomegalovirus infection inducing sensorineural hearing loss. Front. Microbiol. 2021, 12, 649690. [Google Scholar] [CrossRef]
  55. Davis, N.L.; King, C.C.; Kourtis, A.P. Cytomegalovirus infection in pregnancy. Birth Defects Res. 2017, 109, 336–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Naing, Z.W.; Scott, G.M.; Shand, A.; Hamilton, S.T.; van Zuylen, W.J.; Basha, J.; Hall, B.; Craig, M.E.; Rawlinson, W.D. Congenital cytomegalovirus infection in pregnancy: A review of prevalence, clinical features, diagnosis and prevention. Aust. N. Z. J. Obstet. Gynaecol. 2016, 56, 9–18. [Google Scholar] [CrossRef]
  57. Jose Antonio, M.M.; Monica Grisel, R.M.; Alberto, C.S.; Carla Ileana, A.A.; Luis Antonio, U.N.; Maria de Los Angeles, B.S.; Norma Angelica, M.J.; Mara Soraya, R.E.; Victor, R.P.; Jesus Enrique, G.M. Maternal and neonatal risk factors associated with increased mother-to-child transmission of HIV-1 in mexico: Results of a case–control study. Int. J. STD AIDS 2022, 33, 1111–1118. [Google Scholar] [CrossRef]
  58. WHO. HIV/Aids: Mother-to-Child Transmission; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  59. Vidya Vijayan, K.K.; Karthigeyan, K.P.; Tripathi, S.P.; Hanna, L.E. Pathophysiology of cd4+ t-cell depletion in HIV-1 and HIV-2 infections. Front. Immunol. 2017, 8, 580. [Google Scholar] [CrossRef] [Green Version]
  60. Grignolo, S.; Agnello, R.; Gerbaldo, D.; Gotta, C.; Alicino, C.; Del Puente, F.; Taramasso, L.; Bruzzone, B.; Gustavino, C.; Trasino, S.; et al. Pregnancy and neonatal outcomes among a cohort of HIV-infected women in a large italian teaching hospital: A 30-year retrospective study. Epidemiol. Infect. 2017, 145, 1658–1669. [Google Scholar] [CrossRef] [Green Version]
  61. Spinillo, A.; Iasci, A.; Dal Maso, J.; Di Lenardo, L.; Grella, P.; Guaschino, S. The effect of fetal infection with human immunodeficiency virus type 1 on birthweight and length of gestation. Sigo study group of HIV infection in pregnancy. Eur. J. Obstet. Gynecol. Reprod. Biol. 1994, 57, 13–17. [Google Scholar] [CrossRef]
  62. Kumar, M.; Abbas, Z.; Azami, M.; Belopolskaya, M.; Dokmeci, A.K.; Ghazinyan, H.; Jia, J.; Jindal, A.; Lee, H.C.; Lei, W.; et al. Asian pacific association for the study of liver (apasl) guidelines: Hepatitis b virus in pregnancy. Hepatol. Int. 2022, 16, 211–253. [Google Scholar] [CrossRef]
  63. Xiong, Y.; Liu, C.; Huang, S.; Wang, J.; Qi, Y.; Yao, G.; Sun, W.; Qian, Y.; Ye, L.; Liu, H.; et al. Impact of maternal infection with hepatitis b virus on pregnancy complications and neonatal outcomes for women undergoing assisted reproductive technology treatment: A population-based study. J. Viral Hepat. 2021, 28, 613–620. [Google Scholar] [CrossRef]
  64. WHO. Global Progress Report on HIV, Viral Hepatitis and Sexually Transmitted Infections; World Health Organization: Geneva, Switzerland, 2021. [Google Scholar]
  65. Bajema, K.L.; Stankiewicz Karita, H.C.; Tenforde, M.W.; Hawes, S.E.; Heffron, R. Maternal hepatitis b infection and pregnancy outcomes in the united states: A population-based cohort study. Open Forum Infect. Dis. 2018, 5, ofy134. [Google Scholar] [CrossRef] [Green Version]
  66. Deftereou, T.E.; Trypidi, A.; Alexiadi, C.A.; Theotokis, P.; Manthou, M.E.; Meditskou, S.; Simopoulou, M.; Lambropoulou, M. Congenital herpes simplex virus: A histopathological view of the placenta. Cureus 2022, 14, e29101. [Google Scholar] [CrossRef]
  67. Krenn, V.; Bosone, C.; Burkard, T.R.; Spanier, J.; Kalinke, U.; Calistri, A.; Salata, C.; Rilo Christoff, R.; Pestana Garcez, P.; Mirazimi, A.; et al. Organoid modeling of zika and herpes simplex virus 1 infections reveals virus-specific responses leading to microcephaly. Cell Stem Cell 2021, 28, 1362–1379.e7. [Google Scholar] [CrossRef] [PubMed]
  68. Hammad, W.A.B.; Konje, J.C. Herpes simplex virus infection in pregnancy—An update. Eur. J. Obstet. Gynecol. Reprod. Biol. 2021, 259, 38–45. [Google Scholar] [CrossRef] [PubMed]
  69. Heggarty, E.; Sibiude, J.; Mandelbrot, L.; Vauloup-Fellous, C.; Picone, O. Genital herpes and pregnancy: Evaluating practices and knowledge of french health care providers. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 249, 84–91. [Google Scholar] [CrossRef]
  70. Vazquez-Pagan, A.; Schultz-Cherry, S. Serological responses to influenza vaccination during pregnancy. Microorganisms 2021, 9, 2305. [Google Scholar] [CrossRef]
  71. Fuentes-Zacarias, P.; Murrieta-Coxca, J.M.; Gutierrez-Samudio, R.N.; Schmidt, A.; Schmidt, A.; Markert, U.R.; Morales-Prieto, D.M. Pregnancy and pandemics: Interaction of viral surface proteins and placenta cells. Biochim. Biophys. Acta Mol. (BBA)-Basis Dis. 2021, 1867, 166218. [Google Scholar] [CrossRef]
  72. Littauer, E.Q.; Esser, E.S.; Antao, O.Q.; Vassilieva, E.V.; Compans, R.W.; Skountzou, I. H1N1 influenza virus infection results in adverse pregnancy outcomes by disrupting tissue-specific hormonal regulation. PLoS Pathog. 2017, 13, e1006757. [Google Scholar] [CrossRef] [Green Version]
  73. Hewagama, S.; Walker, S.P.; Stuart, R.L.; Gordon, C.; Johnson, P.D.; Friedman, N.D.; O’Reilly, M.; Cheng, A.C.; Giles, M.L. 2009 H1N1 influenza a and pregnancy outcomes in victoria, australia. Clin. Infect. Dis. 2010, 50, 686–690. [Google Scholar] [CrossRef]
  74. Teixeira, F.M.E.; Pietrobon, A.J.; Oliveira, L.M.; Oliveira, L.; Sato, M.N. Maternal-fetal interplay in zika virus infection and adverse perinatal outcomes. Front. Immunol. 2020, 11, 175. [Google Scholar] [CrossRef]
  75. Espino, A.; Gouilly, J.; Chen, Q.; Colin, P.; Guerby, P.; Izopet, J.; Amara, A.; Tabiasco, J.; Al-Daccak, R.; El Costa, H.; et al. The mechanisms underlying the immune control of zika virus infection at the maternal-fetal interface. Front. Immunol. 2022, 13, 1000861. [Google Scholar] [CrossRef] [PubMed]
  76. Ades, A.E.; Soriano-Arandes, A.; Alarcon, A.; Bonfante, F.; Thorne, C.; Peckham, C.S.; Giaquinto, C. Vertical transmission of zika virus and its outcomes: A bayesian synthesis of prospective studies. Lancet Infect. Dis. 2021, 21, 537–545. [Google Scholar] [CrossRef] [PubMed]
  77. de Noronha, L.; Zanluca, C.; Burger, M.; Suzukawa, A.A.; Azevedo, M.; Rebutini, P.Z.; Novadzki, I.M.; Tanabe, L.S.; Presibella, M.M.; Duarte Dos Santos, C.N. Zika virus infection at different pregnancy stages: Anatomopathological findings, target cells and viral persistence in placental tissues. Front. Microbiol. 2018, 9, 2266. [Google Scholar] [CrossRef]
  78. Mourosi, J.T.; Awe, A.; Jain, S.; Batra, H. Nucleic acid vaccine platform for dengue and zika flaviviruses. Vaccines 2022, 10, 834. [Google Scholar] [CrossRef]
  79. Masmejan, S.; Pomar, L.; Favre, G.; Panchaud, A.; Giannoni, E.; Greub, G.; Baud, D. Vertical transmission and materno-fetal outcomes in 13 patients with coronavirus disease 2019. Clin. Microbiol. Infect. 2020, 26, 1585–1587. [Google Scholar] [CrossRef]
  80. Shook, L.L.; Sullivan, E.L.; Lo, J.O.; Perlis, R.H.; Edlow, A.G. COVID-19 in pregnancy: Implications for fetal brain development. Trends Mol. Med. 2022, 28, 319–330. [Google Scholar] [CrossRef]
  81. Shook, L.L.; Fourman, L.T.; Edlow, A.G. Immune responses to SARS-CoV-2 in pregnancy: Implications for the health of the next generation. J. Immunol. 2022, 209, 1465–1473. [Google Scholar] [CrossRef]
  82. Parcial, A.L.N.; Salomao, N.G.; Portari, E.A.; Arruda, L.V.; de Carvalho, J.J.; de Matos Guedes, H.L.; Conde, T.C.; Moreira, M.E.; Batista, M.M.; Paes, M.V.; et al. SARS-CoV-2 is persistent in placenta and causes macroscopic, histopathological, and ultrastructural changes. Viruses 2022, 14, 1885. [Google Scholar] [CrossRef]
  83. Li, P.; Xie, M.; Zhang, W. Clinical characteristics and intrauterine vertical transmission potential of coronavirus disease 2019 infection in 9 pregnant women: A retrospective review of medical records. Am. J. Obstet. Gynecol. 2020, 223, 955–956. [Google Scholar] [CrossRef]
  84. Patane, L.; Morotti, D.; Giunta, M.R.; Sigismondi, C.; Piccoli, M.G.; Frigerio, L.; Mangili, G.; Arosio, M.; Cornolti, G. Vertical transmission of coronavirus disease 2019: Severe acute respiratory syndrome coronavirus 2 rna on the fetal side of the placenta in pregnancies with coronavirus disease 2019-positive mothers and neonates at birth. Am. J. Obstet. Gynecol. MFM 2020, 2, 100145. [Google Scholar] [CrossRef]
