Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2020 Dec 22;95(2):e01381-19. doi: 10.1128/JVI.01381-19

Role of Inflammation in Virus Pathogenesis during Pregnancy

Anna Chudnovets a, Jin Liu a, Harish Narasimhan b, Yang Liu a, Irina Burd a,
Editor: Sabra Kleinc
PMCID: PMC7944452  PMID: 33115865

Viral infections during pregnancy lead to a spectrum of maternal and fetal outcomes, ranging from asymptomatic disease to more critical conditions presenting with severe maternal morbidity, stillbirth, preterm birth, intrauterine growth restriction, and fetal congenital anomalies, either apparent at birth or later in life. In this article, we review the pathogenesis of several viral infections that are particularly relevant in the context of pregnancy and intrauterine inflammation. Understanding the diverse mechanisms employed by viral pathogens as well as the repertoire of immune responses induced in the mother may help to establish novel therapeutic options to attenuate changes in the maternal-fetal interface and prevent adverse pregnancy outcomes.

KEYWORDS: inflammation, pregnancy, viral infection

ABSTRACT

Viral infections during pregnancy lead to a spectrum of maternal and fetal outcomes, ranging from asymptomatic disease to more critical conditions presenting with severe maternal morbidity, stillbirth, preterm birth, intrauterine growth restriction, and fetal congenital anomalies, either apparent at birth or later in life. In this article, we review the pathogenesis of several viral infections that are particularly relevant in the context of pregnancy and intrauterine inflammation. Understanding the diverse mechanisms employed by viral pathogens as well as the repertoire of immune responses induced in the mother may help to establish novel therapeutic options to attenuate changes in the maternal-fetal interface and prevent adverse pregnancy outcomes.

INTRODUCTION

Viral infections during pregnancy are known to increase risks for miscarriages, preterm birth, stillbirth, fetal brain injuries, intrauterine growth restriction, hearing loss, cataracts, and other fetal abnormalities (14). During maternal infection, the placenta serves as an immune organ, protecting the fetus from pathogens. However, viruses have evolved complex mechanisms to induce disease, while heightened maternal immune cytokine activation in response to infection also results in detrimental effects on the fetus.

Specific viruses, among other pathogens, have been established to be associated with adverse pregnancy outcomes and congenital defects, constituting parts of the acronym TORCH (toxoplasma, others, rubella, cytomegalovirus [CMV], and herpes). “Others” includes Zika virus (ZIKV), parvovirus B19, human immunodeficiency virus (HIV), hepatitis viruses, and more (3, 5). More recently, researchers have also proposed including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in this acronym (6). Some congenitally acquired viral infections are more common. For example, CMV, the most common cause of congenital birth defects, reaches 800/1,000,000 live births in the United States (worldwide data are unavailable), while other viruses are less prevalent: hepatitis B reaches <0.1/1,000,000 U.S. population and 240 million worldwide, and HIV reaches 162 infants/year (2010) in the United States and 35.3 million worldwide (3). Among the aforementioned TORCH pathogens, there are vaccine-preventable viruses, which still represent a great burden in low- and middle-income countries, resulting in adverse pregnancy and neonatal sequelae. For example, while cases of congenital rubella syndrome (CRS) were vastly underreported, it was estimated that CRS affected 46,000 infants annually in Southeast Asia alone (1).

Between 2.2 and 8.4% of even asymptomatic pregnant women with normal ultrasound findings have detectable viral genomes in the amniotic fluid. At least one of the eight viruses was found to be adenoviruses, herpes simplex virus (HSV), varicella-zoster virus (VZV), human herpesvirus 6 (HHV6), human cytomegalovirus (HCMV), Epstein-Barr virus (EBV), parvovirus B19, and enteroviruses (1, 7). HHV6 was the most common virus detected in amniotic fluid, at 1%. In this study, viruses were not associated with any changes of the white blood cell counts or cytokine levels, but they can be a predisposing factor to intrauterine inflammation with other pathogens, so further studies are needed (1, 7). Intrauterine inflammation is known to be a predisposing factor to adverse maternal (preterm birth) and fetal (perinatal brain injury in the form of periventricular leukomalacia) outcomes.

Certain infections may disproportionately affect pregnant compared with nonpregnant women (8). Pregnant women are more severely affected by some viral infections, including influenza virus, hepatitis E virus (HEV), and HSV. One of the recent theories proposed that a shift from Th1 to Th2 immunity during pregnancy suppresses the CD4+, CD8+, and natural killer (NK) cell response, decreasing the robustness of cell-mediated immunity against viral infection (810). However, the pathophysiological basis of this increased susceptibility still is not well understood, and the evidence of increased susceptibility of pregnant women to infection is rather weak. These responses may be relevant to a heightened inflammatory mechanism and the Th immune shift.

Our review focuses on the pathogenesis of virus-induced inflammation and its maternal and fetal adverse outcomes. Understanding the mechanisms employed by viral pathogens in the placenta and fetus and studying the gaps in the current research may enable new therapeutic approaches to decrease the adverse pregnancy outcomes and fetal anomalies and improve fetal survival.

METHODOLOGY

We conducted a comprehensive search of peer-reviewed journal articles indexed in the NLM’s MEDLINE and PUBMED databases, available as of 15 May 2020, using the following search terms: “viral infections and pregnancy,” “viral infections and neonatal outcomes,” and “viral infection pathogenesis in pregnancy.” We also searched each of these terms separately for certain viral pathogens of interest for pregnancy. Manual searches of reference lists from relevant papers were performed to identify additional studies. While all studies and review articles were evaluated and reviewed, we mostly focused our review on the clinical outcomes of the viral infections of mother and fetus and the possible immunologic mechanisms of invasion (Table 1).

TABLE 1.

Cytokines and signaling pathways

Virus type Description
DNA virus
    HSV CMV infection triggers innate immune responses resulting in IFN-γ production within the maternal decidua; CMV may stimulate the secretion of various cytokines in the placenta, including IFN-γ, which in turn promotes β2MG production in the fetus
In maternal serum, inflammatory markers CPR and SAA are increased
In placenta, MPP-9 and integrin α1β1 are downregulated
In fetus, the virus exhibits tropism to neural cells and activates an inflammatory response in fetal brain
HCMV interacts with TLR2 on ST-like cells, consequently stimulating the production of TNF-α
β2MG, which is produced by activated lymphocytes and accelerated by inflammatory cytokines, such as IFN-γ, is significantly increased in FGR newborns’ urine with placental CMV detection
Adaptor protein stimulator of interferon genes (STING) is essential for IFN-β induction by HSV-1; increased expression of IFI16 and RIG-I has been observed following HSV-1 infection in human third-trimester placenta, which recognizes dsDNA to induce type I IFNs, the STING/TBK1/IRF3 pathway, and proinflammatory cytokine responses via the NF-κB pathway
Mitochondrial checkpoints have been reported to initiate NF-κB activation in the macrophage after HSV infection
In placenta, there is inhibition of IFN-β expression or enhanced expression of cellular receptors, including both TAM ligand and TAM receptors
IL-12, along with numerous cytokines, induces the production of IFN-γ, mainly in NK cells, to trigger killing of infected cells
HSV inhibits the expression of several proinflammatory cytokines, including IFN-α/β, TNF-α, IL-6, IL-12, and RANTES, by reducing the stability of mRNA in macrophages
    VZV IFN-α, IL-1α, IL-6, CXCL10, and TGF-β are produced by VZV-infected fetal DRG, but exact cellular source of these cytokines is unknown; DRG infection is also associated with increased TGF-β and TGF-β-related cytokines
RNA virus
    Influenza viruses Systematically, there are lower circulating levels of IgG2 following infection of H1N1 during pregnancy
Pregnant women with H1N1 exhibit an attenuated innate interferon response
In pregnant mice with H1N1, there is a prominent association between mortality and induction of proinflammatory cytokines and chemokines (TNF-α, CCL2, CCL3, and CXCL1)
    ZIKV Systemic inflammatory response consists of increased IL-1β, IL2, IL6, IL7, IL15, and IL16
ZIKV infection leads to the inhibition of type I IFN production and downstream ISGs
ZIKV NS1, NS4A, and NS4B proteins function as the primary suppressors of type I IFN induction by blocking TBK1/IRF3 pathways
Damaged fetal brain indicates significantly higher expression of NLRP1, NLRP3, AIM2, IL-1β, IL-6, IL-18, IL-33, and caspase 1 following ZIKV-induced inflammation
    HIV During maternal HIV-1 infection, maternal neutralizing antibodies (NAbs) and virions interact directly with placental macrophages (Hofbauer cells) prior to entering fetal circulation; Hofbauer cells express HIV-1 (co)receptors CD4, CCR5, CXCR4, and DC-SIGN on their surface and sequester Abs and Ab-virion immune complexes, limiting HIV-1 MTCT
Additional host restriction factors expressed by HCs or trophoblasts have been identified, including APOBEC3G and ISGs
Phagocytes (neutrophils, monocytes/macrophages, and DCs) and other innate immune cells (such as plasma cell-like DCs and IFN and NK cells) are initially recruited to eliminate the virus
IP-10 (also called CXCL10, a CXC chemokine) increases, which is associated with low CD4 counts
The major immune cells and associated cellular mediators in HIV-1 infection include (i) dendritic cells (cDC and pDC), macrophages, and NK cells (IFN-γ, IFN-α, IL-6, IL-7, IL-10, IL-12, IL-15, IL-18, IP-10, MCP-1, and TNF-α) and (ii) CD4+ T cells (Th1, Th17, Treg, and Tfh), CD8+ T cells, and B cells (IFN-γ, IL-7, IL-12, IL-15, IP-10, MIP-1β, RANTES, and A-SAA)
Plasma CXCL13 levels increase during early and chronic HIV infection
The level of sTNFR2 and IL-6 change in pregnant women's serum; MiP is associated with increased sTNFR2, sICAM-1, and IL-18BP concentrations during pregnancy
There is a lack of correlation between maternal and neonatal inflammatory markers, suggesting that these biomarkers do not cross the placenta during HIV infection
    HCV Maternal-fetal tolerance mechanisms can induce an inhibitory effect on cellular immunity to suppress HCV-specific CTL activity, preventing hepatocyte destruction
The placenta releases endogenous IFN-α during pregnancy, which can influence ALT levels
Newborns exposed to HCV have activation of inhibitory T cells and proinflammatory markers, which are offset by higher IFN-γ production
Human trophoblast cells infected with HCV-RNA can induce strong upregulation of type I/III IFN and increased expression of chemokines, such as CCL5 (RANTES), CXCL10 (IP-10), and CXCL11 (I-TAC), inducing the recruitment and activation of decidual NK cells; HCV-RNA transfection could induce apoptosis in HTR8 cells, which may affect morphology of placenta
Gradient of the proportion frequency of NKT and γ δ-T cells in placenta is higher than that in umbilical cord blood
Polymorphisms of IL-28B promoter, mannose-binding lectin, TNF-α, IFN-γ, IL-10, and TGF-β have also been used to study the risk of MTCT of HCV
Infected hepatocytes secrete cytokines, such as IL-8, CXCL-9, and CXCL-10, all of which are associated with previous damage, to recruit T cells to the infected liver
Liver TNF-α is associated with increased inflammatory activity, liver fibrosis, and liver injury in patients with chronic hepatitis C
In patients with mixed infection of HCV and HBV, the roles of IL-6 and IL18 are vital, and the effect of interferon on immune status could be evaluated
Serum mediators, such as IL-6, IL-8, CXCL-9, CXCL-10, CXCL-12, and macrophage MIF, are increased
    RV The virus enters the fetus via the maternal bloodstream and infects the epithelium of chorionic villi and the endothelium of the blood vessels of the placenta
RV induces apoptosis in monolayers of human normal-term placental CVE and CTB
Inflammatory chemokines have been observed to be increased in the supernatant of RV-infected endothelial cells
The level of IFNs induced by RV strain in placenta is significantly changed
    SARS-CoV-2 Virus enters alveolar epithelial cells via the membrane-bound ACE2 receptor, which may cause pneumonia, sepsis, or aspiration due to cytokine storms, including cytokines IL-6, IL-2, IL-7, IL-10, TNF-α, G-CSF, MIP1A, MCP1, and IP10
Patients with COVID-19 reveal activation of both Th1 and Th2 immunity, culminating in the presence of IFN-γ and IL-1β as well as IL-4 and IL-10
Elevated levels of IL-6 (a predominantly Th1 response) are associated with a significantly increased risk of mortality
Th1 cell-mediated immunity attenuates due to the physiological transition to a Th2 dominant environment; Th2 dominant environment and hormonal milieu tend to increase the expression of anti-inflammatory cytokines (IL-4 and IL-10) and other unidentified immune adaptations, which may take responsibility for the predominant immune response to SARS-CoV-2, resulting in the lesser severity of COVID-19 compared to that in nonpregnant patients
Changes of the hormonal milieu in pregnancy would influence immunological responses to viral pathogens