  85. AlQahtani, M.A.; AlDajani, S.M. A systemic review of vertical transmission possibility in pregnant women with coronavirus disease 2019-positive status. J. Fam. Med. Prim. Care 2020, 9, 4521–4525. [Google Scholar] [CrossRef] [PubMed]
  86. Angelova, M.; Kovachev, E.; Todorov, N. Cytomegalovirus infection during pregnancy and its impact on the intrauterine fetal development—Case report. Open Access Maced. J. Med. Sci. 2016, 4, 449–452. [Google Scholar] [CrossRef] [Green Version]
  87. Avgil, M.; Ornoy, A. Herpes simplex virus and epstein-barr virus infections in pregnancy: Consequences of neonatal or intrauterine infection. Reprod. Toxicol. 2006, 21, 436–445. [Google Scholar] [CrossRef]
  88. Goncalves, L.F.; Chaiworapongsa, T.; Romero, R. Intrauterine infection and prematurity. Ment. Retard. Dev. Disabil. Res. Rev. 2002, 8, 3–13. [Google Scholar] [CrossRef]
  89. Guan, M.; Johannesen, E.; Tang, C.Y.; Hsu, A.L.; Barnes, C.L.; Burnam, M.; McElroy, J.A.; Wan, X.F. Intrauterine fetal demise in the third trimester of pregnancy associated with mild infection with the SARS-CoV-2 delta variant without protection from vaccination. J. Infect. Dis. 2022, 225, 748–753. [Google Scholar] [CrossRef]
  90. Fuwa, K.; Hayakawa, S. Mechanisms and possible controls of the in utero zika virus infection: Where is the holy grail? Am. J. Reprod. Immunol. 2017, 77, e12605. [Google Scholar] [CrossRef] [Green Version]
  91. Centeno-Tablante, E.; Medina-Rivera, M.; Finkelstein, J.L.; Herman, H.S.; Rayco-Solon, P.; Garcia-Casal, M.N.; Rogers, L.; Ghezzi-Kopel, K.; Zambrano Leal, M.P.; Andrade Velasquez, J.K.; et al. Update on the transmission of zika virus through breast milk and breastfeeding: A systematic review of the evidence. Viruses 2021, 13, 123. [Google Scholar] [CrossRef]
  92. Trinh, Q.D.; Takada, K.; Pham, N.T.K.; Takano, C.; Namiki, T.; Ikuta, R.; Hayashida, S.; Okitsu, S.; Ushijima, H.; Komine-Aizawa, S.; et al. Enhancement of rubella virus infection in immortalized human first-trimester trophoblasts under low-glucose stress conditions. Front. Microbiol. 2022, 13, 904189. [Google Scholar] [CrossRef]
  93. Pereira, L. Congenital viral infection: Traversing the uterine-placental interface. Annu. Rev. Virol. 2018, 5, 273–299. [Google Scholar] [CrossRef] [Green Version]
  94. Kirschen, G.W.; Burd, I. Modeling of vertical transmission and pathogenesis of cytomegalovirus in pregnancy: Opportunities and challenges. Front. Virol. 2023, 3, 1106634. [Google Scholar] [CrossRef]
  95. Weisblum, Y.; Panet, A.; Haimov-Kochman, R.; Wolf, D.G. Models of vertical cytomegalovirus (CMV) transmission and pathogenesis. Semin. Immunopathol. 2014, 36, 615–625. [Google Scholar] [CrossRef] [PubMed]
  96. Sibiude, J.; Le Chenadec, J.; Mandelbrot, L.; Hoctin, A.; Dollfus, C.; Faye, A.; Bui, E.; Pannier, E.; Ghosn, J.; Garrait, V.; et al. Update of perinatal human immunodeficiency virus type 1 transmission in france: Zero transmission for 5482 mothers on continuous antiretroviral therapy from conception and with undetectable viral load at delivery. Clin. Infect. Dis. 2023, 76, e590–e598. [Google Scholar] [CrossRef] [PubMed]
  97. Ter Schiphorst, E.; Hansen, K.C.; Holm, M.; Honge, B.L. Mother-to-child HIV-2 transmission: Comparison with HIV-1 and evaluation of factors influencing the rate of transmission. A systematic review. Trans. R. Soc. Trop. Med. Hyg. 2022, 116, 399–408. [Google Scholar] [CrossRef]
  98. Al-Husaini, A.M. Role of placenta in the vertical transmission of human immunodeficiency virus. J. Perinatol. 2009, 29, 331–336. [Google Scholar] [CrossRef] [Green Version]
  99. WHO. Operationalizing elimination of mother-to-child transmission of hepatitis b virus in the western pacific region. In World Health Organization Regional Office for the Western Pacific, Manila, Licence: CC BY-NC-SA 3.0 IGO; WHO: Geneva, Switzerland, 2021. [Google Scholar]
  100. Lv, N.; Chu, X.D.; Sun, Y.H.; Zhao, S.Y.; Li, P.L.; Chen, X. Analysis on the outcomes of hepatitis b virus perinatal vertical transmission: Nested case-control study. Eur. J. Gastroenterol. Hepatol. 2014, 26, 1286–1291. [Google Scholar] [CrossRef]
  101. Xu, D.Z.; Yan, Y.P.; Choi, B.C.; Xu, J.Q.; Men, K.; Zhang, J.X.; Liu, Z.H.; Wang, F.S. Risk factors and mechanism of transplacental transmission of hepatitis b virus: A case-control study. J. Med. Virol. 2002, 67, 20–26. [Google Scholar] [CrossRef]
  102. Belanger, B.G.; Lui, F. Embryology, teratology torch. In Statpearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  103. Felker, A.M.; Nguyen, P.; Kaushic, C. Primary HSV-2 infection in early pregnancy results in transplacental viral transmission and dose-dependent adverse pregnancy outcomes in a novel mouse model. Viruses 2021, 13, 1929. [Google Scholar] [CrossRef]
  104. Westhoff, G.L.; Little, S.E.; Caughey, A.B. Herpes simplex virus and pregnancy: A review of the management of antenatal and peripartum herpes infections. Obstet. Gynecol. Surv. 2011, 66, 629–638. [Google Scholar] [CrossRef]
  105. Kriebs, J.M. Understanding herpes simplex virus: Transmission, diagnosis, and considerations in pregnancy management. J. Midwifery Women’s Health 2008, 53, 202–208. [Google Scholar] [CrossRef]
  106. Nunes, M.C.; Madhi, S.A. Prevention of influenza-related illness in young infants by maternal vaccination during pregnancy. F1000Research 2018, 7, 122. [Google Scholar] [CrossRef]
  107. Irving, W.L.; James, D.K.; Stephenson, T.; Laing, P.; Jameson, C.; Oxford, J.S.; Chakraverty, P.; Brown, D.W.; Boon, A.C.; Zambon, M.C. Influenza virus infection in the second and third trimesters of pregnancy: A clinical and seroepidemiological study. BJOG 2000, 107, 1282–1289. [Google Scholar] [CrossRef] [PubMed]
  108. Jabrane-Ferrat, N.; Veas, F. Zika virus targets multiple tissues and cell types during the first trimester of pregnancy. Methods Mol. Biol. 2020, 2142, 235–249. [Google Scholar] [PubMed]
  109. Schwartz, D.A.; Morotti, D.; Beigi, B.; Moshfegh, F.; Zafaranloo, N.; Patane, L. Confirming vertical fetal infection with coronavirus disease 2019: Neonatal and pathology criteria for early onset and transplacental transmission of severe acute respiratory syndrome coronavirus 2 from infected pregnant mothers. Arch. Pathol. Lab. Med. 2020, 144, 1451–1456. [Google Scholar] [CrossRef]
  110. Gupta, A.; Malhotra, Y.; Patil, U.; Muradas, A.R.; Lee, W.T.; Krammer, F.; Amanat, F.; Clare, C.A.; Vinod, S.; Ghaly, E. In utero vertical transmission of coronavirus disease 2019 in a severely ill 29-week preterm infant. Am. J. Perinatol. Rep. 2020, 10, e270–e274. [Google Scholar] [CrossRef]
  111. Mankertz, A.; Chen, M.-H.; Goldberg, T.L.; Hübschen, J.M.; Pfaff, F.; Ulrich, R.G.; ICTV Report Consortium. ICTV virus taxonomy profile: Matonaviridae 2022. J. Gen. Virol. 2022, 103, 001817. [Google Scholar] [CrossRef]
  112. Fonseca, I.C.; Wong, J.; Mireskandari, K.; Chitayat, D. Newborn with bilateral congenital cataracts: Never forget congenital rubella syndrome. Paediatr. Child Health 2020, 25, 72–76. [Google Scholar] [CrossRef]
  113. Chauhan, N.; Sen, M.S.; Jhanda, S.; Grover, S. Psychiatric manifestations of congenital rubella syndrome: A case report and review of literature. J. Pediatr. Neurosci. 2016, 11, 137–139. [Google Scholar]
  114. Centers for Disease Control and Prevention. Nationwide rubella epidemic—Japan, 2013. MMWR Morb. Mortal. Wkly. Rep. 2013, 62, 457–462. [Google Scholar]
  115. Kanai, M.; Kamiya, H.; Okuno, H.; Sunagawa, T.; Tanaka-Taya, K.; Matsui, T.; Oishi, K.; Kitajima, H.; Takeda, M.; Mori, Y. Epidemiology of congenital rubella syndrome related to the 2012–2013 rubella epidemic in Japan. J. Pediatric. Infect. Dis. Soc. 2022, 11, 400–403. [Google Scholar] [CrossRef]
  116. Ujiie, M. Rubella resurgence in Japan 2018–2019. J. Travel Med. 2019, 26, taz047. [Google Scholar] [CrossRef]
  117. Tilahun, G.T.; Tolasa, T.M.; Wole, G.A. Modeling the dynamics of rubella disease with vertical transmission. Heliyon 2022, 8, e11797. [Google Scholar] [CrossRef] [PubMed]
  118. Bate, S.L.; Dollard, S.C.; Cannon, M.J. Cytomegalovirus seroprevalence in the united states: The national health and nutrition examination surveys, 1988–2004. Clin. Infect. Dis. 2010, 50, 1439–1447. [Google Scholar] [CrossRef] [Green Version]
  119. Cannon, M.J.; Schmid, D.S.; Hyde, T.B. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev. Med. Virol. 2010, 20, 202–213. [Google Scholar] [CrossRef]
  120. Syggelou, A.; Iacovidou, N.; Kloudas, S.; Christoni, Z.; Papaevangelou, V. Congenital cytomegalovirus infection. Ann. N. Y. Acad. Sci. 2010, 1205, 144–147. [Google Scholar] [CrossRef]
  121. Fowler, K.B.; Stagno, S.; Pass, R.F. Maternal age and congenital cytomegalovirus infection: Screening of two diverse newborn populations, 1980-1990. J. Infect. Dis. 1993, 168, 552–556. [Google Scholar] [CrossRef]
  122. Reynolds, D.W.; Stagno, S.; Hosty, T.S.; Tiller, M.; Alford, C.A., Jr. Maternal cytomegalovirus excretion and perinatal infection. N. Engl. J. Med. 1973, 289, 1–5. [Google Scholar] [CrossRef]
  123. Fisher, S.; Genbacev, O.; Maidji, E.; Pereira, L. Human cytomegalovirus infection of placental cytotrophoblasts in vitro and in utero: Implications for transmission and pathogenesis. J. Virol. 2000, 74, 6808–6820. [Google Scholar] [CrossRef] [Green Version]
  124. Pass, R.F. Epidemiology and transmission of cytomegalovirus. J. Infect. Dis. 1985, 152, 243–248. [Google Scholar] [CrossRef]
  125. Stagno, S.; Pass, R.F.; Cloud, G.; Britt, W.J.; Henderson, R.E.; Walton, P.D.; Veren, D.A.; Page, F.; Alford, C.A. Primary cytomegalovirus infection in pregnancy. Incidence, transmission to fetus, and clinical outcome. JAMA 1986, 256, 1904–1908. [Google Scholar] [CrossRef]
  126. Hemmings, D.G.; Kilani, R.; Nykiforuk, C.; Preiksaitis, J.; Guilbert, L.J. Permissive cytomegalovirus infection of primary villous term and first trimester trophoblasts. J. Virol. 1998, 72, 4970–4979. [Google Scholar] [CrossRef] [Green Version]
  127. Toth, F.D.; Mosborg-Petersen, P.; Kiss, J.; Aboagye-Mathiesen, G.; Hager, H.; Juhl, C.B.; Gergely, L.; Zdravkovic, M.; Aranyosi, J.; Lampe, L. Interactions between human immunodeficiency virus type 1 and human cytomegalovirus in human term syncytiotrophoblast cells coinfected with both viruses. J. Virol. 1995, 69, 2223–2232. [Google Scholar] [CrossRef] [Green Version]
  128. Vijayan, V.; Naeem, F.; Veesenmeyer, A.F. Management of infants born to mothers with HIV infection. Am. Fam. Physician 2021, 104, 58–62. [Google Scholar] [PubMed]
  129. Chigwedere, P.; Seage, G.R.; Lee, T.H.; Essex, M. Efficacy of antiretroviral drugs in reducing mother-to-child transmission of HIV in Africa: A meta-analysis of published clinical trials. AIDS Res. Hum. Retrovir. 2008, 24, 827–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Zijenah, L.S.; Bandason, T.; Bara, W.; Chipiti, M.M.; Katzenstein, D.A. Impact of option b(+) combination antiretroviral therapy on mother-to-child transmission of HIV-1, maternal and infant virologic responses to combination antiretroviral therapy, and maternal and infant mortality rates: A 24-month prospective follow-up study at a primary health care clinic, in harare, zimbabwe. AIDS Patient Care STDS 2022, 36, 145–152. [Google Scholar] [PubMed]
  131. Soriano-Arandes, A.; Noguera-Julian, A.; Lopez-Lacort, M.; Soler-Palacin, P.; Mur, A.; Mendez, M.; Mayol, L.; Vallmanya, T.; Almeda, J.; Carnicer-Pont, D.; et al. Pregnancy as an opportunity to diagnose human-immunodeficiency virus immigrant women in catalonia. Enferm. Infecc. Microbiol. Clin. 2018, 36, 9–15. [Google Scholar] [CrossRef]
  132. Cooper, E.R.; Charurat, M.; Mofenson, L.; Hanson, I.C.; Pitt, J.; Diaz, C.; Hayani, K.; Handelsman, E.; Smeriglio, V.; Hoff, R.; et al. Combination antiretroviral strategies for the treatment of pregnant HIV-1-infected women and prevention of perinatal HIV-1 transmission. J. Acquir. Immune Defic. Syndr. 2002, 29, 484–494. [Google Scholar] [CrossRef]
  133. Mano, H.; Chermann, J.C. Replication of human immunodeficiency virus type 1 in primary cultured placental cells. Res. Virol. 1991, 142, 95–104. [Google Scholar] [CrossRef]
  134. McGann, K.A.; Collman, R.; Kolson, D.L.; Gonzalez-Scarano, F.; Coukos, G.; Coutifaris, C.; Strauss, J.F.; Nathanson, N. Human immunodeficiency virus type 1 causes productive infection of macrophages in primary placental cell cultures. J. Infect. Dis. 1994, 169, 746–753. [Google Scholar] [CrossRef]
  135. Kilani, R.T.; Chang, L.J.; Garcia-Lloret, M.I.; Hemmings, D.; Winkler-Lowen, B.; Guilbert, L.J. Placental trophoblasts resist infection by multiple human immunodeficiency virus (HIV) type 1 variants even with cytomegalovirus coinfection but support HIV replication after provirus transfection. J. Virol. 1997, 71, 6359–6372. [Google Scholar] [CrossRef] [Green Version]
  136. Vidricaire, G.; Tardif, M.R.; Tremblay, M.J. The low viral production in trophoblastic cells is due to a high endocytic internalization of the human immunodeficiency virus type 1 and can be overcome by the pro-inflammatory cytokines tumor necrosis factor-alpha and interleukin-1. J. Biol. Chem. 2003, 278, 15832–15841. [Google Scholar] [CrossRef] [Green Version]
  137. Alter, M.J. Epidemiology of hepatitis b in europe and worldwide. J. Hepatol. 2003, 39 (Suppl. S1), S64–S69. [Google Scholar] [CrossRef]
  138. Eke, A.C.; Eleje, G.U.; Eke, U.A.; Xia, Y.; Liu, J. Hepatitis b immunoglobulin during pregnancy for prevention of mother-to-child transmission of hepatitis b virus. Cochrane Database Syst. Rev. 2017, 2, CD008545. [Google Scholar] [CrossRef]
  139. Goudeau, A.; Yvonnet, B.; Lesage, G.; Barin, F.; Denis, F.; Coursaget, P.; Chiron, J.P.; Diop Mar, I. Lack of anti-hbc igm in neonates with hbsag carrier mothers argues against transplacental transmission of hepatitis b virus infection. Lancet 1983, 2, 1103–1104. [Google Scholar] [CrossRef]
  140. Schweitzer, I.L. Vertical transmission of the hepatitis b surface antigen. Am. J. Med. Sci. 1975, 270, 287–291. [Google Scholar] [CrossRef]
  141. Zhang, S.L.; Yue, Y.F.; Bai, G.Q.; Shi, L.; Jiang, H. Mechanism of intrauterine infection of hepatitis b virus. World J. Gastroenterol. 2004, 10, 437–438. [Google Scholar] [CrossRef]
  142. Bai, G.; Wang, Y.; Zhang, L.; Tang, Y.; Fu, F. The study on the role of hepatitis b virus x protein and apoptosis in HBV intrauterine infection. Arch. Gynecol. Obstet. 2012, 285, 943–949. [Google Scholar] [CrossRef]
  143. Wang, X.P.; Li, F.J.; Xu, D.Z.; Yan, Y.P.; Men, K.; Zhang, J.X. Uptake of hepatitis b virus into choriocarcinoma cells in the presence of proinflammatory cytokine tumor necrosis factor-alpha. Am. J. Obstet. Gynecol. 2004, 191, 1971–1978. [Google Scholar] [CrossRef]
  144. Singhal, P.; Naswa, S.; Marfatia, Y.S. Pregnancy and sexually transmitted viral infections. Indian J. Sex Transm. Dis. AIDS 2009, 30, 71–78. [Google Scholar]
  145. Brown, Z.A.; Selke, S.; Zeh, J.; Kopelman, J.; Maslow, A.; Ashley, R.L.; Watts, D.H.; Berry, S.; Herd, M.; Corey, L. The acquisition of herpes simplex virus during pregnancy. N. Engl. J. Med. 1997, 337, 509–515. [Google Scholar] [CrossRef]
  146. Baker, D.A. Consequences of herpes simplex virus in pregnancy and their prevention. Curr. Opin. Infect. Dis. 2007, 20, 73–76. [Google Scholar] [CrossRef]
  147. Mertz, G.J.; Rosenthal, S.L.; Stanberry, L.R. Is herpes simplex virus type 1 (HSV-1) now more common than HSV-2 in first episodes of genital herpes? Sex. Transm. Dis. 2003, 30, 801–802. [Google Scholar] [CrossRef] [PubMed]
  148. Brown, Z.A.; Wald, A.; Morrow, R.A.; Selke, S.; Zeh, J.; Corey, L. Effect of serologic status and cesarean delivery on transmission rates of herpes simplex virus from mother to infant. JAMA 2003, 289, 203–209. [Google Scholar] [CrossRef] [PubMed]
  149. Pinninti, S.G.; Kimberlin, D.W. Preventing herpes simplex virus in the newborn. Clin. Perinatol. 2014, 41, 945–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Florman, A.L.; Gershon, A.A.; Blackett, P.R.; Nahmias, A.J. Intrauterine infection with herpes simplex virus: Resultant congenital malformations. JAMA 1973, 225, 129–132. [Google Scholar] [CrossRef]
  151. Hutto, C.; Arvin, A.; Jacobs, R.; Steele, R.; Stagno, S.; Lyrene, R.; Willett, L.; Powell, D.; Andersen, R.; Werthammer, J.; et al. Intrauterine herpes simplex virus infections. J. Pediatr. 1987, 110, 97–101. [Google Scholar] [CrossRef]
  152. Koi, H.; Zhang, J.; Makrigiannakis, A.; Getsios, S.; MacCalman, C.D.; Strauss, J.F., 3rd; Parry, S. Syncytiotrophoblast is a barrier to maternal-fetal transmission of herpes simplex virus. Biol. Reprod. 2002, 67, 1572–1579. [Google Scholar] [CrossRef] [Green Version]