DNA VIRUSES AND PREGNANCY

CMV, or herpesvirus type 5, is a member of the Betaherpesvirinae subfamily and persists lifelong following primary infection (11). The specific immunologic mechanisms controlling lifelong latency of the disease and the possibility of reinfection with a new CMV strain are not well understood (12). Human CMV is the most common cause of congenital infection, affecting 0.2 to 2% of all live births (12). Congenital CMV transmission rates are as high as 50% in women who acquire primary CMV infection during pregnancy and less than 2% in women with nonprimary infection (13). The two known main sources of primary CMV infection during pregnancy are through sexual activity and contact with young children, where infection transmission can occur in utero, intrapartum, and during breastfeeding (14). Among the mentioned sources, in utero CMV transmission has the highest risk of congenital infection (15). The severity of congenital CMV infection decreases with advancing gestation; however, the risk of transmission to the fetus rises from 40% in first and second trimesters to 60% in the third trimester (16). Interestingly, even the occurrence of primary CMV infection in the preconception and periconception periods have been associated with increased risk of in utero CMV transmission (5% and 16%) (17).

According to several studies, women who did not transmit CMV to the fetus had earlier production of neutralizing antibodies against CMV (gH/gL/pUL128L) (18) in addition to an early CD4+ response (19). Furthermore, it was found that decreased reexpression of CD45RA on the surface of HCMV-specific effector memory cells correlates with HCMV transmission to the fetus (20). In the placenta, CMV replicates in differentiating cytotrophoblasts (CTB), where it induces downregulation of matrix metalloproteinase-9 (MPP-9) and integrin α1β1, impairing cell invasiveness (21). The resultant impairment of cytotrophoblast integrity facilitates the spread of the virus to the fetal compartment. Within fetal tissues, the virus exhibits tropism to neural cells. However, the neuropathogenesis of CMV-induced brain disorders has not yet been fully elucidated. It was observed that HCMV-infected cells localize predominately in the ventricular and subventricular zones (SVZ) (22), where the virus disrupts the functions of neural stem cells (23).

The primary infection during pregnancy is asymptomatic in 75 to 95%; alternatively, it can manifest as a mild mononucleosis or flu‐like syndrome with persistent fever, fatigue, and a variety of eruptions (in one-third of patients), with rare incidence of CMV-induced organ-specific complications (24). There is no evidence that pregnancy can increase maternal CMV disease severity, but cervical shedding may be more common among pregnant women: 5% in the first trimester, 6% to 10% in the second trimester, and 11% to 28% in the third trimester (25). Furthermore, 30% of seropositive women intermittently secrete CMV in the breastmilk.

Most infants with congenital CMV are asymptomatic at birth. Congenital CMV is the leading cause of nonhereditary sensorineural hearing loss, growth failure, and vision loss (26). The clinical findings of symptomatic congenital CMV infection include jaundice, petechiae, thrombocytopenia, hepatosplenomegaly, growth restriction, myocarditis, and nonimmune hydrops (15). Both symptomatic and asymptomatic children excrete the virus for several years after birth, whereas in adults, viral excretion is usually controlled within several months. This reduced control of CMV replication suggests a limitation in cell-mediated immune responses of infants. Around 10% to 15% of asymptomatic infants will develop long‐term neurological sequelae, primarily hearing loss (12, 26).

One study (27) indicates that chronic villitis was commonly observed in the placentas with CMV infection. Studies demonstrate CMV antigens induce placental inflammation in a Toll-like receptor-2 (TLR2)-dependent manner as an innate defense response instead of a direct infection (28, 29). Specifically, HCMV interacts with TLR2 on syncytiotrophoblast (ST)-like cells, consequently stimulating the production of tumor necrosis factor alpha (TNF-α) and inducing the apoptosis of adjoining cells.

In addition, placental inflammation caused by CMV-induced chronic villitis results in the elevation of inflammatory markers in the serum of pregnant women. One cohort study (27) demonstrated that increased C-reactive protein (CRP) or serum amyloid A (SAA) levels in the serum was frequently discovered in fetal growth restriction (FGR) cases with placental CMV detection, indicating that CRP and SAA can serve as biomarkers to predict CMV-induced placental inflammation during pregnancy. Intrauterine growth restriction in light of placental CMV infection may be attributed to the intrauterine inflammation without vertical transmission of the virus.

Furthermore, beta-2 microglobulin (β2MG), which is produced by activated lymphocytes and accelerated by inflammatory cytokines, such as gamma interferon (IFN-γ), was significantly higher in FGR newborns’ urine with placental CMV detection (27). Previous reports (27, 30, 31) suggest that β2MG is an efficient diagnostic marker for symptomatic CMV congenital infections. CMV infection could trigger innate immune responses, resulting in IFN-γ production within the maternal decidua (32). Thus, CMV may stimulate the secretion of various cytokines in the placenta, including IFN-γ, which in turn promotes β2MG production in the fetus.

All in all, the inflammatory biomarkers in the serum of pregnant women may be useful in establishing a diagnosis of placental chronic villitis induced by CMV. Maternal CMV serologic tests may also be important to comprehensively predict the possibility of congenital CMV infection.

Antiviral treatment prenatally and postnatally was not proven to be effective at preventing congenital or postnatal CMV infection. There are several studies that found antiviral treatment with ganciclovir to be effective in the treatment of symptomatic congenital CMV infection, indicating improved neurologic outcomes compared to those of nontreated groups (33).

HSV.

Herpes simplex virus (HSV) belongs to the Herpesviridae family. HSV infection is prevalent worldwide among women of childbearing age. There are two groups of diseases caused by HSV. The orolabial herpesvirus infection, mostly caused by HSV-1, usually occurs in young children and results in asymptomatic infection. Genital HSV infection, one of the most common sexually transmitted diseases, can be caused by HSV-2 or HSV-1. The United States National Health and Nutrition Examination Survey (NHANES) estimated that the seroprevalence of HSV-1 and HSV-2 in pregnant women from 2007 to 2014 is 59.3 and 21.1% (34). Latency and neurovirulence are the most important biologic properties of HSV infection for humans, where the virus will persist lifelong in the sensory ganglia following primary infection.

HSV infections have similar clinical presentation in pregnant and nonpregnant women, usually asymptomatic or associated with mild symptoms. The orofacial HSV is associated with single or clustered vesicles on the face and lips, while genital HSV vesicles are located on the genitalia, perineum, buttocks, upper thighs, or perianal areas. HSV rash can ulcerate before resolving. The symptoms of primary HSV infection may include malaise, fever, or localized adenopathy. However, transmission of genital herpes to the fetus results in neonatal infection, causing serious morbidity and mortality (35).

HSV is transmitted mainly through direct contact with infected individuals but also can be transmitted to infants from infected pregnant mothers (35). Neonates can be infected in utero, peripartum, and postpartum. Peripartum transmission is most common (∼85%), while in utero has the lowest probability of transmission (5%) (32). Intrauterine HSV infection can be acquired from maternal viremia and is associated with primary HSV infection or ascending infection during pregnancy.

There are three clinical groups of genital HSV infections: primary, nonprimary first episode, and recurrent. Primary maternal genital HSV infection poses the highest risk of neonatal transmission at delivery, while the recurrent HSV infection is the most common form. Primary genital HSV-2 infection is more common; furthermore, it has a higher rate of reactivation. Almost 100% of women with HSV-2 experience clinical or subclinical recurrences throughout their lives. Perinatal transmission is almost the exclusive cause of neonatal HSV-2, occurring as mucocutaneous lesions and/or central nervous system (CNS) disease (3638). Neonatal HSV can be divided into three main categories: local skin, eyes, and oral cavity (SEM); with or without SEM CNS; and transmitted diseases.

During pregnancy, HSV infection is associated with spontaneous abortion, intrauterine growth restriction, preterm delivery, skin scars, chorioretinitis, and microphthalmia (30). The infection that occurs early during pregnancy poses a higher risk, but it may also have severe fetal consequences in late gestation (31).

The intrauterine infection caused by primary HSV infection with viremia in pregnant women is associated with placental infarction, necrotizing calcifying mycosis (inflammation of the umbilical cord), plasma cell infiltration (39), fetal hydrops, and even death in utero. The neonate may have the characteristic triad of skin vesicles, ulcers, and scars, eye injuries, and severe abnormalities of the CNS, including microcephaly or edema.

Ascending infection usually occurs after prolonged rupture of fetal membranes in mothers with HSV infection near the active stage during delivery. Sometimes it occurs in the absence of a ruptured amniotic membrane (40). The cellular targets in herpesvirus are epithelial cells of skin and mucosa and neurons. In terms of pathogenesis, HSV infection can lead to loss of HLA-G, since the virus can block HLA-G intracellular transport in placenta (41). HSV can infect human trophoblast cells, leading to cell death or reduced secretion of human chorionic gonadotropin (42).

Aldo et al. (2) proposed that HSV‐2 infection could lead to an increased sensitivity of trophoblast cells to ZIKV infection via inhibition of IFN-β expression or enhanced expression of cellular receptors required for ZIKA entry, including both TAM (members of the phosphatidylserine receptor family) ligand (Gas6) and TAM receptors (Axl, Mer, and Tyro), which facilitate ZIKV entry into trophoblast cells. Several reports have shown that HSV‐1 DNA and double‐stranded RNA (dsRNA), a side product of viral replication, are detected by various kinds of pattern recognition receptors (PRRs), which play a significant role in the maternal-fetal interface (211). In addition, it was demonstrated that the adaptor protein stimulator of interferon genes (STING) is essential for IFN-β induction by HSV‐1 (2, 12). Increased expression of IFI16 and RIG-I was observed following HSV-1 infection, and double-stranded DNA (dsDNA) to induce type I IFNs via the STING/TBK1/IRF3 pathway and proinflammatory cytokine responses via the nuclear factor kappa B (NF‐κB) pathway were recognized (43). Mitochondrial checkpoints have been reported to initiate NF-κB activation after HSV infection (44). These studies indicate the ability of cellular pathogen sensors to induce downstream signaling pathways in response to viral infection and to prevent immunological rejection of the fetal allograft in order to decrease the incidence of stillbirth and preterm birth. However, HSV is capable of inhibiting the expression of several proinflammatory cytokines, including IFN-α/β, TNF-α, IL-6, IL-12, and RANTES, by reducing the stability of mRNA. Thus, HSV can hinder the host’s antiviral response to infection (45). IL-12, along with numerous cytokines, induces the production of IFN-γ, mainly in NK cells, to trigger killing of infected cells (46).

Chickenpox virus/VZV.

Varicella (chickenpox) infection is caused by a highly infectious DNA virus known as varicella-zoster virus (VZV). While children exhibit only mild symptoms following infection, adults are at risk of significant complications, such as secondary bacterial infections, lesions, encephalitis, and hepatitis (47). Following primary infection, VZV is capable of remaining dormant within the sensory nerve ganglia and causes latent infection in 10 to 20% of cases via reactivation, resulting in shingles or herpes zoster (47).