  153. Norskov-Lauritsen, N.; Aboagye-Mathisen, G.; Juhl, C.B.; Petersen, P.M.; Zachar, V.; Ebbesen, P. Herpes simplex virus infection of cultured human term trophoblast. J. Med. Virol. 1992, 36, 162–166. [Google Scholar] [CrossRef]
  154. Oliveira, L.H.; Fonseca, M.E.; de-Bonis, M. Echovirus type 19 and herpes simplex type 2 infection in human placenta tissue explants. Braz. J. Med. Biol. Res. 1993, 26, 703–717. [Google Scholar]
  155. Coffey, V.P.; Jessop, W.J. Maternal influenza and congenital deformities: A prospective study. Lancet 1959, 2, 935–938. [Google Scholar] [CrossRef]
  156. Kwit, K.; Pomorska-Mol, M.; Markowska-Daniel, I. Pregnancy outcome and clinical status of gilts following experimental infection by H1N2, H3N2 and H1N1pdm09 influenza a viruses during the last month of gestation. Arch. Virol. 2015, 160, 2415–2425. [Google Scholar] [CrossRef] [Green Version]
  157. Kourtis, A.P.; Read, J.S.; Jamieson, D.J. Pregnancy and infection. N. Engl. J. Med. 2014, 370, 2211–2218. [Google Scholar] [CrossRef] [Green Version]
  158. Harris, J.W. Influenza occurring in pregnant women: A statistical study of thirteen hundred and fifty cases. J. Am. Med. Assoc. 1919, 72, 978–980. [Google Scholar] [CrossRef]
  159. Nuzum, J.W.; Pilot, I.; Stangl, F.H.; Bonar, B.E. Pandemic influenza and pneumonia in a large civilian hospital. J. Am. Med. Assoc. 1918, 71, 1562–1567. [Google Scholar] [CrossRef] [Green Version]
  160. Siston, A.M.; Rasmussen, S.A.; Honein, M.A.; Fry, A.M.; Seib, K.; Callaghan, W.M.; Louie, J.; Doyle, T.J.; Crockett, M.; Lynfield, R.; et al. Pandemic 2009 influenza A(H1N1) virus illness among pregnant women in the united states. JAMA 2010, 303, 1517–1525. [Google Scholar] [CrossRef]
  161. Jamieson, D.J.; Honein, M.A.; Rasmussen, S.A.; Williams, J.L.; Swerdlow, D.L.; Biggerstaff, M.S.; Lindstrom, S.; Louie, J.K.; Christ, C.M.; Bohm, S.R.; et al. H1N1 2009 influenza virus infection during pregnancy in the USA. Lancet 2009, 374, 451–458. [Google Scholar] [CrossRef]
  162. Wilson, M.G.; Stein, A.M. Teratogenic effects of asian influenza. An extended study. JAMA 1969, 210, 336–337. [Google Scholar] [CrossRef]
  163. Zou, S. Potential impact of pandemic influenza on blood safety and availability. Transfus. Med. Rev. 2006, 20, 181–189. [Google Scholar] [CrossRef]
  164. Likos, A.M.; Kelvin, D.J.; Cameron, C.M.; Rowe, T.; Kuehnert, M.J.; Norris, P.J.; National Heart, Lung, Blood Institute Retrovirus Epidemiology Donor Study-II (REDS-II). Influenza viremia and the potential for blood-borne transmission. Transfusion 2007, 47, 1080–1088. [Google Scholar] [CrossRef] [Green Version]
  165. Tsuruoka, H.; Xu, H.; Kuroda, K.; Hayashi, K.; Yasui, O.; Yamada, A.; Ishizaki, T.; Yamada, Y.; Watanabe, T.; Hosaka, Y. Detection of influenza virus rna in peripheral blood mononuclear cells of influenza patients. Jpn. J. Med. Sci. Biol. 1997, 50, 27–34. [Google Scholar] [CrossRef] [Green Version]
  166. Komine-Aizawa, S.; Suzaki, A.; Trinh, Q.D.; Izumi, Y.; Shibata, T.; Kuroda, K.; Hayakawa, S. H1N1/09 influenza a virus infection of immortalized first trimester human trophoblast cell lines. Am. J. Reprod. Immunol. 2012, 68, 226–232. [Google Scholar] [CrossRef]
  167. Trinh, Q.D.; Izumi, Y.; Komine-Aizawa, S.; Shibata, T.; Shimotai, Y.; Kuroda, K.; Mizuguchi, M.; Ushijima, H.; Mor, G.; Hayakawa, S. H3N2 influenza a virus replicates in immortalized human first trimester trophoblast cell lines and induces their rapid apoptosis. Am. J. Reprod. Immunol. 2009, 62, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Besnard, M.; Lastere, S.; Teissier, A.; Cao-Lormeau, V.; Musso, D. Evidence of perinatal transmission of zika virus, french polynesia, december 2013 and february 2014. Eurosurveillance 2014, 19, 20751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Cao-Lormeau, V.M.; Roche, C.; Teissier, A.; Robin, E.; Berry, A.L.; Mallet, H.P.; Sall, A.A.; Musso, D. Zika virus, french polynesia, south pacific, 2013. Emerg. Infect. Dis. 2014, 20, 1085–1086. [Google Scholar] [CrossRef] [PubMed]
  170. Duffy, M.R.; Chen, T.H.; Hancock, W.T.; Powers, A.M.; Kool, J.L.; Lanciotti, R.S.; Pretrick, M.; Marfel, M.; Holzbauer, S.; Dubray, C.; et al. Zika virus outbreak on yap island, federated states of micronesia. N. Engl. J. Med. 2009, 360, 2536–2543. [Google Scholar] [CrossRef] [PubMed]
  171. Brasil, P.; Calvet, G.A.; Siqueira, A.M.; Wakimoto, M.; De Sequeira, P.C.; Nobre, A.; Quintana, M.D.S.B.; De Mendonça, M.C.L.; Lupi, O.; De Souza, R.V.; et al. Zika virus outbreak in rio de janeiro, brazil: Clinical characterization, epidemiological and virological aspects. PLoS Negl. Trop. Dis. 2016, 10, e0004636. [Google Scholar] [CrossRef] [Green Version]
  172. Metsky, H.C.; Matranga, C.B.; Wohl, S.; Schaffner, S.F.; Freije, C.A.; Winnicki, S.M.; West, K.; Qu, J.; Baniecki, M.L.; Gladden-Young, A.; et al. Zika virus evolution and spread in the americas. Nature 2017, 546, 411–415. [Google Scholar] [CrossRef] [Green Version]
  173. Hamanaka, T.; Ribeiro, C.T.M.; Pone, S.; Gomes, S.C.; Nielsen-Saines, K.; Brickley, E.B.; Moreira, M.E.; Pone, M. Longitudinal follow-up of gross motor function in children with congenital zika virus syndrome from a cohort in rio de janeiro, brazil. Viruses 2022, 14, 1173. [Google Scholar] [CrossRef]
  174. Lazear, H.M.; Diamond, M.S. Zika virus: New clinical syndromes and its emergence in the western hemisphere. J. Virol. 2016, 90, 4864–4875. [Google Scholar] [CrossRef] [Green Version]
  175. Miao, J.; Yuan, H.; Rao, J.; Zou, J.; Yang, K.; Peng, G.; Cao, S.; Chen, H.; Song, Y. Identification of a small compound that specifically inhibits zika virus in vitro and in vivo by targeting the ns2b-ns3 protease. Antivir. Res. 2022, 199, 105255. [Google Scholar] [CrossRef]
  176. Luria-Perez, R.; Sanchez-Vargas, L.A.; Munoz-Lopez, P.; Mellado-Sanchez, G. Mucosal vaccination: A promising alternative against flaviviruses. Front. Cell. Infect. Microbiol. 2022, 12, 887729. [Google Scholar] [CrossRef]
  177. Kim, I.J.; Lanthier, P.A.; Clark, M.J.; De La Barrera, R.A.; Tighe, M.P.; Szaba, F.M.; Travis, K.L.; Low-Beer, T.C.; Cookenham, T.S.; Lanzer, K.G.; et al. Efficacy of an inactivated zika vaccine against virus infection during pregnancy in mice and marmosets. NPJ Vaccines 2022, 7, 9. [Google Scholar] [CrossRef]
  178. Kale, A.; Joshi, D.; Menon, I.; Bagwe, P.; Patil, S.; Vijayanand, S.; Braz Gomes, K.; D’Souza, M. Novel microparticulate zika vaccine induces a significant immune response in a preclinical murine model after intramuscular administration. Int. J. Pharm. 2022, 624, 121975. [Google Scholar] [CrossRef]
  179. Tabata, T.; Petitt, M.; Puerta-Guardo, H.; Michlmayr, D.; Wang, C.; Fang-Hoover, J.; Harris, E.; Pereira, L. Zika virus targets different primary human placental cells, suggesting two routes for vertical transmission. Cell Host Microbe 2016, 20, 155–166. [Google Scholar] [CrossRef] [Green Version]
  180. Quicke, K.M.; Bowen, J.R.; Johnson, E.L.; McDonald, C.E.; Ma, H.; O’Neal, J.T.; Rajakumar, A.; Wrammert, J.; Rimawi, B.H.; Pulendran, B.; et al. Zika virus infects human placental macrophages. Cell Host Microbe 2016, 20, 83–90. [Google Scholar] [CrossRef] [Green Version]
  181. Weisblum, Y.; Oiknine-Djian, E.; Vorontsov, O.M.; Haimov-Kochman, R.; Zakay-Rones, Z.; Meir, K.; Shveiky, D.; Elgavish, S.; Nevo, Y.; Roseman, M.; et al. Zika virus infects early- and midgestation human maternal decidual tissues, inducing distinct innate tissue responses in the maternal-fetal interface. J. Virol. 2017, 91, e01905-16. [Google Scholar] [CrossRef] [Green Version]
  182. El Costa, H.; Gouilly, J.; Mansuy, J.M.; Chen, Q.; Levy, C.; Cartron, G.; Veas, F.; Al-Daccak, R.; Izopet, J.; Jabrane-Ferrat, N. Zika virus reveals broad tissue and cell tropism during the first trimester of pregnancy. Sci. Rep. 2016, 6, 35296. [Google Scholar] [CrossRef] [Green Version]
  183. Ashraf, M.A.; Keshavarz, P.; Hosseinpour, P.; Erfani, A.; Roshanshad, A.; Pourdast, A.; Nowrouzi-Sohrabi, P.; Chaichian, S.; Poordast, T. Coronavirus disease 2019 (COVID-19): A systematic review of pregnancy and the possibility of vertical transmission. J. Reprod. Infertil. 2020, 21, 157–168. [Google Scholar]