In the context of pregnancy, the incidence of varicella pneumonia is similar to that of nonpregnant individuals; however, it results in significantly higher morbidity and mortality (up to 20%) in pregnant women (47, 48). Increased severity has been attributed to changes in the immunological profile during pregnancy or the mechanical effect of an enlarged uterus in restricting the movement of the diaphragm, although further studies are required (48). The ability of VZV to traverse the placenta following infection (∼24%) of pregnant women and to infect fetal neurons and neuronal precursors has been shown to result in congenital defects, known as congenital varicella syndrome (CVS) and neonatal chickenpox (49). CVS is typically characterized by low birth weight and intrauterine growth restriction with occasional incidence of fetal mortality, neurological defects, skin lesions, limb abnormalities, ocular disease, and muscular hypoplasia (48, 50). A prominent hypothesis suggests CVS is caused by the reactivation of VZV rather than primary infection, which is supported by the dermatological distribution of skin lesions and segmental maldevelopment (51). Furthermore, the immature fetal cell-mediated immunity would also shorten the latent period between primary infection and reinfection (52). Several studies have demonstrated the highest risk (up to 25%) of intrauterine infection following maternal varicella infection in the first and second trimester and CVS in around 12% of infected fetuses (48). Although there exists a theoretical risk of CVS during maternal herpes zoster infection, no cases have been reported.

The VSV viral envelope glycoprotein gE is essential for infection. This protein binds to the insulin-degrading enzyme (IDE) receptor and employs heparan sulfate to facilitate host cell infection. Various cytokines and chemokines are produced in VZV-infected fetal dorsal root ganglia (DRG), including IFN-α, IL-1α, IL-6, CXCL10, and transforming growth factor beta (TGF-β) (53). While inflammatory cytokines were increased, DRG infection was also associated with increased TGF-β and TGF-β-related cytokines (54). The exact cellular source of these cytokines is unknown, but satellite glial cells (SGC) have been hypothesized to be candidates (47). SGC are a specialized ganglionic cell type that share phenotypic and functional features with antigen-presenting cells. Cytokine changes in response to CVS in the placenta during pregnancy are unknown.

The administration of the varicella vaccines effectively reduces risk of infection, but suboptimal vaccination coverage has resulted in the susceptibility of relatively high numbers of high-risk populations, including pregnant women (48, 49). As a live vaccine, it is contraindicated in pregnant women, making antiviral therapy and varicella-zoster immunoglobulin (VZIG) the only available modes of prophylaxis/treatment for severe disease during pregnancy (55). While effective in nonpregnant individuals, controlled studies are still required to demonstrate efficacy in the context of pregnancy.

RNA VIRUSES AND PREGNANCY

Influenza viruses.

Influenza viruses are a major public health burden, having caused four pandemics since 1918 in addition to annual epidemics, resulting in a substantial number of illnesses and deaths (56). They are characterized by segmented, negative-strand RNA genomes whose error-prone RNA-dependent RNA polymerases enable antigenic drift and antigenic shift (57). As a result, we are required to update the seasonal vaccine annually based on surveillance across the world to match the ever-changing antigenic domain(s) of the virus.

In addition to young children and the elderly, pregnant women are at high risk of developing severe or possibly fatal influenza infections during seasonal epidemics as well as pandemics (58, 59). Data from the 1918 H1N1 and the 1957 H2N2 pandemics have revealed increased risk of complications following influenza A virus (IAV) infection (58). More recently, during the 2009 H1N1 pandemic (H1N1pdm09), approximately 5% of total fatalities were pregnant women, while they only constituted 1% of the total population (60). In addition to adverse effects on maternal health, influenza infection may also result in adverse fetal outcomes, including fetal demise, reduced weight, and preterm delivery (58, 61). However, in most cases, vertical transmission of the virus was not observed, suggesting indirect effects of infection on fetal outcomes (62, 63).

Interestingly, women specifically in their third trimester of pregnancy were at the greatest risk of severe influenza (58, 64, 65). Over 50% of deaths due to secondary pneumonia during the 1918 H1N1 pandemic were of women in their third trimester of pregnancy (58). A similar trend was observed in the example of seasonal influenza cases between 1974 and 1993, where women in the third trimester of pregnancy were more than 3 times more likely to die due to influenza-related illness than nonpregnant women (64). While anatomical changes during pregnancy may contribute to increased severity, various physiological changes leading to altered cardiopulmonary and immunologic function are considered more significant contributors to severe outcomes. Remarkably, pregnant women without pregnancy-associated cardiopulmonary comorbidities were also shown to be at a higher risk of influenza-related hospitalization and death following infection, suggesting the altered immunological profile itself is sufficient to cause severe disease (64). Additionally, sex hormones estradiol and progesterone are known to significantly affect immune responses to influenza (66, 67). Thus, it is imperative to understand the impact of pregnancy-associated changes on immune function in the context of influenza.

The role of altered cytokine and chemokine production, immune cell infiltration, and resultant tissue damage has been extensively studied in the context of influenza pathology (6871). Due to several physiological changes, including elevated levels of estradiol and progesterone, cytokine concentration, as well as immune cell population, size and function are dramatically altered during pregnancy (7274). Increased numbers of peripheral neutrophils, monocytes, and dendritic cells (DCs) are observed (74). However, NK cells and CD4 T cells produce smaller amounts of inflammatory cytokines and chemokines following in vitro stimulation (74). While these studies are helpful for our understanding of immunity during pregnancy, responses to influenza infection during pregnancy are still not very well characterized. Compared to postpartum and nonpregnant women, pregnant women exhibited lower circulating levels of IgG2 (antibody isotype correlated with protection) following infection during the 2009 H1N1 pandemic (75). Additionally, they also exhibited an attenuated innate interferon response, which may permit the presence of higher viral titers within the lung (76). Vaccination studies indicate that the adaptive immune responses in pregnant women are comparable to those of nonpregnant controls, evidenced by similar rates of seroconversion and seroprotection (7779). However, one group demonstrated diminished induction of IgG1, IgG2, and IgG3 responses following vaccination with a pronounced effect during later stages of pregnancy (80).

These studies suggest that there is a paucity of data, specifically regarding the molecular mechanisms responsible for the altered immune responses to influenza infection observed during pregnancy. To address this, several animal studies have been conducted to provide insight into the mechanism of disease.

Pregnant mice infected with H1N1pdm09 experienced significantly higher mortality than nonpregnant females (75, 81). While some groups observed increased viral replication in the lungs of pregnant females, others suggest replication is unaffected by pregnancy (75, 81). Alternatively, a prominent association was observed between mortality and induction of proinflammatory cytokines and chemokines (TNF-α, CCL2, CCL3, and CXCL1) (75, 81). Increases in populations of pulmonary macrophages as well as Treg cells were observed, while CD8 T cells and neutralizing antibody titers were unaffected following infection (81). Pregnant mice have been shown to induce a robust antibody response following H1N1pdm09 infection (75); however, another study suggested a diminished response compared to that of nonpregnant mice (81). Thus, it appears that although profound changes were observed in the innate response to IAV infection, adaptive responses are much less affected.

The role of pregnancy-associated hormonal changes in mediating protection or increased severity of infection has also been explored (66, 67, 82). Elevation of estradiol (E2) has been shown to be protective in the context of both inactivated influenza vaccination as well as IAV infection in nonpregnant females (83). Another estrogen, estriol (E3), specifically induced in high quantities during pregnancy, has also been shown to confer protection following infection by reducing tissue inflammation (82). Similarly, progesterone treatment in nonpregnant females promoted accelerated recovery in influenza-infected females, possibly via the upregulation of the epidermal growth factor amphiregulin in lungs (84). Furthermore, reduced concentrations of progesterone in pregnant females correlated with increased severity and adverse fetal outcomes (85). Thus, it is unlikely that increased levels of E2, E3, and progesterone during pregnancy are responsible for severe disease. However, the impact of other estrogens (E1 and E4) and glucocorticoids, which are also altered during pregnancy, is unknown and requires further investigation. Limited studies have been conducted in other models, such as guinea pigs, ferrets, and pigs, with no major differences in viral replication or morbidity between pregnant and nonpregnant females (50, 8688).

It is evident across clinical and mouse data that pandemic influenza strains are more pathogenic in pregnant women than seasonal strains, reiterating the role of viral factors, such as antigenic shift, in the severity of disease (5961, 89). However, there is also strong evidence indicating that the immunological shift, among other physiological changes observed during pregnancy, contributes to increased severity of influenza infection (58, 59, 64). Significant human and mouse literature suggests that this phenotype is a result of increased tissue damage within the respiratory tract due to the influx of inflammatory cells and the induction of proinflammatory cytokines and chemokines. However, there are major inconsistencies across studies with regard to the dynamics of specific cell populations, associated cytokines, and functions due to differences in strain of virus, dose of inoculum, mouse strain, and timing of infection. Some studies described an increase in cytokine induction (75, 81), whereas others reported an opposite phenotype following infection (90, 91). Several groups have demonstrated higher viral replication in pregnant mice than nonpregnant females (75, 81, 90, 92, 93), while one study reported no differences in viral replication (81).

Therefore, it is important to further systematically study the pathogenesis of IAV and associated immune responses in pregnant women as well as in animal models to improve our understanding and, thus, outcomes following infection. Studies accounting for host factors like pregnancy would also greatly help to develop more effective influenza vaccines for such high-risk populations.

ZIKV.

Zika virus (ZIKV) is a single-stranded RNA virus in the family Flaviviridae. The transmission of ZIKV infection mostly commonly occurs through the bite of the Aedes mosquito. Furthermore, there are additional routes of ZIKV transmission: sexual, blood transfusion, and vertical transmission from mother to child (9496). Clinical observation of birth defects during the recent epidemic emphasized ZIKV as an important threat to public health in 2015. The number of ZIKV disease cases reached 216,207 in Brazil, and 8,604 neonates were born with malformations (9698).

Accumulating evidence suggests that primary human placental trophoblast cell lines, endothelial and Hoffbauer cells, are permissive to ZIKV infection and facilitate viral replication (99, 100). These observations highlight the potential role of the placenta as a reservoir and entry for the virus to reach the fetus, where ZIKV mostly infects neocortical neuroepithelial and neuronal stem cells (NSCs). In the NSCs, ZIKV induces apoptosis, reduced proliferation, and transcriptional alterations (101103), which result in cortical alterations and neurodevelopmental anomalies (104).

The majority (50% to 80%) of ZIKV infections are asymptomatic. The most common signs and symptoms of ZIKV infection occur within a period of 3 to 14 days and last for up to 1 week and manifest as a rash, low-grade fever (72%), arthralgia and myalgia (65%), conjunctivitis (63%), headache (46%), fatigue, and/or rash (105107). Complications are infrequent (<1%), but when they occur, they are severe and may be fatal: Guillain-Barre syndrome, transverse myelitis, iridocyclitis, maculopathy, thrombocytopenic purpura, and myocarditis (106). The manifestations of acute ZIKV infection are similar across age groups, in both sexes, and in pregnant women. ZIKV-infected mothers transmit ZIKV in 20 to 30% of cases to the fetus. Most fetuses and newborns are asymptomatic, but 4 to 7% of pregnancies exhibit miscarriage and 5 to 14% have congenital Zika syndrome (CZS; 4 to 6% microcephaly) (106).

CZS shares the symptoms and signs with other TORCH congenital infections, such as cognitive, sensory, and motor disabilities. However, there are signs that are unique to CZS: (i) severe microcephaly with partially collapsed skull; (ii) thin cerebral cortices with subcortical calcifications; (iii) macular scarring and focal pigmentary retinal mottling; (iv) congenital contractures; and (v) marked early hypertonia and symptoms of extrapyramidal involvement (94, 108). Furthermore, CZS can cause malformations of the important brain structures, such as the hippocampus, corpus callosum, basal ganglia, thalamus, cerebellum, and brainstem (109112). The subcortical calcifications (as opposed to periventricular in CMV) are observed when the infection occurs later in pregnancy and the brain is macroscopically well formed. CZS is also associated with intrauterine growth restriction, craniofacial malformations, and pulmonary hypoplasia (94).

The long-term development of infants with CZS most likely will be severely impacted. Sensorineural hearing impairment, hypertonia, spasticity dysphagia, and tremors are residual long-term manifestations of CZS (113, 114).

According to widely studied animal models of CZS, during Zika infection, the mother exhibits a systemic inflammatory response with increased IL-1β IL-2, IL-6, IL-7, IL-15, and IL-16. Furthermore, ZIKV infection leads to the inhibition of type I IFN production and downstream interferon-stimulated genes (ISGs) (115). ZIKV NS1, NS4A, and NS4B proteins function as the primary suppressors of type I IFN induction by blocking TBK1/IRF3 pathways. Multiple studies of the damaged fetal brain indicate significantly higher expression of NLRP1, NLRP3, and AIM2 and cytokines IL-1β, IL-6, IL-18, IL-33, and caspase 1 following ZIKV-induced inflammation (100, 115118).

HIV.

HIV-1 is a lentivirus that belongs to the family Retroviridae. Perinatally acquired HIV-1 infection is less common in the United States as a result of earlier identification of maternal HIV infection and comprehensive HIV antiretroviral treatment (3), reducing the perinatal transmission rate in the preantiretroviral era from 18% to 32% to 1% to 2% (3, 119). However, in resource-limited settings, such as sub-Saharan Africa, ∼15 to 30% of infants become infected with HIV during pregnancy, with an additional 5 to 20% infected through breastfeeding (200,000 infants were infected in 2013) (1, 120).

The factors that increase the risk of perinatal HIV transmission are the maternal plasma viral load, maternal CD4 count, advanced WHO clinical disease stage, breastfeeding and mastitis, and acute maternal infection (3, 121, 122).

During maternal HIV-1 infection, maternal neutralizing antibodies (NAbs) and virions interact directly with placental macrophages (Hofbauer cells) prior to entering fetal circulation (123, 124). According to several studies, Hofbauer cells express HIV-1 (co)receptors CD4, CCR5, CXCR4, and DC-SIGN on their surface and sequester Abs and Ab-virion immune complexes, limiting HIV-1 mother-to-child transmission (MTCT) (123). Additional host restriction factors expressed by HCs or trophoblasts have been identified, including APOBEC3G and ISGs (124). Despite multiple studies, the strategy of HIV-1 evasion of the placenta and its transmission are poorly understood.

Several studies have confirmed the relationship of HIV seropositivity with increased risk of spontaneous abortion, stillbirth, intrauterine growth retardation, low birth weight, preterm birth (125129), and neurodevelopmental delay (130). HIV is also associated with an increased risk of chorioamnionitis and deciduitis, which may in part explain the elevated risk for preterm birth (131, 132).

It is important to mention that postnatal growth of the children of HIV-infected women is often delayed even without the occurrence of vertical transmission. Furthermore, HIV-exposed uninfected infants (HEU) suffer from up to 4-fold higher rates of morbidity and mortality from diarrheal and respiratory illnesses than unexposed children (133136). The pathophysiology of such increased risk of illness and death in HEU infants is multifactorial and can be related to the severity of maternal HIV disease (137), poor placental transfer of protective maternal antibodies (138, 139), replacement feeding rather than breast feeding (140), perinatal exposure to antiretroviral drugs (141143), and increased exposure to pathogens from immunodeficient individuals in the household (144, 145).

Acute HIV infection results in the production of proinflammatory cytokines and chemokines, which help to recruit inflammatory cells to the site of infection. Phagocytes (neutrophils, monocytes/macrophages, and DCs) and other innate immune cells (such as plasma cell-like DCs that produce IFN and NK cells) are initially recruited to eliminate the infecting virus (146, 147). Early studies (147, 148) proved that neopterin and β2-microglobulin were increased in HIV infection. Elevated levels of IP-10 (also called CXCL10, a CXC chemokine) were associated with low CD4 counts, strongly indicating this marker as a predictor of HIV infection progression.

Following infection, DCs present antigen to T cells and B cells within lymphoid tissues to trigger the adaptive immune response. The major immune cells and associated cellular mediators in HIV-1 infection include (i) dendritic cells (cDC and pDC), macrophages, and NK cells (IFN-γ, IFN-α, IL-6, IL-7, IL-10, IL-12, IL-15, IL-18, IP-10, MCP-1, and TNF-α) and (ii) CD4+ T cells (Th1, Th17, Treg, and Tfh), CD8+ T cells, and B cells (IFN-γ, IL-7, IL-12, IL-15, IP-10, MIP-1β, RANTES, and A-SAA) (147).

Significant elevation of plasma CXCL13 levels during early and chronic HIV infection represents a novel marker of systemic immune activation during early and chronic HIV infection and may be used to predict the development of non-AIDS events.

One study (149) demonstrated that maternal HIV infection induces a state of fetal hyperferritinemia, which is unrelated to maternal ferritin level and does not appear to be secondary to the fetal proinflammatory state. Another study (150) demonstrated that women infected with HIV have an increased risk of malaria in pregnancy (MiP). MiP also was associated with increased sTNFR2, sICAM-1, and IL-18BP concentrations during pregnancy. Pregnant women who delivered preterm had increased concentrations of sTNFR2 and altered levels of IL-6. Interestingly, pregnant women with sTNFR2 concentrations in the highest quartile within 6 weeks of delivery had an elevated relative risk of preterm birth.

One study (151) of Brazilian mother-infant pairs found that levels of inflammation, monocyte activation, and oxidized low-density lipoprotein in HIV-exposed infants are higher than levels in unexposed infants at birth, some of which remain elevated for up to 6 months of life. Interestingly, the levels of these inflammatory markers were not related to maternal inflammatory status. Inflammation may contribute to the increased HIV-exposed, uninfected infectious morbidity and slow growth. In addition, the lack of correlation between maternal and neonatal inflammatory markers suggests that these biomarkers do not cross the placenta during HIV infection (152).

In summary, further research is required to determine the contribution of maternal and fetal immune responses to adverse outcomes among HIV-infected mothers and infants. Furthermore, advances in this area could aid in the development of novel therapeutic strategies targeting pathological inflammation in HIV-infected mothers and infants.

HCV.

The worldwide prevalence of hepatitis C virus (HCV) infection is between 1% and 8% in pregnant women and between 0.05% and 5% in children (153). MTCT is the main cause of HCV infection in children, with an incidence from 3% to 10% (154) and a 2- to 4-fold increase upon coinfection with HIV-1 (155, 156). HCV may directly impact maternal health during pregnancy or influence pregnancy outcomes due to MTCT or maternal infection. Chronic HCV itself does not seem to have a negative effect on pregnancy, while there are few related pathological studies.

Serum alanine aminotransferase (ALT) levels were observed to be elevated in nonpregnant individuals infected with HCV as a result of leakage from the liver due to immune-mediated damage of parenchymal liver cells. However, recent studies found that in pregnant women with hepatitis C, maternal serum ALT levels decrease during the first trimester of pregnancy and rise after delivery, while HCV RNA levels increase during the second and third trimesters (157, 158). This inconsistency between ALT levels of pregnant and nonpregnant individuals might be due to maternal-fetal tolerance mechanisms that induce an inhibitory effect on cellular immunity to suppress HCV-specific cytotoxic T-lymphocyte (CTL) activity, thereby preventing hepatocyte destruction (159). Additionally, the placenta also releases endogenous IFN-α during pregnancy, which can influence ALT levels; however, its effect on viral clearance is unclear (160).

Research found that HCV coinfection was significantly associated with cholestasis (161, 162). HCV infection during pregnancy may increase the risk of adverse perinatal outcomes, such as preterm delivery, FGR, congenital malformations, and overall perinatal mortality (163166). However, there is a lack of consistency between reports of maternal and fetal morbidity and outcome across different studies, primarily due to small sample sizes.

There is about a 5% risk of transmission of the virus from mother to baby. HCV may pass from mother to child during pregnancy and childbirth, while breastfeeding does not appear to contribute to an increase in HCV transmission rate (167, 168). Furthermore, the mode of delivery, vaginal or cesarean section, did not show differences in rates of HCV vertical transmission (169).

HCV may be transmitted to infant blood through maternal blood leakage or through placenta, but the underlying molecular mechanisms are unclear. Studies have indicated the ability of HCV to infect placental cytotrophoblasts in vitro (170). Additionally, peripheral blood mononuclear cell (PBMC) infection was demonstrated to be a critical risk factor for perinatal transmission of HCV. PBMCs can act as a vector of the virus and transfer it to the target cells of a new host or produce foreign bodies containing HCV, which can enter the fetal bloodstream independently and cause vertical transmission (171).

Research found that newborns exposed to HCV have activation of inhibitory T cells and proinflammatory markers, which are offset by higher IFN-γ production (172). This suggests a balance between proinflammatory and anti-inflammatory responses in newborns exposed to HCV, which may be responsible for prevention of HCV infection in utero.

Anti-HCV antibodies (anti-HCV Ab) can be transmitted across the placenta from a pregnant woman to the fetus, and neonatal serum can be detected as anti-HCV positive up to 13 months after birth. The American Academy of Pediatrics and the U.S. Centers for Disease Control and Prevention (CDC) recommend anti-HCV screening for babies born to HCV-positive women after 18 months or HCV RNA screening for 1-month-old babies twice (173). Ribavirin is banned during pregnancy because of its known teratogenicity. In addition, after discontinuation of ribavirin, the risk of teratogenesis persists for up to 6 months and applies to women taking ribavirin and male and female partners taking ribavirin.

It has been found that human trophoblast cells infected with HCV-RNA can induce strong upregulation of type I/III IFN and increased expression of chemokines such as CCL5 (RANTES), CXCL10 (IP-10), and CXCL11 (I-TAC), inducing the recruitment and activation of decidual NK cells (1). At the same time, it was observed that HCV-RNA transfection could induce apoptosis in HTR8 cells, which may affect the morphology of placenta.

The difference between ALT and HCV RNA observed in postpartum HCV RNA+ women may be due to the different proportions of Th1 cytokines produced (2). A study of mothers and infants infected with HCV examined umbilical cord blood, placenta, and decidua by isolating mononuclear cells. This study found that the gradient of the proportion frequency of NKT and γ δ-T cells in placenta was higher than that in umbilical cord blood. HCV infection can enhance the cytotoxicity of NK and NKT cells in placenta (3). The polymorphisms of the IL-28B promoter, mannose-binding lectin, TNF-α, IFN-γ, IL-10, and TGF-β have also been used to study the risk of mother-to-child transmission of HCV (4, 5).

In the case of uncontrolled replication, infected hepatocytes secrete cytokines, such as IL-8, CXCL-9, and CXCL-10, to recruit T cells to the infected liver, all of which are associated with previous damage (174, 175). During HCV infection, TNF-α is considered the key molecule to promote insulin resistance and diabetes. The ratio of TNF-α to interleukin-10 is higher in patients with severe liver disease and liver cancer (176, 177). Liver TNF-α is associated with increased inflammatory activity, liver fibrosis, and liver injury in patients with chronic hepatitis C (178, 179). In patients with mixed infection of HCV and HBV, the concentration of IL-6 was positively correlated with the course of disease and viral load, while the concentration of IL-18 was positively correlated with the levels of ALT and aspartate transaminase, and the effect of interferon on immune status could be evaluated (180).

The researchers found that serum mediators such as IL-6, IL-8, CXCL-9, CXCL-10, CXCL-12, and macrophage migration inhibitory factor (MIF) can be used as potential markers to evaluate the progression of chronic hepatitis to liver cirrhosis (181, 182).

RV.

Rubella virus (RV), also called German measles or 3-day measles, is a member of the family Togaviridae, genus Rubivirus. Humans are the only known reservoir for rubella infection. The proportion of women of childbearing age who are susceptible to rubella varies greatly among nations, where infection rates are negatively correlated with vaccine coverage.

Maternal acquired rubella is usually a mild, self-limited disease. Erythematous maculopapular eruption as well as prodromal symptoms, such as low fever, conjunctivitis, rhinitis, sore throat, and cough, are observed. Polyarthritis and polyarthralgia are potential sequelae. However, in spite of the mother being asymptomatic or having mild symptoms, rubella infection can have devastating effects for the developing fetus during pregnancy.

Clinical outcomes of maternal infection with rubella are dependent on gestational age. Risk to the fetus is highest during the first trimester and reduces with gestational age. Infection during pregnancy can result in stillbirths, spontaneous abortion, or several anomalies associated with congenital rubella syndrome (CRS). This syndrome can cause one or more problems to infants, such as growth delays, cataracts, glaucoma, deafness, congenital heart defects, defects in other organs, and intellectual disabilities (183).