  184. Tossetta, G.; Fantone, S.; Muti, N.D.; Balercia, G.; Ciavattini, A.; Giannubilo, S.R.; Marzioni, D. Preeclampsia and severe acute respiratory syndrome coronavirus 2 infection: A systematic review. J. Hypertens. 2022, 40, 1629–1638. [Google Scholar] [CrossRef]
  185. He, Z.; Fang, Y.; Zuo, Q.; Huang, X.; Lei, Y.; Ren, X.; Liu, D. Vertical transmission and kidney damage in newborns whose mothers had coronavirus disease 2019 during pregnancy. Int. J. Antimicrob. Agents 2021, 57, 106260. [Google Scholar] [CrossRef]
  186. Kuhrt, K.; McMicking, J.; Nanda, S.; Nelson-Piercy, C.; Shennan, A. Placental abruption in a twin pregnancy at 32 weeks’ gestation complicated by coronavirus disease 2019 without vertical transmission to the babies. Am. J. Obstet. Gynecol. MFM 2020, 2, 100135. [Google Scholar] [CrossRef]
  187. Malhotra, Y.; Rossberg, M.C.; Bajaj, K.; Shtern, A.; Moore, R.M. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vertical transmission in neonates born to mothers with coronavirus disease 2019 (COVID-19) pneumonia. Obstet. Gynecol. 2020, 136, 632–633. [Google Scholar] [CrossRef] [PubMed]
  188. Marzollo, R.; Aversa, S.; Prefumo, F.; Saccani, B.; Perez, C.R.; Sartori, E.; Motta, M. Possible coronavirus disease 2019 pandemic and pregnancy: Vertical transmission is not excluded. Pediatr. Infect. Dis. J. 2020, 39, e261–e262. [Google Scholar] [CrossRef] [PubMed]
  189. Kotlyar, A.M.; Grechukhina, O.; Chen, A.; Popkhadze, S.; Grimshaw, A.; Tal, O.; Taylor, H.S.; Tal, R. Vertical transmission of coronavirus disease 2019: A systematic review and meta-analysis. Am. J. Obstet. Gynecol. 2021, 224, 35–53.e3. [Google Scholar] [CrossRef] [PubMed]
  190. Kotlyar, A.M.; Tal, O.; Tal, R. Vertical transmission of coronavirus disease 2019, a response. Am. J. Obstet. Gynecol. 2021, 224, 329–330. [Google Scholar] [CrossRef] [PubMed]
  191. Li, M.; Chen, L.; Zhang, J.; Xiong, C.; Li, X. The SARS-CoV-2 receptor ace2 expression of maternal-fetal interface and fetal organs by single-cell transcriptome study. PLoS ONE 2020, 15, e0230295. [Google Scholar] [CrossRef] [Green Version]
  192. Algarroba, G.N.; Hanna, N.N.; Rekawek, P.; Vahanian, S.A.; Khullar, P.; Palaia, T.; Peltier, M.R.; Chavez, M.R.; Vintzileos, A.M. Confirmatory evidence of the visualization of severe acute respiratory syndrome coronavirus 2 invading the human placenta using electron microscopy. Am. J. Obstet. Gynecol. 2020, 223, 953–954. [Google Scholar] [CrossRef]
  193. Algarroba, G.N.; Rekawek, P.; Vahanian, S.A.; Khullar, P.; Palaia, T.; Peltier, M.R.; Chavez, M.R.; Vintzileos, A.M. Visualization of severe acute respiratory syndrome coronavirus 2 invading the human placenta using electron microscopy. Am. J. Obstet. Gynecol. 2020, 223, 275–278. [Google Scholar] [CrossRef]
  194. Latifi, Z.; Nejabati, H.R.; Abroon, S.; Mihanfar, A.; Farzadi, L.; Hakimi, P.; Hajipour, H.; Nouri, M.; Fattahi, A. Dual role of tgf-beta in early pregnancy: Clues from tumor progression. Biol. Reprod. 2019, 100, 1417–1430. [Google Scholar] [CrossRef]
  195. Zhang, Y.; Alexander, P.B.; Wang, X.F. Tgf-beta family signaling in the control of cell proliferation and survival. Cold Spring Harb. Perspect. Biol. 2017, 9, a022145. [Google Scholar] [CrossRef] [Green Version]
  196. Hata, A.; Chen, Y.G. Tgf-beta signaling from receptors to smads. Cold Spring Harb. Perspect. Biol. 2016, 8, a022061. [Google Scholar] [CrossRef] [Green Version]
  197. Marinelli Busilacchi, E.; Costantini, A.; Mancini, G.; Tossetta, G.; Olivieri, J.; Poloni, A.; Viola, N.; Butini, L.; Campanati, A.; Goteri, G.; et al. Nilotinib treatment of patients affected by chronic graft-versus-host disease reduces collagen production and skin fibrosis by downmodulating the tgf-beta and p-smad pathway. Biol. Blood Marrow Transplant. 2020, 26, 823–834. [Google Scholar] [CrossRef]
  198. Monsivais, D.; Matzuk, M.M.; Pangas, S.A. The tgf-beta family in the reproductive tract. Cold Spring Harb. Perspect. Biol. 2017, 9, a022251. [Google Scholar] [CrossRef] [Green Version]
  199. Wu, M.Y.; Hill, C.S. Tgf-beta superfamily signaling in embryonic development and homeostasis. Dev. Cell 2009, 16, 329–343. [Google Scholar] [CrossRef] [Green Version]
  200. Jones, R.L.; Stoikos, C.; Findlay, J.K.; Salamonsen, L.A. Tgf-beta superfamily expression and actions in the endometrium and placenta. Reproduction 2006, 132, 217–232. [Google Scholar] [CrossRef]
  201. Ciebiera, M.; Wlodarczyk, M.; Wrzosek, M.; Meczekalski, B.; Nowicka, G.; Lukaszuk, K.; Ciebiera, M.; Slabuszewska-Jozwiak, A.; Jakiel, G. Role of transforming growth factor beta in uterine fibroid biology. Int. J. Mol. Sci. 2017, 18, 2435. [Google Scholar] [CrossRef] [Green Version]
  202. Licini, C.; Tossetta, G.; Avellini, C.; Ciarmela, P.; Lorenzi, T.; Toti, P.; Gesuita, R.; Voltolini, C.; Petraglia, F.; Castellucci, M.; et al. Analysis of cell-cell junctions in human amnion and chorionic plate affected by chorioamnionitis. Histol. Histopathol. 2016, 31, 759–767. [Google Scholar]
  203. Tossetta, G.; Paolinelli, F.; Avellini, C.; Salvolini, E.; Ciarmela, P.; Lorenzi, T.; Emanuelli, M.; Toti, P.; Giuliante, R.; Gesuita, R.; et al. Il-1beta and tgf-beta weaken the placental barrier through destruction of tight junctions: An in vivo and in vitro study. Placenta 2014, 35, 509–516. [Google Scholar] [CrossRef]
  204. Ma, W.; Qin, Y.; Chapuy, B.; Lu, C. Lrrc33 is a novel binding and potential regulating protein of tgf-beta1 function in human acute myeloid leukemia cells. PLoS ONE 2019, 14, e0213482. [Google Scholar] [CrossRef] [Green Version]
  205. Lash, G.E.; Naruse, K.; Innes, B.A.; Robson, S.C.; Searle, R.F.; Bulmer, J.N. Secretion of angiogenic growth factors by villous cytotrophoblast and extravillous trophoblast in early human pregnancy. Placenta 2010, 31, 545–548. [Google Scholar] [CrossRef]
  206. Simpson, H.; Robson, S.C.; Bulmer, J.N.; Barber, A.; Lyall, F. Transforming growth factor beta expression in human placenta and placental bed during early pregnancy. Placenta 2002, 23, 44–58. [Google Scholar] [CrossRef]
  207. Svensson-Arvelund, J.; Mehta, R.B.; Lindau, R.; Mirrasekhian, E.; Rodriguez-Martinez, H.; Berg, G.; Lash, G.E.; Jenmalm, M.C.; Ernerudh, J. The human fetal placenta promotes tolerance against the semiallogeneic fetus by inducing regulatory t cells and homeostatic m2 macrophages. J. Immunol. 2015, 194, 1534–1544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Takano, C.; Horie, M.; Taiko, I.; Trinh, Q.D.; Kanemaru, K.; Komine-Aizawa, S.; Hayakawa, S.; Miki, T. Inhibition of epithelial-mesenchymal transition maintains stemness in human amniotic epithelial cells. Stem Cell Rev. Rep. 2022, 18, 3083–3091. [Google Scholar] [CrossRef] [PubMed]
  209. Schliefsteiner, C.; Ibesich, S.; Wadsack, C. Placental hofbauer cell polarization resists inflammatory cues in vitro. Int. J. Mol. Sci. 2020, 21, 736. [Google Scholar] [CrossRef] [Green Version]
  210. Del Gobbo, V.; Giganti, M.G.; Zenobi, R.; Villani, V.; Premrov, M.G. The immunosuppressive cytokines influence the fetal survival in patients with pregnancy-induced hypertension. Am. J. Reprod. Immunol. 2000, 44, 214–221. [Google Scholar] [CrossRef] [PubMed]
  211. Huang, N.; Chi, H.; Qiao, J. Role of regulatory t cells in regulating fetal-maternal immune tolerance in healthy pregnancies and reproductive diseases. Front. Immunol. 2020, 11, 1023. [Google Scholar] [CrossRef]
  212. Jorgensen, N.; Persson, G.; Hviid, T.V.F. The tolerogenic function of regulatory t cells in pregnancy and cancer. Front. Immunol. 2019, 10, 911. [Google Scholar] [CrossRef] [Green Version]
  213. Benian, A.; Madazli, R.; Aksu, F.; Uzun, H.; Aydin, S. Plasma and placental levels of interleukin-10, transforming growth factor-beta1, and epithelial-cadherin in preeclampsia. Obstet. Gynecol. 2002, 100, 327–331. [Google Scholar]
  214. Djurovic, S.; Schjetlein, R.; Wisloff, F.; Haugen, G.; Husby, H.; Berg, K. Plasma concentrations of lp(a) lipoprotein and tgf-beta1 are altered in preeclampsia. Clin. Genet. 1997, 52, 371–376. [Google Scholar] [CrossRef]
  215. Abudukeyoumu, A.; Li, M.Q.; Xie, F. Transforming growth factor-beta1 in intrauterine adhesion. Am. J. Reprod. Immunol. 2020, 84, e13262. [Google Scholar] [CrossRef]