The pathogenesis of congenital rubella is not completely understood. The virus enters the fetus via the maternal bloodstream and infects the epithelium of chorionic villi and the endothelium of the blood vessels of the placenta (184). Pathological evidence suggests that rubella virus spreads through the vascular system of the developing fetus after infecting the placenta and results in defects due to cytopathic effects within fetal organs (185, 186). RV induces apoptosis in monolayers of human normal-term placental chorionic explants (CVE) and cytotrophoblasts (CTB) but does not induce apoptosis in human fetal fibroblast (HEF) culture of primary human embryonic fibroblasts (187, 188). RV replication in the host cells can directly or indirectly affect the expression of genes involved in the development of sensory organs (189). The lifelong effect of RV on developing fetus is cumulative, which is related to the interaction of host with RV protein (189). The replication cycle of RV was longer, and the replication dynamics was slower (190).

Inflammatory chemokines were observed to be increased in the supernatant of RV-infected endothelial cells (191). The level of IFNs induced by the RV strain in placenta was significantly different, but there was no significant difference in lung or leukocyte culture (192). The cellular immunity of 738 pupils vaccinated with RV vaccine has shifted to the proinflammatory cytokine spectrum, in which the levels of IL-6, granulocyte macrophage colony-stimulating factor (GM-CSF), and TNF-α are increased while the detection rate of IL-10 is the lowest (193).

SARS-CoV-2.

In December 2019, a novel type of coronavirus was identified in patients with infectious respiratory disease in Wuhan, China, with the ability of human-to-human transmission. The disease, termed coronavirus disease 2019 (COVID-19), has expanded globally exceptionally swiftly. COVID-19 is caused by the virus severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), a novel enveloped RNA beta-coronavirus. The primary phenotype of COVID-19 is severe acute respiratory distress syndrome (ARDS). With the spread of this novel coronavirus, interests have expanded to the role of the virus in pregnancy, including potential intrauterine transmission from mother to fetus as well as maternal and fetal outcomes in pregnant women infected with SARS-CoV-2.

In a systematic review including 356 cases (33 studies) in pregnant women, the most frequent symptoms were fever (67%), cough (66%), dyspnea (7%), sore throat (7%), fatigue (7%), and myalgia (6%) (194). Rhinorrhea/nasal congestion, anorexia, nausea/vomiting, headache, and possible abnormalities in smell and/or taste have also been reported. Laboratory findings included lymphopenia (14%), mild increase in liver enzymes (5%), and thrombocytopenia (1%). These clinical manifestations are similar to those in nonpregnant individuals reported in a 138-case cohort study (195).

Some data showed that pregnancy and childbirth do not increase the risk of contracting SARS-CoV-2 and do not worsen the clinical course of COVID-19 compared with those of age-matched nonpregnant patients (196199). The majority of pregnant women who develop severe COVID-19 are younger (200), while within the general population, those affected by severe disease are older (201). Pregnant patients with severe disease also may suffer from other comorbidities that may increase or exacerbate the risk of severe disease (such as hypertension, diabetes, severe obesity, severe asthma, serious heart disease, and immunocompromise) (202).

Some studies (203, 204) reported that patients with severe COVID-19 have laboratory evidence of a cytokine storm-related syndrome induced by a strong inflammatory response, which has been proven to cause severe and fatal illnesses. However, whether the normal alterations within the immune system during pregnancy can influence the course of this response remains an open question.

A review analyzing 51 pregnant patients with confirmed COVID-19 reported that the preterm birth rate (delivered before 37 weeks of gestation) is 39%, where 96% were delivered by cesarean (198). A larger systematic review of 252 pregnant COVID-19 patients reported that the preterm birth rate is 15.2% when 69.4% were delivered by cesarean (194). The frequency of spontaneous abortion (0.8%) (194) is not increased, per current data (194, 205). However, there are limited data available for pregnant patients who were infected during the first trimester. Two reports (206, 207) described two critically ill women who had fetal deaths, where one of the two women died while the other was treated with extracorporeal membrane oxygenation.

For neonatal outcomes, among 256 newborns from one review (194), more than 95% of newborns were healthy at birth. Neonatal complications have been linked mainly to preterm birth (194). Specifically, preterm birth (<37 weeks of gestation) occurred in 39 (15.2%) newborns. Birthweight ranged from 1,520 to 4,050 g, among which a low birth weight (<2,500 g) was reported in 20 (7.8%) newborns, while intrauterine fetal distress was observed in 20 (7.8%) newborns. In addition, 3.1% were admitted to the neonatal intensive care unit, 1.2% were put on neonatal mechanical ventilation, 4.7% had respiratory distress syndrome, 1.2% had neonatal pneumonia (3 cases), and 1.2% had disseminated intravascular coagulation. Mortality occurred in 3 cases.

Four (1.6%) newborns, delivered by cesarean section, had a positive reverse transcription PCR test result, while samples from their cord blood, placenta, and amniotic fluid were negative (194). Three (1.2%) newborns had positive IgM, and 6 (2.3%) had a positive IgG test. PCR test for COVID-19 in samples from 30 cord blood, 23 amniotic fluid, and 12 placenta specimens were all negative. All samples of breast milk from 26 women tested negative for COVID-19 (194).

In addition, hyperthermia is the most common clinical manifestation in pregnant women with COVID-19, as mentioned above. Considering this, a theoretical concern should be focused on the fact that high maternal core temperature from a febrile illness such as COVID-19 during organogenesis in the first trimester may be associated with an increased risk of congenital anomalies, especially neural tube defects, or miscarriage. However, the incidence of these outcomes has not been observed. Longitudinal monitoring of the neonates born to infected mothers would provide further insight into subtle consequences of maternal infection during pregnancy.

There is a hypothetical opportunity of vertical transmission, similar to that seen in SARS, as the aminopeptidase-angiotensin-converting enzyme 2 (ACE2) receptor is generally expressed in the placenta (208) and a similar receptor-binding domain between SARS-CoV and SARS-CoV-2 (209). However, maternal viremia rates appear to be low (1% in one study [210]), indicating that placental seeding and vertical transmission are highly unlikely. Besides, no confirmed cases of intrauterine transmission have been reported in reviews including 51 pregnant women with COVID-19 (197, 198, 211). In addition, several suspected cases based on newborn laboratory and/or clinical findings have been reported (212215), but SARS-CoV-2 testing on fetal blood, amniotic fluid, and placenta was either negative or not performed. The majority of these infants were delivered by cesarean and had positive nasopharyngeal cultures for SARS-CoV-2 on day 1 or 2 of life, an increased IgM level, and/or pneumonia. Positive IgM results are not definitive evidence of in utero infection, and, in most of these cases, early infant infection may have been due to close contact with infected parents or caregivers (216).

Based on the current data available, there have been no confirmed cases of vertical transmission among the neonates born to COVID-19-infected mothers reported. However, it is important to note that the majority of the mothers acquired COVID-19 in the third trimester, and there are limited data for infection earlier in pregnancy.

In pregnant and nonpregnant patients, the main phenotype of COVID-19 is ARDS, a potentially lethal syndrome caused by pneumonia, sepsis, or aspiration due to cytokine storms (217). This occurs following viral entry into alveolar epithelial cells via the membrane-bound ACE2 receptor. This results in the activation of the innate immune and adaptive immune systems, resulting in the release of a large number of cytokines, including IL-6, IL-2, IL-7, IL-10, TNF-α, granulocyte colony-stimulating factor (G-CSF), macrophage inflammatory protein 1 alpha (MIP1A), monocyte chemoattractant protein (MCP1), and interferon-γ-inducible protein (IP10), inducing a cytokine storm (209, 217219).

Interestingly, patients with COVID-19 reveal the activation of both Th1 and Th2 immunity, culminating in the presence of IFN-γ and IL-1β as well as IL-4 and IL-10 (203). Furthermore, elevated levels of IL-6 (a predominantly Th1 response) are associated with a significantly increased risk of mortality in COVID-19 patients (220). Based on this observation, tocilizumab, a humanized monoclonal antibody against the interleukin-6 receptor (IL-6R), has been experimentally applied for the treatment of critical COVID-19 pneumonia in China and Italy with promising results.

In pregnancy, there is an attenuation in Th1 cell-mediated immunity due to the physiological transition to a Th2-dominant environment. In addition, changes of the hormonal milieu in pregnancy would influence immunological responses to viral pathogens. Th2-dominant environment and hormonal milieu tend to increase the expression of anti-inflammatory cytokines (IL-4 and IL-10) and other unidentified immune adaptations, which may be responsible for the predominant immune response to SARS-CoV-2, resulting in the lesser severity of COVID-19 than that in nonpregnant patients (221).

One study reported (222) that SARS-CoV-2 might attack hemoglobin, causing critically low oxygen saturation. According to this theory, the fetus may receive less oxygen due to reduced maternal hemoglobin. The placenta has the role of exchanging carbon dioxide and oxygen between mother and fetus. Therefore, COVID-19 attacking hemoglobin would produce iron, carbon dioxide, and oxygen, which might induce toxicity in placental and fetal cells. In addition, capillaries would be easily broken due to inflammation. Proteins such as fibrinogen fill the capillaries' cracks via the coagulation reaction, which will also lead to severe placental inflammation.

CONCLUSIONS AND FUTURE DIRECTIONS

Perinatal outcomes from viral infections during pregnancy can range from mild to asymptomatic disease to pregnancy loss by spontaneous abortion as well as fetal infection, resulting in congenital viral syndromes. Prenatal care has no true standards for antenatal management for most viral infections during pregnancy, with no available standardized treatment or preventative strategies.

Most viral infections during pregnancy interfere with IFN and TNF signaling, but, due to diverse antigenic properties, they exhibit different tissue tropism and outcomes. CMV interferes with TLR-2 responses and is neurotropic, resulting in congenital fetal anomalies. HSV and VZV are also neurotropic, while VZV additionally induces TGF-β, IL-1α, IL-6, and CXCL10 production. Influenza virus causes IL-1α, IL-6, CXCL10, and TGF-β elevation and has a more pronounced effect on the mother than the fetus. In contrast, ZIKV is neurotropic and causes fetal brain damage by inhibiting type I IFN responses and inducing IL-1β, IL-6, IL-18, NLRP1, NLRP3, and AIM2 expression. Rubella, similarly, can cause congenital rubella syndrome in the fetus and has endothelial and neural tropism, inducing IL-6, GM-CSF, and TNF-α. In addition, we reviewed other viruses, such as HIV and HCV, which do not cause severe congenital anomalies but are of interest because of possible transmission to the fetus. Lastly, SARS-CoV2, which is currently highly clinically relevant due to the large number of cases during the pandemic, is tropic to the ACE2 receptor in the lung tissues and causes IL-6 and IFN-γ activation. Importantly, pregnancy and childbirth do not exacerbate the risk of acquiring SARS-CoV-2 or severe disease and do not result in worse pregnancy outcomes.

There is a dearth of information with regard to viral pathogenesis and inflammation during pregnancy. Several studies are required to investigate the diverse mechanisms employed by viruses to induce disease in pregnant women as well as the complex, potentially pathological immune responses induced in response to infection. Further insight into specific molecular mechanisms responsible for the increased severity of disease in mothers and/or neonates due to viral infections and associated inflammation would be greatly beneficial in improving clinical care during pregnancy. Additionally, this knowledge could aid in efforts to develop novel therapeutics targeting uncontrolled inflammatory responses to viral infection during pregnancy to prevent adverse fetal outcomes.

ACKNOWLEDGMENTS

Financial support was received from NIH Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) grants R01HD097608 and R21HD099000.

We have no conflicts of interest to report.