  216. Goteri, G.; Altobelli, E.; Tossetta, G.; Zizzi, A.; Avellini, C.; Licini, C.; Lorenzi, T.; Castellucci, M.; Ciavattini, A.; Marzioni, D. High temperature requirement a1, transforming growth factor beta1, phosphosmad2 and ki67 in eutopic and ectopic endometrium of women with endometriosis. Eur. J. Histochem. 2015, 59, 2570. [Google Scholar] [CrossRef] [Green Version]
  217. Rallon, N.I.; Barreiro, P.; Soriano, V.; Garcia-Samaniego, J.; Lopez, M.; Benito, J.M. Elevated tgf-beta1 levels might protect HCV/HIV-coinfected patients from liver fibrosis. Eur. J. Clin. Investig. 2011, 41, 70–76. [Google Scholar] [CrossRef]
  218. Vadaq, N.; van de Wijer, L.; van Eekeren, L.E.; Koenen, H.; de Mast, Q.; Joosten, L.A.B.; Netea, M.G.; Matzaraki, V.; van der Ven, A. Targeted plasma proteomics reveals upregulation of distinct inflammatory pathways in people living with HIV. iScience 2022, 25, 105089. [Google Scholar] [CrossRef]
  219. Brancaccio, M.; Mennitti, C.; Calvanese, M.; Gentile, A.; Musto, R.; Gaudiello, G.; Scamardella, G.; Terracciano, D.; Frisso, G.; Pero, R.; et al. Diagnostic and therapeutic potential for hnp-1, hbd-1 and hbd-4 in pregnant women with COVID-19. Int. J. Mol. Sci. 2022, 23, 3450. [Google Scholar] [CrossRef]
  220. Gwon, Y.D.; Mahani, S.A.N.; Nagaev, I.; Mincheva-Nilsson, L.; Evander, M. Rift valley fever virus propagates in human villous trophoblast cell lines and induces cytokine mrna responses known to provoke miscarriage. Viruses 2021, 13, 2265. [Google Scholar] [CrossRef]
  221. Kumar, A.; Devi, S.G.; Kar, P.; Agarwal, S.; Husain, S.A.; Gupta, R.K.; Sharma, S. Association of cytokines in hepatitis e with pregnancy outcome. Cytokine 2014, 65, 95–104. [Google Scholar] [CrossRef]
  222. Periolo, N.; Avaro, M.; Czech, A.; Russo, M.; Benedetti, E.; Pontoriero, A.; Campos, A.; Peralta, L.M.; Baumeister, E. Pregnant women infected with pandemic influenza A(H1N1)pdm09 virus showed differential immune response correlated with disease severity. J. Clin. Virol. 2015, 64, 52–58. [Google Scholar] [CrossRef]
  223. Salomao, N.; Rabelo, K.; Avvad-Portari, E.; Basilio-de-Oliveira, C.; Basilio-de-Oliveira, R.; Ferreira, F.; Ferreira, L.; de Souza, T.M.; Nunes, P.; Lima, M.; et al. Histopathological and immunological characteristics of placentas infected with chikungunya virus. Front. Microbiol. 2022, 13, 1055536. [Google Scholar] [CrossRef]
  224. Helantera, I.; Loginov, R.; Koskinen, P.; Tornroth, T.; Gronhagen-Riska, C.; Lautenschlager, I. Persistent cytomegalovirus infection is associated with increased expression of tgf-beta1, pdgf-aa and icam-1 and arterial intimal thickening in kidney allografts. Nephrol. Dial. Transplant. 2005, 20, 790–796. [Google Scholar] [CrossRef] [Green Version]
  225. Shimamura, M.; Murphy-Ullrich, J.E.; Britt, W.J. Human cytomegalovirus induces tgf-beta1 activation in renal tubular epithelial cells after epithelial-to-mesenchymal transition. PLoS Pathog. 2010, 6, e1001170. [Google Scholar] [CrossRef] [Green Version]
  226. Chen, G.; Yang, X.; Wang, B.; Cheng, Z.; Zhao, R. Human cytomegalovirus promotes the activation of tgf-beta1 in human umbilical vein endothelial cells by mmp-2 after endothelial mesenchymal transition. Adv. Clin. Exp. Med. 2019, 28, 1441–1450. [Google Scholar] [CrossRef]
  227. Zhu, X.; Hu, B.; Hu, M.; Qian, D.; Wang, B. Human cytomegalovirus infection enhances invasiveness and migration of glioblastoma cells by epithelial-to-mesenchymal transition. Int. J. Clin. Exp. Pathol. 2020, 13, 2637–2647. [Google Scholar] [PubMed]
  228. Reinhold, D.; Wrenger, S.; Kahne, T.; Ansorge, S. HIV-1 tat: Immunosuppression via tgf-beta1 induction. Immunol. Today 1999, 20, 384–385. [Google Scholar] [CrossRef] [PubMed]
  229. Lin, W.; Weinberg, E.M.; Tai, A.W.; Peng, L.F.; Brockman, M.A.; Kim, K.A.; Kim, S.S.; Borges, C.B.; Shao, R.X.; Chung, R.T. HIV increases HCV replication in a tgf-beta1-dependent manner. Gastroenterology 2008, 134, 803–811. [Google Scholar] [CrossRef] [PubMed]
  230. Kafka, J.K.; Osborne, B.J.; Sheth, P.M.; Nazli, A.; Dizzell, S.; Huibner, S.; Kovacs, C.; Verschoor, C.P.; Bowdish, D.M.; Kaul, R.; et al. Latent tgf-beta1 is compartmentalized between blood and seminal plasma of HIV-positive men and its activation in semen is negatively correlated with viral load and immune activation. Am. J. Reprod. Immunol. 2015, 73, 151–161. [Google Scholar] [CrossRef] [PubMed]
  231. Dickinson, M.; Kliszczak, A.E.; Giannoulatou, E.; Peppa, D.; Pellegrino, P.; Williams, I.; Drakesmith, H.; Borrow, P. Dynamics of transforming growth factor (tgf)-beta superfamily cytokine induction during HIV-1 infection are distinct from other innate cytokines. Front. Immunol. 2020, 11, 596841. [Google Scholar] [CrossRef]
  232. Chinnapaiyan, S.; Parira, T.; Dutta, R.; Agudelo, M.; Morris, A.; Nair, M.; Unwalla, H.J. HIV infects bronchial epithelium and suppresses components of the mucociliary clearance apparatus. PLoS ONE 2017, 12, e0169161. [Google Scholar] [CrossRef] [Green Version]
  233. Lien, K.; Mayer, W.; Herrera, R.; Padilla, N.T.; Cai, X.; Lin, V.; Pholcharoenchit, R.; Palefsky, J.; Tugizov, S.M. HIV-1 proteins gp120 and tat promote epithelial-mesenchymal transition and invasiveness of HPV-positive and HPV-negative neoplastic genital and oral epithelial cells. Microbiol. Spectr. 2022, 10, e0362222. [Google Scholar] [CrossRef]
  234. Pan, J.; Clayton, M.; Feitelson, M.A. Hepatitis b virus x antigen promotes transforming growth factor-beta1 (tgf-beta1) activity by up-regulation of tgf-beta1 and down-regulation of alpha2-macroglobulin. J. Gen. Virol. 2004, 85, 275–282. [Google Scholar] [CrossRef]
  235. Flisiak, R.; Jaroszewicz, J.; Lapinski, T.W.; Flisiak, I.; Rogalska, M.; Prokopowicz, D. Plasma transforming growth factor beta1, metalloproteinase-1 and tissue inhibitor of metalloproteinases-1 in acute viral hepatitis type b. Regul. Pept. 2005, 131, 54–58. [Google Scholar] [CrossRef]
  236. Guo, G.H.; Tan, D.M.; Zhu, P.A.; Liu, F. Hepatitis b virus x protein promotes proliferation and upregulates tgf-beta1 and ctgf in human hepatic stellate cell line, lx-2. Hepatobiliary Pancreat. Dis. Int. 2009, 8, 59–64. [Google Scholar]
  237. Ming, D.; Yu, X.; Guo, R.; Deng, Y.; Li, J.; Lin, C.; Su, M.; Lin, Z.; Su, Z. Elevated tgf-beta1/il-31 pathway is associated with the disease severity of hepatitis b virus-related liver cirrhosis. Viral Immunol. 2015, 28, 209–216. [Google Scholar] [CrossRef]
  238. Li, M.H.; Chen, Q.Q.; Zhang, L.; Lu, H.H.; Sun, F.F.; Zeng, Z.; Lu, Y.; Yi, W.; Xie, Y. Association of cytokines with hepatitis b virus and its antigen. J. Med. Virol. 2020, 92, 3426–3435. [Google Scholar] [CrossRef]
  239. Li, M.H.; Lu, Y.; Sun, F.F.; Chen, Q.Q.; Zhang, L.; Lu, H.H.; Zeng, Z.; Yi, W.; Xie, Y. Transforming growth factor beta as a possible independent factor in chronic hepatitis b. Arch. Virol. 2021, 166, 1853–1858. [Google Scholar] [CrossRef]
  240. Yu, X.; Guo, R.; Ming, D.; Deng, Y.; Su, M.; Lin, C.; Li, J.; Lin, Z.; Su, Z. The transforming growth factor beta1/interleukin-31 pathway is upregulated in patients with hepatitis b virus-related acute-on-chronic liver failure and is associated with disease severity and survival. Clin. Vaccine Immunol. 2015, 22, 484–492. [Google Scholar] [CrossRef] [Green Version]
  241. Li, W.; Duan, X.; Zhu, C.; Liu, X.; Jeyarajan, A.J.; Xu, M.; Tu, Z.; Sheng, Q.; Chen, D.; Zhu, C.; et al. Hepatitis b and hepatitis c virus infection promote liver fibrogenesis through a tgf-beta1-induced oct4/nanog pathway. J. Immunol. 2022, 208, 672–684. [Google Scholar] [CrossRef]
  242. Wang, Y.; Zhao, L.F.; Wang, R.R.; Zhi, S.W. [effect of transforming growth factor-beta1 on HBV replication and antigen synthesis in hepg2.2.15 cells with steatosis]. Zhonghua Gan Zang Bing Za Zhi 2017, 25, 732–737. [Google Scholar]
  243. Mendez-Samperio, P.; Hernandez, M.; Ayala, H.E. Induction of transforming growth factor-beta 1 production in human cells by herpes simplex virus. J. Interf. Cytokine Res. 2000, 20, 273–280. [Google Scholar] [CrossRef]
  244. Kaygusuz, I.; Godekmerdan, A.; Keles, E.; Karlidag, T.; Yalcin, S.; Yildiz, M.; Tazegul, A. The role of viruses in idiopathic peripheral facial palsy and cellular immune response. Am. J. Otolaryngol. 2004, 25, 401–406. [Google Scholar] [CrossRef]
  245. Nie, Y.; Cui, D.; Pan, Z.; Deng, J.; Huang, Q.; Wu, K. HSV-1 infection suppresses tgf-beta1 and smad3 expression in human corneal epithelial cells. Mol. Vis. 2008, 14, 1631–1638. [Google Scholar]