REFERENCES

  • 1.Kristina MAW, Ryan MM. 2013. Influence of infection during pregnancy on fetal development. Reproduction 146:R151–R162. doi: 10.1530/REP-13-0232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Silasi M, Cardenas I, Kwon J-Y, Racicot K, Aldo P, Mor G. 2015. Viral infections during pregnancy. Am J Reprod Immunol 73:199–213. doi: 10.1111/aji.12355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Neu N, Duchon J, Zachariah P. 2015. TORCH infections. Clin Perinatol 42:77–103. doi: 10.1016/j.clp.2014.11.001. [DOI] [PubMed] [Google Scholar]
  • 4.Leeper C, Lutzkanin A. 2018. Infections during pregnancy. Prim Care 45:567–586. doi: 10.1016/j.pop.2018.05.013. [DOI] [PubMed] [Google Scholar]
  • 5.Rasti S, Ghasemi FS, Abdoli A, Piroozmand A, Mousavi SGA, Fakhrie-Kashan Z. 2016. ToRCH “co-infections” are associated with increased risk of abortion in pregnant women. Congenit Anom 56:73–78. doi: 10.1111/cga.12138. [DOI] [PubMed] [Google Scholar]
  • 6.Muldoon KM, Fowler KB, Pesch MH, Schleiss MR. 2020. SARS-CoV-2: is it the newest spark in the TORCH? J Clin Virol 127:104372. doi: 10.1016/j.jcv.2020.104372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gervasi M-T, Romero R, Bracalente G, Chaiworapongsa T, Erez O, Dong Z, Hassan SS, Yeo L, Yoon BH, Mor G, Barzon L, Franchin E, Militello V, Palù G. 2012. Viral invasion of the amniotic cavity (VIAC) in the midtrimester of pregnancy. J Matern Fetal Neonatal Med 25:2002–2013. doi: 10.3109/14767058.2012.683899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kourtis AP, Read JS, Jamieson DJ. 2014. Pregnancy and infection. N Engl J Med 370:2211–2218. doi: 10.1056/NEJMra1213566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Straub RH. 2007. The complex role of estrogens in inflammation. Endocr Rev 28:521–574. doi: 10.1210/er.2007-0001. [DOI] [PubMed] [Google Scholar]
  • 10.Anonymous. . 2012. Pregnancy and pregnancy-associated hormones alter immune responses and disease pathogenesis. Horm Behav 62:263–271. doi: 10.1016/j.yhbeh.2012.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Alford CA, Stagno S, Pass RF, Britt WJ. 1990. Congenital and perinatal cytomegalovirus infections. Rev Infect Dis 12:S745–S753. doi: 10.1093/clinids/12.Supplement_7.S745. [DOI] [PubMed] [Google Scholar]
  • 12.Davis NL, King CC, Kourtis AP. 2017. Cytomegalovirus infection in pregnancy. Birth Defects Res 109:336–346. doi: 10.1002/bdra.23601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kenneson A, Cannon MJ. 2007. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol 17:253–276. doi: 10.1002/rmv.535. [DOI] [PubMed] [Google Scholar]
  • 14.Fowler KB, Pass RF. 2006. Risk factors for congenital cytomegalovirus infection in the offspring of young women: exposure to young children and recent onset of sexual activity. Pediatrics 118:e286–e292. doi: 10.1542/peds.2005-1142. [DOI] [PubMed] [Google Scholar]
  • 15.Anonymous. . 2015. Practice bulletin no. 151: cytomegalovirus, parvovirus B19, varicella zoster, and toxoplasmosis in pregnancy. Obstet Gynecol 125:1510–1525. doi: 10.1097/01.AOG.0000466430.19823.53. [DOI] [PubMed] [Google Scholar]
  • 16.Pass RF, Fowler KB, Boppana SB, Britt WJ, Stagno S. 2006. Congenital cytomegalovirus infection following first trimester maternal infection: symptoms at birth and outcome. J Clin Virol 35:216–220. doi: 10.1016/j.jcv.2005.09.015. [DOI] [PubMed] [Google Scholar]
  • 17.Picone O, Vauloup-Fellous C, Cordier AG, Guitton S, Senat MV, Fuchs F, Ayoubi JM, Grangeot Keros L, Benachi A. 2013. A series of 238 cytomegalovirus primary infections during pregnancy: description and outcome. Prenat Diagn 33:751–758. doi: 10.1002/pd.4118. [DOI] [PubMed] [Google Scholar]
  • 18.Lilleri D, Kabanova A, Revello MG, Percivalle E, Sarasini A, Genini E, Sallusto F, Lanzavecchia A, Corti D, Gerna G. 2013. Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130–131 complex during primary infection. PLoS One 8:e59863. doi: 10.1371/journal.pone.0059863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lilleri D, Fornara C, Furione M, Zavattoni M, Revello MG, Gerna G. 2007. Development of human cytomegalovirus—specific T cell immunity during primary infection of pregnant women and its correlation with virus transmission to the fetus. J Infect Dis 195:1062–1070. doi: 10.1086/512245. [DOI] [PubMed] [Google Scholar]
  • 20.Fornara C, Furione M, Arossa A, Gerna G, Lilleri D. 2016. Comparative magnitude and kinetics of human cytomegalovirus-specific CD4+ and CD8+ T-cell responses in pregnant women with primary versus remote infection and in transmitting versus non-transmitting mothers: its utility for dating primary infection in pregnancy. J Med Virol 88:1238–1246. doi: 10.1002/jmv.24449. [DOI] [PubMed] [Google Scholar]
  • 21.Tabata T, McDonagh S, Kawakatsu H, Pereira L. 2007. Cytotrophoblasts infected with a pathogenic human cytomegalovirus strain dysregulate cell–matrix and cell–cell adhesion molecules: a quantitative analysis. Placenta 28:527–537. doi: 10.1016/j.placenta.2006.05.006. [DOI] [PubMed] [Google Scholar]
  • 22.Perlman JM, Argyle C. 1992. Lethal cytomegalovirus infection in preterm infants: clinical, radiological, and neuropathological findings. Ann Neurol 31:64–68. doi: 10.1002/ana.410310112. [DOI] [PubMed] [Google Scholar]
  • 23.Odeberg J, Wolmer N, Falci S, Westgren M, Seiger Å, Söderberg-Nauclér C. 2006. Human cytomegalovirus inhibits neuronal differentiation and induces apoptosis in human neural precursor cells. J Virol 80:8929–8939. doi: 10.1128/JVI.00676-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Nigro G. 2009. Maternal–fetal cytomegalovirus infection: from diagnosis to therapy. J Matern Fetal Neonatal Med 22:169–174. doi: 10.1080/14767050802609767. [DOI] [PubMed] [Google Scholar]
  • 25.Cannon MJ, Hyde TB, Schmid DS. 2011. Review of cytomegalovirus shedding in bodily fluids and relevance to congenital cytomegalovirus infection. Rev Med Virol 21:240–255. doi: 10.1002/rmv.695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Dollard SC, Grosse SD, Ross DS. 2007. New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection. Rev Med Virol 17:355–363. doi: 10.1002/rmv.544. [DOI] [PubMed] [Google Scholar]
  • 27.Tsuge M, Hida AI, Minematsu T, Honda N, Oshiro Y, Yokoyama M, Kondo Y. 2019. Prospective cohort study of congenital cytomegalovirus infection during pregnancy with fetal growth restriction: serologic analysis and placental pathology. J Pediatr 206:42–48. doi: 10.1016/j.jpeds.2018.10.003. [DOI] [PubMed] [Google Scholar]
  • 28.Chan G, Guilbert LJ. 2006. Ultraviolet-inactivated human cytomegalovirus induces placental syncytiotrophoblast apoptosis in a Toll-like receptor-2 and tumour necrosis factor-alpha dependent manner. J Pathol 210:111–120. doi: 10.1002/path.2025. [DOI] [PubMed] [Google Scholar]
  • 29.Chaudhuri S, Lowen B, Chan G, Davey A, Riddell M, Guilbert LJ. 2009. Human cytomegalovirus interacts with toll-like receptor 2 and CD14 on syncytiotrophoblasts to stimulate expression of TNFalpha mRNA and apoptosis. Placenta 30:994–1001. doi: 10.1016/j.placenta.2009.09.001. [DOI] [PubMed] [Google Scholar]
  • 30.Fabbri E, Revello MG, Furione M, Zavattoni M, Lilleri D, Tassis B, Quarenghi A, Rustico M, Nicolini U, Ferrazzi E, Gerna G. 2011. Prognostic markers of symptomatic congenital human cytomegalovirus infection in fetal blood. BJOG 118:448–456. doi: 10.1111/j.1471-0528.2010.02822.x. [DOI] [PubMed] [Google Scholar]
  • 31.Enders M, Daiminger A, Exler S, Ertan K, Enders G, Bald R. 2017. Prenatal diagnosis of congenital cytomegalovirus infection in 115 cases: a 5 years' single center experience. Prenat Diagn 37:389–398. doi: 10.1002/pd.5025. [DOI] [PubMed] [Google Scholar]
  • 32.Weisblum Y, Panet A, Zakay-Rones Z, Vitenshtein A, Haimov-Kochman R, Goldman-Wohl D, Oiknine-Djian E, Yamin R, Meir K, Amsalem H, Imbar T, Mandelboim O, Yagel S, Wolf DG. 2015. Human cytomegalovirus induces a distinct innate immune response in the maternal-fetal interface. Virology 485:289–296. doi: 10.1016/j.virol.2015.06.023. [DOI] [PubMed] [Google Scholar]
  • 33.Kimberlin DW, Lin C-Y, Sánchez PJ, Demmler GJ, Dankner W, Shelton M, Jacobs RF, Vaudry W, Pass RF, Kiell JM, Soong S-J, Whitley RJ. 2003. Effect of ganciclovir therapy on hearing in symptomatic congenital cytomegalovirus disease involving the central nervous system: a randomized, controlled trial. J Pediatr 143:16–25. doi: 10.1016/S0022-3476(03)00192-6. [DOI] [PubMed] [Google Scholar]
  • 34.Patton ME, Bernstein K, Liu G, Zaidi A, Markowitz LE. 2018. Seroprevalence of herpes simplex virus types 1 and 2 among pregnant women and sexually active, nonpregnant women in the United States. Clin Infect Dis 67:1535–1542. doi: 10.1093/cid/ciy318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Simmons A. 2002. Clinical manifestations and treatment considerations of herpes simplex virus infection. J Infect Dis 186(Suppl 1):S71–S77. doi: 10.1086/342967. [DOI] [PubMed] [Google Scholar]
  • 36.Kimberlin DW. 2004. Neonatal herpes simplex infection. Clin Microbiol Rev 17:1–13. doi: 10.1128/cmr.17.1.1-13.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kimberlin DW, Lin CY, Jacobs RF, Powell DA, Corey L, Gruber WC, Rathore M, Bradley JS, Diaz PS, Kumar M, Arvin AM, Gutierrez K, Shelton M, Weiner LB, Sleasman JW, de Sierra TM, Weller S, Soong SJ, Kiell J, Lakeman FD, Whitley RJ, National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group. 2001. Safety and efficacy of high-dose intravenous acyclovir in the management of neonatal herpes simplex virus infections. Pediatrics 108:230–238. doi: 10.1542/peds.108.2.230. [DOI] [PubMed] [Google Scholar]
  • 38.Whitley R, Arvin A, Prober C, Corey L, Burchett S, Plotkin S, Starr S, Jacobs R, Powell D, Nahmias A. 1991. Predictors of morbidity and mortality in neonates with herpes simplex virus infections. The National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group. N Engl J Med 324:450–454. doi: 10.1056/NEJM199102143240704. [DOI] [PubMed] [Google Scholar]
  • 39.Hyde SR, Giacoia GP. 1993. Congenital herpes infection: placental and umbilical cord findings. Obstet Gynecol 81:852–855. [PubMed] [Google Scholar]
  • 40.Peng J, Krause PJ, Kresch M. 1996. Neonatal herpes simplex virus infection after cesarean section with intact amniotic membranes. J Perinatol 16:397–399. [PubMed] [Google Scholar]
  • 41.Schust DJ, Hill AB, Ploegh HL. 1996. Herpes simplex virus blocks intracellular transport of HLA-G in placentally derived human cells. J Immunol 157:3375–3380. [PubMed] [Google Scholar]
  • 42.Nørskov-Lauritsen N, Aboagye-Mathisen G, Juhl CB, Petersen PM, Zachar V, Ebbesen P. 1992. Herpes simplex virus infection of cultured human term trophoblast. J Med Virol 36:162–166. doi: 10.1002/jmv.1890360303. [DOI] [PubMed] [Google Scholar]
  • 43.Jabłońska A, Studzińska M, Suski P, Kalinka J, Paradowska E. 2018. Enhanced expression of IFI16 and RIG-I in human third-trimester placentas following HSV-1 infection. Clin Exp Immunol 193:255–263. doi: 10.1111/cei.13143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Mogensen TH, Melchjorsen J, Hollsberg P, Paludan SR. 2003. Activation of NF-kappa B in virus-infected macrophages is dependent on mitochondrial oxidative stress and intracellular calcium: downstream involvement of the kinases TGF-beta-activated kinase 1, mitogen-activated kinase/extracellular signal-regulated kinase kinase 1, and I kappa B kinase. J Immunol 170:6224–6233. doi: 10.4049/jimmunol.170.12.6224. [DOI] [PubMed] [Google Scholar]
  • 45.Mogensen TH, Melchjorsen J, Malmgaard L, Casola A, Paludan SR. 2004. Suppression of proinflammatory cytokine expression by herpes simplex virus type 1. J Virol 78:5883–5890. doi: 10.1128/JVI.78.11.5883-5890.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ellermann-Eriksen S. 2005. Macrophages and cytokines in the early defence against herpes simplex virus. Virol J 2:59. doi: 10.1186/1743-422X-2-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Gershon AA, Breuer J, Cohen JI, Cohrs RJ, Gershon MD, Gilden D, Grose C, Hambleton S, Kennedy PGE, Oxman MN, Seward JF, Yamanishi K. 2015. Varicella zoster virus infection. Nat Rev Dis Primers 1:15016. doi: 10.1038/nrdp.2015.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lamont RF, Sobel JD, Carrington D, Mazaki-Tovi S, Kusanovic JP, Vaisbuch E, Romero R. 2011. Varicella-zoster virus (chickenpox) infection in pregnancy. BJOG 118:1155–1162. doi: 10.1111/j.1471-0528.2011.02983.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Smith CK, Arvin AM. 2009. Varicella in the fetus and newborn. Semin Fetal Neonat Med 14:209–217. doi: 10.1016/j.siny.2008.11.008. [DOI] [PubMed] [Google Scholar]
  • 50.Collie MH, Sweet C, Cavanagh D, Smith H. 1978. Association of foetal wastage with influenza infection during ferret pregnancy. Br J Exp Pathol 59:190–195. [PMC free article] [PubMed] [Google Scholar]
  • 51.Higa K, Dan K, Manabe H. 1987. Varicella-zoster virus infections during pregnancy: hypothesis concerning the mechanisms of congenital malformations. Obstet Gynecol 69:214–222. [PubMed] [Google Scholar]
  • 52.Grose C. 1989. Congenital varicella-zoster virus infection and the failure to establish virus-specific cell-mediated immunity. Mol Biol Med 6:453–462. [PubMed] [Google Scholar]
  • 53.Zerboni L, Arvin A. 2015. Neuronal subtype and satellite cell tropism are determinants of varicella-zoster virus virulence in human dorsal root ganglia xenografts in vivo. PLoS Pathog 11:e1004989. doi: 10.1371/journal.ppat.1004989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Biber K, Neumann H, Inoue K, Boddeke HWGM. 2007. Neuronal “on” and “off” signals control microglia. Trends Neurosci 30:596–602. doi: 10.1016/j.tins.2007.08.007. [DOI] [PubMed] [Google Scholar]
  • 55.Huang YC, Lin TY, Lin YJ, Lien RI, Chou YH. 2001. Prophylaxis of intravenous immunoglobulin and acyclovir in perinatal varicella. Eur J Pediatr 160:91–94. doi: 10.1007/s004310000640. [DOI] [PubMed] [Google Scholar]
  • 56.Gordon A, Reingold A. 2018. The burden of influenza: a complex problem. Curr Epidemiol Rep 5:1–9. doi: 10.1007/s40471-018-0136-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bouvier NM, Palese P. 2008. The biology of influenza viruses. Vaccine 26(Suppl 4):D49–D53. doi: 10.1016/j.vaccine.2008.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Harris JW. 1919. Influenza occurring in pregnant women: a statistical study of thirteen hundred and fifty cases. JAMA 72:978–980. doi: 10.1001/jama.1919.02610140008002. [DOI] [Google Scholar]
  • 59.Jamieson DJ, Honein MA, Rasmussen SA, Williams JL, Swerdlow DL, Biggerstaff MS, Lindstrom S, Louie JK, Christ CM, Bohm SR, Fonseca VP, Ritger KA, Kuhles DJ, Eggers P, Bruce H, Davidson HA, Lutterloh E, Harris ML, Burke C, Cocoros N, Finelli L, MacFarlane KF, Shu B, Olsen SJ, Novel Influenza A (H1N1) Pregnancy Working Group. 2009. H1N1 2009 influenza virus infection during pregnancy in the USA. Lancet 374:451–458. doi: 10.1016/S0140-6736(09)61304-0. [DOI] [PubMed] [Google Scholar]
  • 60.Rasmussen SA, Jamieson DJ. 2012. Influenza and pregnancy in the United States: before, during, and after 2009 H1N1. Clin Obstet Gynecol 55:487–497. doi: 10.1097/GRF.0b013e31824df23e. [DOI] [PubMed] [Google Scholar]
  • 61.Creanga AA, Kamimoto L, Newsome K, D'Mello T, Jamieson DJ, Zotti ME, Arnold KE, Baumbach J, Bennett NM, Farley MM, Gershman K, Kirschke D, Lynfield R, Meek J, Morin C, Reingold A, Ryan P, Schaffner W, Thomas A, Zansky S, Finelli L, Honein MA. 2011. Seasonal and 2009 pandemic influenza A (H1N1) virus infection during pregnancy: a population-based study of hospitalized cases. Am J Obstet Gynecol 204:S38–S45. doi: 10.1016/j.ajog.2011.02.037. [DOI] [PubMed] [Google Scholar]
  • 62.Ramphal R, Donnelly WH, Small PA. 1980. Fatal influenzal pneumonia in pregnancy: failure to demonstrate transplacental transmission of influenza virus. Am J Obstet Gynecol 138:347–348. doi: 10.1016/0002-9378(80)90265-3. [DOI] [PubMed] [Google Scholar]
  • 63.Meijer WJ, Wensing AM, Bruinse HW, Nikkels PG. 2014. High rate of chronic villitis in placentas of pregnancies complicated by influenza A/H1N1 infection. Infect Dis Obstet Gynecol 2014:768380. doi: 10.1155/2014/768380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Neuzil KM, Reed GW, Mitchel EF, Simonsen L, Griffin MR. 1998. Impact of influenza on acute cardiopulmonary hospitalizations in pregnant women. Am J Epidemiol 148:1094–1102. doi: 10.1093/oxfordjournals.aje.a009587. [DOI] [PubMed] [Google Scholar]
  • 65.Van Kerkhove MD, Vandemaele KA, Shinde V, Jaramillo-Gutierrez G, Koukounari A, Donnelly CA, Carlino LO, Owen R, Paterson B, Pelletier L, Vachon J, Gonzalez C, Hongjie Y, Zijian F, Chuang SK, Au A, Buda S, Krause G, Haas W, Bonmarin I, Taniguichi K, Nakajima K, Shobayashi T, Takayama Y, Sunagawa T, Heraud JM, Orelle A, Palacios E, van der Sande MA, Wielders CC, Hunt D, Cutter J, Lee VJ, Thomas J, Santa-Olalla P, Sierra-Moros MJ, Hanshaoworakul W, Ungchusak K, Pebody R, Jain S, Mounts AW, on behalf of the WHO Working Group for Risk Factors for Severe H1N1pdm Infection. 2011. Risk factors for severe outcomes following 2009 influenza A (H1N1) infection: a global pooled analysis. PLoS Med 8:e1001053. doi: 10.1371/journal.pmed.1001053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Potluri T, Fink AL, Sylvia KE, Dhakal S, Vermillion MS, Vom Steeg L, Deshpande S, Narasimhan H, Klein SL. 2019. Age-associated changes in the impact of sex steroids on influenza vaccine responses in males and females. NPJ Vaccines 4:29–29. doi: 10.1038/s41541-019-0124-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Vom Steeg LG, Klein SL. 2019. Sex and sex steroids impact influenza pathogenesis across the life course. Semin Immunopathol 41:189–194. doi: 10.1007/s00281-018-0718-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hennet T, Ziltener HJ, Frei K, Peterhans E. 1992. A kinetic study of immune mediators in the lungs of mice infected with influenza A virus. J Immunol 149:932–939. [PubMed] [Google Scholar]
  • 69.Kash JC, Basler CF, Garcia-Sastre A, Carter V, Billharz R, Swayne DE, Przygodzki RM, Taubenberger JK, Katze MG, Tumpey TM. 2004. Global host immune response: pathogenesis and transcriptional profiling of type A influenza viruses expressing the hemagglutinin and neuraminidase genes from the 1918 pandemic virus. J Virol 78:9499–9511. doi: 10.1128/JVI.78.17.9499-9511.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Perrone LA, Plowden JK, Garcia-Sastre A, Katz JM, Tumpey TM. 2008. H5N1 and 1918 pandemic influenza virus infection results in early and excessive infiltration of macrophages and neutrophils in the lungs of mice. PLoS Pathog 4:e1000115. doi: 10.1371/journal.ppat.1000115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tumpey TM, Garcia-Sastre A, Taubenberger JK, Palese P, Swayne DE, Pantin-Jackwood MJ, Schultz-Cherry S, Solorzano A, Van Rooijen N, Katz JM, Basler CF. 2005. Pathogenicity of influenza viruses with genes from the 1918 pandemic virus: functional roles of alveolar macrophages and neutrophils in limiting virus replication and mortality in mice. J Virol 79:14933–14944. doi: 10.1128/JVI.79.23.14933-14944.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bachy V, Williams DJ, Ibrahim MA. 2008. Altered dendritic cell function in normal pregnancy. J Reprod Immunol 78:11–21. doi: 10.1016/j.jri.2007.09.004. [DOI] [PubMed] [Google Scholar]
  • 73.Della Bella S, Giannelli S, Cozzi V, Signorelli V, Cappelletti M, Cetin I, Villa ML. 2011. Incomplete activation of peripheral blood dendritic cells during healthy human pregnancy. Clin Exp Immunol 164:180–192. doi: 10.1111/j.1365-2249.2011.04330.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kraus TA, Engel SM, Sperling RS, Kellerman L, Lo Y, Wallenstein S, Escribese MM, Garrido JL, Singh T, Loubeau M, Moran TM. 2012. Characterizing the pregnancy immune phenotype: results of the viral immunity and pregnancy (VIP) study. J Clin Immunol 32:300–311. doi: 10.1007/s10875-011-9627-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chan JF, To KK, Tse H, Lau CC, Li IW, Hung IF, Chan KH, Cheng VC, Lai TS, Woo PC, Chan EY, Yuen KY. 2011. The lower serum immunoglobulin G2 level in severe cases than in mild cases of pandemic H1N1 2009 influenza is associated with cytokine dysregulation. Clin Vaccine Immunol 18:305–310. doi: 10.1128/CVI.00363-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Forbes RL, Wark PA, Murphy VE, Gibson PG. 2012. Pregnant women have attenuated innate interferon responses to 2009 pandemic influenza A virus subtype H1N1. J Infect Dis 206:646–653. doi: 10.1093/infdis/jis377. [DOI] [PubMed] [Google Scholar]