  246. Choi, J.A.; Ju, H.H.; Kim, J.E.; Lee, J.; Jee, D.; Park, C.K.; Paik, S.Y. Cytokine profile and cytoskeletal changes after herpes simplex virus type 1 infection in human trabecular meshwork cells. J. Cell. Mol. Med. 2021, 25, 9295–9305. [Google Scholar] [CrossRef]
  247. Drevets, P.; Chucair-Elliott, A.; Shrestha, P.; Jinkins, J.; Karamichos, D.; Carr, D.J. The use of human cornea organotypic cultures to study herpes simplex virus type 1 (HSV-1)-induced inflammation. Graefes Arch. Clin. Exp. Ophthalmol. 2015, 253, 1721–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Wen, Y.; Deng, B.C.; Zhou, Y.; Wang, Y.; Cui, W.; Wang, W.; Liu, P. Immunological features in patients with pneumonitis due to influenza a H1N1 infection. J. Investig. Allergol. Clin. Immunol. 2011, 21, 44–50. [Google Scholar] [PubMed]
  249. Li, C.; Jiao, S.; Wang, G.; Gao, Y.; Liu, C.; He, X.; Zhang, C.; Xiao, J.; Li, W.; Zhang, G.; et al. The immune adaptor adap regulates reciprocal tgf-beta1-integrin crosstalk to protect from influenza virus infection. PLoS Pathog. 2015, 11, e1004824. [Google Scholar] [CrossRef] [PubMed]
  250. Bischoff, A.L.; Folsgaard, N.V.; Vissing, N.H.; Birch, S.; Brix, S.; Bisgaard, H. Airway mucosal immune-suppression in neonates of mothers receiving A(H1N1)pnd09 vaccination during pregnancy. Pediatr. Infect. Dis. J. 2015, 34, 84–90. [Google Scholar] [CrossRef] [Green Version]
  251. BustosRivera-Bahena, G.; Lopez-Guerrero, D.V.; Marquez-Bandala, A.H.; Esquivel-Guadarrama, F.R.; Montiel-Hernandez, J.L. Tgf-beta1 signaling inhibit the in vitro apoptotic, infection and stimulatory cell response induced by influenza H1N1 virus infection on a549 cells. Virus Res. 2021, 297, 198337. [Google Scholar] [CrossRef]
  252. de Sousa, J.R.; Azevedo, R.S.S.; Martins Filho, A.J.; Araujo, M.T.F.; Moutinho, E.R.C.; Baldez Vasconcelos, B.C.; Cruz, A.C.R.; Oliveira, C.S.; Martins, L.C.; Baldez Vasconcelos, B.H.; et al. Correlation between apoptosis and in situ immune response in fatal cases of microcephaly caused by zika virus. Am. J. Pathol. 2018, 188, 2644–2652. [Google Scholar] [CrossRef] [Green Version]
  253. Jiyarom, B.; Giannakopoulos, S.; Strange, D.P.; Panova, N.; Gale, M., Jr.; Verma, S. Rig-i and mda5 are modulated by bone morphogenetic protein (bmp6) and are essential for restricting zika virus infection in human sertoli cells. Front. Microbiol. 2022, 13, 1062499. [Google Scholar] [CrossRef]
  254. Karadeniz, H.; Avanoglu Guler, A.; Ozger, H.S.; Yildiz, P.A.; Erbas, G.; Bozdayi, G.; Deveci Bulut, T.; Gulbahar, O.; Yapar, D.; Kucuk, H.; et al. The prognostic value of lung injury and fibrosis markers, kl-6, tgf-beta1, fgf-2 in COVID-19 patients. Biomark. Insights 2022, 17, 11772719221135443. [Google Scholar] [CrossRef]
  255. Laloglu, E.; Alay, H. Role of transforming growth factor-beta 1 and connective tissue growth factor levels in coronavirus disease-2019-related lung injury: A prospective, observational, cohort study. Rev. Soc. Bras. Med. Trop. 2022, 55, e06152021. [Google Scholar] [CrossRef]
  256. Mezger, M.C.; Conzelmann, C.; Weil, T.; von Maltitz, P.; Albers, D.P.J.; Munch, J.; Stamminger, T.; Schilling, E.M. Inhibitors of activin receptor-like kinase 5 interfere with SARS-CoV-2 s-protein processing and spike-mediated cell fusion via attenuation of furin expression. Viruses 2022, 14, 1308. [Google Scholar] [CrossRef]
  257. Zachar, V.; Fink, T.; Koppelhus, U.; Ebbesen, P. Role of placental cytokines in transcriptional modulation of HIV type 1 in the isolated villous trophoblast. AIDS Res. Hum. Retroviruses 2002, 18, 839–847. [Google Scholar] [CrossRef]
  258. Cui, H.; Li, Q.L.; Chen, J.; Na, Q.; Liu, C.X. Hepatitis b virus x protein modifies invasion, proliferation and the inflammatory response in an htr-8/svneo cell model. Oncol. Rep. 2015, 34, 2090–2098. [Google Scholar] [CrossRef] [Green Version]
  259. Liu, T.; Zheng, X.; Li, Q.; Chen, J.; Yin, Z.; Xiao, J.; Zhang, D.; Li, W.; Qiao, Y.; Chen, S. Role of human cytomegalovirus in the proliferation and invasion of extravillous cytotrophoblasts isolated from early placentae. Int. J. Clin. Exp. Med. 2015, 8, 17248–17260. [Google Scholar]
  260. Busnadiego, O.; Gonzalez-Santamaria, J.; Lagares, D.; Guinea-Viniegra, J.; Pichol-Thievend, C.; Muller, L.; Rodriguez-Pascual, F. LOXL4 is induced by transforming growth factor β1 through Smad and JunB/Fra2 and contributes to vascular matrix remodeling. Mol. Cell. Biol. 2013, 33, 2388–2401. [Google Scholar] [CrossRef] [Green Version]
  261. Booth, L.; Roberts, J.L.; Cash, D.R.; Tavallai, S.; Jean, S.; Fidanza, A.; Cruz-Luna, T.; Siembiba, P.; Cycon, K.A.; Cornelissen, C.N.; et al. GRP78/BiP/HSPA5/Dna K is a universal therapeutic target for human disease. J. Cell Physiol. 2015, 230, 1661–1676. [Google Scholar] [CrossRef] [Green Version]
  262. Elfiky, A.A.; Ibrahim, I.M.; Elgohary, A.M. SARS-CoV-2 delta variant is recognized through GRP78 host-cell surface receptor, in silico perspective. Int. J. Pept. Res. Ther. 2022, 28, 146. [Google Scholar] [CrossRef]
  263. Gonzalez-Gronow, M.; Gopal, U.; Austin, R.C.; Pizzo, S.V. Glucose-regulated protein (GRP78) is an important cell surface receptor for viral invasion, cancers, and neurological disorders. IUBMB Life 2021, 73, 843–854. [Google Scholar] [CrossRef]
  264. Khongwichit, S.; Sornjai, W.; Jitobaom, K.; Greenwood, M.; Greenwood, M.P.; Hitakarun, A.; Wikan, N.; Murphy, D.; Smith, D.R. A functional interaction between GRP78 and zika virus e protein. Sci. Rep. 2021, 11, 393. [Google Scholar] [CrossRef]
  265. Nain, M.; Mukherjee, S.; Karmakar, S.P.; Paton, A.W.; Paton, J.C.; Abdin, M.Z.; Basu, A.; Kalia, M.; Vrati, S. GRP78 is an important host factor for Japanese encephalitis virus entry and replication in mammalian cells. J. Virol. 2017, 91, e02274-16. [Google Scholar] [CrossRef] [Green Version]
  266. Prusty, B.K.; Siegl, C.; Gulve, N.; Mori, Y.; Rudel, T. Gp96 interacts with hhv-6 during viral entry and directs it for cellular degradation. PLoS ONE 2014, 9, e113962. [Google Scholar] [CrossRef] [Green Version]
  267. Pujhari, S.; Brustolin, M.; Macias, V.M.; Nissly, R.H.; Nomura, M.; Kuchipudi, S.V.; Rasgon, J.L. Heat shock protein 70 (hsp70) mediates zika virus entry, replication, and egress from host cells. Emerg. Microbes Infect. 2019, 8, 8–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Ma, J.; Jia, J.; Jiang, X.; Xu, M.; Guo, J.; Tang, T.; Xu, X.; Wu, Z.; Hu, B.; Yao, K.; et al. Gp96 is critical for both human herpesvirus 6a (hhv-6a) and hhv-6b infections. J. Virol. 2020, 94, e00311-20. [Google Scholar] [CrossRef] [PubMed]
  269. Taylor, J.R.; Skeate, J.G.; Kast, W.M. Annexin a2 in virus infection. Front. Microbiol. 2018, 9, 2954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  270. Dawar, F.U.; Tu, J.; Khattak, M.N.; Mei, J.; Lin, L. Cyclophilin a: A key factor in virus replication and potential target for anti-viral therapy. Curr. Issues Mol. Biol. 2017, 21, 1–20. [Google Scholar]
  271. Yang, S.L.; Chou, Y.T.; Wu, C.N.; Ho, M.S. Annexin ii binds to capsid protein vp1 of enterovirus 71 and enhances viral infectivity. J. Virol. 2011, 85, 11809–11820. [Google Scholar] [CrossRef] [Green Version]
  272. Gonzalez-Reyes, S.; Garcia-Manso, A.; Del Barrio, G.; Dalton, K.P.; Gonzalez-Molleda, L.; Arrojo-Fernandez, J.; Nicieza, I.; Parra, F. Role of annexin a2 in cellular entry of rabbit vesivirus. J. Gen. Virol. 2009, 90, 2724–2730. [Google Scholar] [CrossRef]
  273. Braaten, D.; Franke, E.K.; Luban, J. Cyclophilin a is required for an early step in the life cycle of human immunodeficiency virus type 1 before the initiation of reverse transcription. J. Virol. 1996, 70, 3551–3560. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Viral transmission to pregnant mother and her fetus/baby. (A) Transmission routes to pregnant mother. (B) Vertical transmission routes. (C) Intrauterine transmission-possible mechanisms of virus invasion into the placenta barrier. Abbreviations: RuV, rubella virus; HCMV, human cytomegalovirus; HIV, human immunodeficiency virus; HBV, hepatitis B virus; HSV, herpes simplex virus; IAV, influenza A virus; ZIKV, Zika virus; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; CBT, cytotrophoblast; STB, syncytiotrophoblast; EVT, extravillous trophoblast; EC, endothelial cells; T reg, regulatory T cells.
Figure 1. Viral transmission to pregnant mother and her fetus/baby. (A) Transmission routes to pregnant mother. (B) Vertical transmission routes. (C) Intrauterine transmission-possible mechanisms of virus invasion into the placenta barrier. Abbreviations: RuV, rubella virus; HCMV, human cytomegalovirus; HIV, human immunodeficiency virus; HBV, hepatitis B virus; HSV, herpes simplex virus; IAV, influenza A virus; ZIKV, Zika virus; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; CBT, cytotrophoblast; STB, syncytiotrophoblast; EVT, extravillous trophoblast; EC, endothelial cells; T reg, regulatory T cells.
Ijms 24 06489 g001
Figure 2. Trophoblasts, microvillous structure and the published effects of TGF-β1 on viral infection in trophoblasts at the maternal–fetal interface. ❶, TGF-β1 increases the infection and replication of HCMV in STBs through macrophage contact [26]; ❷, enhancement of ZIKV infection and replication in trophoblasts by TGF-β1 [34]; ❸, no effect on HIV construct replication in trophoblasts under TGF-β1 treatment [257]; ❹, increases HBx-transfected trophoblast proliferation and invasion [258]. Abbreviations: HCMV, human cytomegalovirus; HBV, hepatitis B virus; HIV, human immunodeficiency virus; ZIKV, Zika virus; CBT, cytotrophoblast; STB, syncytiotrophoblast; EVT, extravillous trophoblast; T reg, regulatory T cells.
Figure 2. Trophoblasts, microvillous structure and the published effects of TGF-β1 on viral infection in trophoblasts at the maternal–fetal interface. ❶, TGF-β1 increases the infection and replication of HCMV in STBs through macrophage contact [26]; ❷, enhancement of ZIKV infection and replication in trophoblasts by TGF-β1 [34]; ❸, no effect on HIV construct replication in trophoblasts under TGF-β1 treatment [257]; ❹, increases HBx-transfected trophoblast proliferation and invasion [258]. Abbreviations: HCMV, human cytomegalovirus; HBV, hepatitis B virus; HIV, human immunodeficiency virus; ZIKV, Zika virus; CBT, cytotrophoblast; STB, syncytiotrophoblast; EVT, extravillous trophoblast; T reg, regulatory T cells.
Ijms 24 06489 g002
Figure 3. The Smad pathway and possible underlying mechanisms of the published effects of TGF-β1 on ZIKV infection in relation to the virus life cycle.
Figure 3. The Smad pathway and possible underlying mechanisms of the published effects of TGF-β1 on ZIKV infection in relation to the virus life cycle.
Ijms 24 06489 g003
Table 1. Summary of general information related to the reviewed viral infections.
Table 1. Summary of general information related to the reviewed viral infections.
VirusTransmission RoutesMajor Infected Cells/OrgansPregnant and Fetal OutcomesVaccine AvailabilityRepresentative References
RuVRespiratory tract. Direct or droplet contactRespiratory mucosa and cervical lymph nodes. Others: skin, eye, brainCRSYes[48,49,50,51,52]
HCMVThrough bodily fluids: saliva, urine, blood, breast milkEpithelial cells, fibroblasts, endothelial cells, and immune cellsCongenital CMV infectionNo[53,54,55,56]
HIVSexual contact, sharing injecting equipmentImmune system, primarily targeting CD4+ T cellsIncrease miscarriage, stillbirth, or premature delivery. Congenital HIV infectionNo[57,58,59,60,61]
HBVSexual contact, sharing injecting equipment Liver cellsPremature delivery or low birth weight, chronic HBVYes[62,63,64,65]
HSVSexual contact, infected skin or mucous membranesSkin and mucous membranes, nerve cellsCongenital HSV infection, lead to neurological damage, blindness, and deathNo[66,67,68,69]
IAVRespiratory tract, through respiratory dropletsPrimarily infects respiratory tract cellsIncreased risk of pneumonia,
premature delivery, or stillbirth.
Yes[6,45,70,71,72,73]
ZIKVAedes mosquito bite, sexual contact, blood transfusionInfects skin, lymph nodes, and other tissues including placentaFetal loss, stillbirth, miscarriage, CZS with brain abnormalitiesNo[74,75,76,77,78]
SARS-CoV-2Respiratory dropletsPrimarily infects cells in the respiratory tractPreterm delivery, fetal distress, and stillbirthYes[46,79,80,81,82,83,84,85]
Table 2. Primary routes of vertical transmission of the eight viruses.
Table 2. Primary routes of vertical transmission of the eight viruses.
VirusMain Routes of Vertical TransmissionRepresentative
References
RuVThe virus can cross the placenta. Transplacental infection can occur at any stage of pregnancy, highest incidence during the first trimester (organogenesis period)[9,92,93]
HCMVPlacental and perinatal transmissions, especially if the mother has a primary infection during pregnancy or at the time of delivery; through breastfeeding[55,93,94,95]
HIVThe majority of MTCT of HIV occurs during delivery or through breastfeeding [57,58,96,97,98]
HBVPerinatal transmission during delivery is the primary route [62,99,100,101]
HSVAny stage of pregnancy, highest during delivery when the fetus passes through the infected birth canal[68,102,103,104,105]
IAVThrough respiratory secretions. The risk of vertical transmission is low compared to other viruses[3,45,106,107]
ZIKVCan cross the placenta. Vertically transplacental infection is highest during the first and second trimesters of pregnancy[75,76,90,108]
SARS-CoV-2Risk of vertical transmission is generally low. Higher in certain situations: severe maternal COVID-19, infected close to the time of delivery[79,83,109,110]
Table 3. Summary of findings from up-to-date reported studies on the roles of TGF-β1 in viral infections at non-maternal–fetal interface.
Table 3. Summary of findings from up-to-date reported studies on the roles of TGF-β1 in viral infections at non-maternal–fetal interface.
VirusType of Studies, Involved Organs/Cell TypesEffects/RolesReference
RuVIn vitro, lung epithelial cellsIncrease the virus binding and infection in A549 cells[20]
HCMVIn vitro, renal tubular epithelial cells and umbilical vein endothelial cellsIncreased expression and activation of TGF-β1 by HCMV infection[225,226]
HIVEx vivo, bronchial epithelial cells. In vitro, macrophagesIncrease CXCR4 expression in macrophages, increase the viral burden in bronchial epithelial cells[24,25,232]
HBVIn vitro, hepatocellular carcinoma cellsInhibit the expression of HBsAg and HBeAg, and suppress HBV replication in HepG2 cells[28,30]
HSVEx vivo, human cornea organotypic cultureEnhance HSV-1 replication in 3-dimensional human corneal keratocytes [247]
IAVIn vitro, lung epithelial cellsInhibit apoptosis induced by IAV infection on A549 cells[251]
ZIKVIn vitro, Sertoli cellsNot affect ZIKV replication in human Sertoli cells[253]
SARS-CoV-2In vitro, airway epithelial cellsIncrease furin expression leading to enhanced susceptibility to SARS-CoV-2[31,256]
Table 4. Summary of findings from up-to-date reported studies on the roles of TGF-β1 in viral infections during pregnancy. Abbreviation: NR, not reported.
Table 4. Summary of findings from up-to-date reported studies on the roles of TGF-β1 in viral infections during pregnancy. Abbreviation: NR, not reported.
VirusType of Studies, Organs/Cells Involved, Pregnancy PeriodEffects/RolesReference
RuVNRNR
HCMVIn vitro, STBTGF-β1 and IL-8 promote HCMV replication in STB[26]
HIVIn vitro, STBNot increase HIV construct replication[257]
HBV
In vitro, first-trimester trophoblast HTR-8/SVneo cells Increases HBx-transfected HTR-8/SVneo cell proliferation and invasion[258]
HSVNRNR
IAVNRNR
ZIKVIn vitro, first-trimester tropho-blast cellsIncrease the virus binding and replication in trophoblasts[34]
SARS-CoV-2NRNR
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Trinh, Q.D.; Pham, N.T.K.; Takada, K.; Ushijima, H.; Komine-Aizawa, S.; Hayakawa, S. Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives. Int. J. Mol. Sci. 2023, 24, 6489. https://doi.org/10.3390/ijms24076489

AMA Style

Trinh QD, Pham NTK, Takada K, Ushijima H, Komine-Aizawa S, Hayakawa S. Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives. International Journal of Molecular Sciences. 2023; 24(7):6489. https://doi.org/10.3390/ijms24076489

Chicago/Turabian Style

Trinh, Quang Duy, Ngan Thi Kim Pham, Kazuhide Takada, Hiroshi Ushijima, Shihoko Komine-Aizawa, and Satoshi Hayakawa. 2023. "Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives" International Journal of Molecular Sciences 24, no. 7: 6489. https://doi.org/10.3390/ijms24076489

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

Article Metrics

Back to TopTop