  • 77.Kay AW, Bayless NL, Fukuyama J, Aziz N, Dekker CL, Mackey S, Swan GE, Davis MM, Blish CA. 2015. Pregnancy does not attenuate the antibody or plasmablast response to inactivated influenza vaccine. J Infect Dis 212:861–870. doi: 10.1093/infdis/jiv138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sperling RS, Engel SM, Wallenstein S, Kraus TA, Garrido J, Singh T, Kellerman L, Moran TM. 2012. Immunogenicity of trivalent inactivated influenza vaccination received during pregnancy or postpartum. Obstet Gynecol 119:631–639. doi: 10.1097/AOG.0b013e318244ed20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Jackson LA, Patel SM, Swamy GK, Frey SE, Creech CB, Munoz FM, Artal R, Keitel WA, Noah DL, Petrie CR, Wolff M, Edwards KM. 2011. Immunogenicity of an inactivated monovalent 2009 H1N1 influenza vaccine in pregnant women. J Infect Dis 204:854–863. doi: 10.1093/infdis/jir440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Schlaudecker EP, Ambroggio L, McNeal MM, Finkelman FD, Way SS. 2018. Declining responsiveness to influenza vaccination with progression of human pregnancy. Vaccine 36:4734–4741. doi: 10.1016/j.vaccine.2018.05.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Marcelin G, Aldridge JR, Duan S, Ghoneim HE, Rehg J, Marjuki H, Boon AC, McCullers JA, Webby RJ. 2011. Fatal outcome of pandemic H1N1 2009 influenza virus infection is associated with immunopathology and impaired lung repair, not enhanced viral burden, in pregnant mice. J Virol 85:11208–11219. doi: 10.1128/JVI.00654-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Vermillion MS, Ursin RL, Attreed SE, Klein SL. 2018. Estriol reduces pulmonary immune cell recruitment and inflammation to protect female mice from severe influenza. Endocrinology 159:3306–3320. doi: 10.1210/en.2018-00486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Robinson DP, Lorenzo ME, Jian W, Klein SL. 2011. Elevated 17beta-estradiol protects females from influenza A virus pathogenesis by suppressing inflammatory responses. PLoS Pathog 7:e1002149. doi: 10.1371/journal.ppat.1002149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hall OJ, Nachbagauer R, Vermillion MS, Fink AL, Phuong V, Krammer F, Klein SL. 2017. Progesterone-based contraceptives reduce adaptive immune responses and protection against sequential influenza A virus infections. J Virol 91:e02160-16. doi: 10.1128/JVI.02160-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Vermillion MS, Nelson A, Vom Steeg L, Loube J, Mitzner W, Klein SL. 2018. Pregnancy preserves pulmonary function following influenza virus infection in C57BL/6 mice. Am J Physiol Lung Cell Mol Physiol 315:L517–L525. doi: 10.1152/ajplung.00066.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sweet C, Collie MH, Toms GL, Smith H. 1977. The pregnant guinea-pig as a model for studying influenza virus infection in utero: infection of foetal tissues in organ culture and in vivo. Br J Exp Pathol 58:133–139. [PMC free article] [PubMed] [Google Scholar]
  • 87.Kwit K, Pomorska-Mol M, Markowska-Daniel I. 2014. The influence of experimental infection of gilts with swine H1N2 influenza A virus during the second month of gestation on the course of pregnancy, reproduction parameters and clinical status. BMC Vet Res 10:123. doi: 10.1186/1746-6148-10-123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kwit K, Pomorska-Mol M, Markowska-Daniel I. 2015. 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 160:2415–2425. doi: 10.1007/s00705-015-2518-8. [DOI] [PubMed] [Google Scholar]
  • 89.Kim HM, Kang YM, Song BM, Kim HS, Seo SH. 2012. The 2009 pandemic H1N1 influenza virus is more pathogenic in pregnant mice than seasonal H1N1 influenza virus. Viral Immunol 25:402–410. doi: 10.1089/vim.2012.0007. [DOI] [PubMed] [Google Scholar]
  • 90.Pazos MA, Kraus TA, Munoz-Fontela C, Moran TM. 2012. Estrogen mediates innate and adaptive immune alterations to influenza infection in pregnant mice. PLoS One 7:e40502. doi: 10.1371/journal.pone.0040502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Engels G, Hierweger AM, Hoffmann J, Thieme R, Thiele S, Bertram S, Dreier C, Resa-Infante P, Jacobsen H, Thiele K, Alawi M, Indenbirken D, Grundhoff A, Siebels S, Fischer N, Stojanovska V, Muzzio D, Jensen F, Karimi K, Mittrucker HW, Arck PC, Gabriel G. 2017. Pregnancy-related immune adaptation promotes the emergence of highly virulent H1N1 influenza virus strains in allogenically pregnant mice. Cell Host Microbe 21:321–333. doi: 10.1016/j.chom.2017.02.020. [DOI] [PubMed] [Google Scholar]
  • 92.Kim JC, Kim HM, Kang YM, Ku KB, Park EH, Yum J, Kim JA, Kang YK, Lee JS, Kim HS, Seo SH. 2014. Severe pathogenesis of influenza B virus in pregnant mice. Virology 448:74–81. doi: 10.1016/j.virol.2013.10.001. [DOI] [PubMed] [Google Scholar]
  • 93.Littauer EQ, Esser ES, Antao OQ, Vassilieva EV, Compans RW, Skountzou I. 2017. H1N1 influenza virus infection results in adverse pregnancy outcomes by disrupting tissue-specific hormonal regulation. PLoS Pathog 13:e1006757. doi: 10.1371/journal.ppat.1006757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Moore CA, Staples JE, Dobyns WB, Pessoa A, Ventura CV, Fonseca EB, Ribeiro EM, Ventura LO, Neto NN, Arena JF, Rasmussen SA. 2017. Characterizing the pattern of anomalies in congenital Zika syndrome for pediatric clinicians. JAMA Pediatr 171:288–295. doi: 10.1001/jamapediatrics.2016.3982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Anonymous. . 2017. Zika, sexual transmission and prudent public health policy. Public Health 148:66–68. doi: 10.1016/j.puhe.2017.02.018. [DOI] [PubMed] [Google Scholar]
  • 96.Anonymous. . 2017. Zika virus: history, epidemiology, transmission, and clinical presentation. J Neuroimmunol 308:50–64. doi: 10.1016/j.jneuroim.2017.03.001. [DOI] [PubMed] [Google Scholar]
  • 97.Lowe R, Barcellos C, Brasil P, Cruz OG, Honório NA, Kuper H, Carvalho MS. 2018. The Zika virus epidemic in Brazil: from discovery to future implications. Int J Environ Res Public Health 15:96. doi: 10.3390/ijerph15010096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Anonymous. . 2016. Anticipating the international spread of Zika virus from Brazil. Lancet 387:335–336. doi: 10.1016/S0140-6736(16)00080-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Anonymous. . 2016. Zika virus infects human placental macrophages. Cell Host Microbe 20:83–90. doi: 10.1016/j.chom.2016.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Vermillion MS, Lei J, Shabi Y, Baxter VK, Crilly NP, McLane M, Griffin DE, Pekosz A, Klein SL, Burd I. 2017. Intrauterine Zika virus infection of pregnant immunocompetent mice models transplacental transmission and adverse perinatal outcomes. Nat Commun 8:14575. doi: 10.1038/ncomms14575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Onorati M, Li Z, Liu F, Sousa AMM, Nakagawa N, Li M, Dell'Anno MT, Gulden FO, Pochareddy S, Tebbenkamp ATN, Han W, Pletikos M, Gao T, Zhu Y, Bichsel C, Varela L, Szigeti-Buck K, Lisgo S, Zhang Y, Testen A, Gao XB, Mlakar J, Popovic M, Flamand M, Strittmatter SM, Kaczmarek LK, Anton ES, Horvath TL, Lindenbach BD, Sestan N. 2016. Zika virus disrupts phospho-TBK1 localization and mitosis in human neuroepithelial stem cells and radial glia. Cell Rep 16:2576–2592. doi: 10.1016/j.celrep.2016.08.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wu Y, Liu Q, Zhou J, Xie W, Chen C, Wang Z, Yang H, Cui J. 2017. Zika virus evades interferon-mediated antiviral response through the co-operation of multiple nonstructural proteins in vitro. Cell Discov 3:17006. doi: 10.1038/celldisc.2017.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li C, Xu D, Ye Q, Hong S, Jiang Y, Liu X, Zhang N, Shi L, Qin CF, Xu Z. 2016. Zika virus disrupts neural progenitor development and leads to microcephaly in mice. Cell Stem Cell 19:120–126. doi: 10.1016/j.stem.2016.04.017. [DOI] [PubMed] [Google Scholar]
  • 104.Anonymous. . 2020. An immunocompetent mouse model of Zika virus infection. Cell Host Microbe 23:672–685. doi: 10.1016/j.chom.2018.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Musso D, Bossin H, Mallet HP, Besnard M, Broult J, Baudouin L, Levi JE, Sabino EC, Ghawche F, Lanteri MC, Baud D. 2018. Zika virus in French Polynesia 2013–14: anatomy of a completed outbreak. Lancet Infect Dis 18:e172–e182. doi: 10.1016/S1473-3099(17)30446-2. [DOI] [PubMed] [Google Scholar]
  • 106.Musso D, Ko AI, Baud D. 2019. Zika virus infection—after the pandemic. N Engl J Med 381:1444–1457. doi: 10.1056/NEJMra1808246. [DOI] [PubMed] [Google Scholar]
  • 107.Musso D, Gubler DJ. 2016. Zika virus. Clin Microbiol Rev 29:487–524. doi: 10.1128/CMR.00072-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Anonymous. . 2020. Experience in diagnosing congenital Zika syndrome in Brazilian children born to asymptomatic mothers. Acta Tropica 206:105438. doi: 10.1016/j.actatropica.2020.105438. [DOI] [PubMed] [Google Scholar]
  • 109.Centers for Disease Control and Prevention. 2019. Clinical evaluation & disease. Zika virus. Centers for Disease Control and Prevention, Atlanta, GA. [Google Scholar]
  • 110.Anonymous. . 2015. Neurological syndrome, congenital malformations, and Zika virus infection. World Health Organization, Geneva, Switzerland. https://www.paho.org/hq/dmdocuments/2015/2015-dec-1-cha-epi-alert-zika-neuro-syndrome.pdf. [Google Scholar]
  • 111.Zare Mehrjardi M, Carteaux G, Poretti A, Sanei Taheri M, Bermudez S, Werner H, Hygino da Cruz LC, Jr. 2017. Neuroimaging findings of postnatally acquired Zika virus infection: a pictorial essay. Jpn J Radiol 35:341–349. doi: 10.1007/s11604-017-0641-z. [DOI] [PubMed] [Google Scholar]
  • 112.Cugola FR, Fernandes IR, Russo FB, Freitas BC, Dias JLM, Guimarães KP, Benazzato C, Almeida N, Pignatari GC, Romero S, Polonio CM, Cunha I, Freitas CL, Brandão WN, Rossato C, Andrade DG, Faria D. d P, Garcez AT, Buchpigel CA, Braconi CT, Mendes E, Sall AA, Zanotto P. M d A, Peron JPS, Muotri AR, Beltrão-Braga PCB. 2016. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 534:267–271. doi: 10.1038/nature18296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Anonymous. . 2018. Review: evidence of neurological sequelae in children with acquired Zika virus infection. Pediatr Neurol 85:16–20. doi: 10.1016/j.pediatrneurol.2018.03.001. [DOI] [PubMed] [Google Scholar]
  • 114.Souza INO, Barros-Aragao FGQ, Frost PS, Figueiredo CP, Clarke JR. 2019. Late neurological consequences of Zika virus infection: risk factors and pharmaceutical approaches. Pharmaceuticals 12:60. doi: 10.3390/ph12020060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Lee JK, Shin OS. 2019. Advances in Zika virus-host cell interaction: current knowledge and future perspectives. Int J Mol Sci 20:1101. doi: 10.3390/ijms20051101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Gurung S, Reuter N, Preno A, Dubaut J, Nadeau H, Hyatt K, Singleton K, Martin A, Parks WT, Papin JF, Myers DA. 2019. Zika virus infection at mid-gestation results in fetal cerebral cortical injury and fetal death in the olive baboon. PLoS Pathog 15:e1007507. doi: 10.1371/journal.ppat.1007507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Seferovic M, Sánchez-San Martín C, Tardif SD, Rutherford J, Castro ECC, Li T, Hodara VL, Parodi LM, Giavedoni L, Layne-Colon D, Tamhankar M, Yagi S, Martyn C, Reyes K, Suter MA, Aagaard KM, Chiu CY, Patterson JL. 2018. Experimental Zika virus infection in the pregnant common marmoset induces spontaneous fetal loss and neurodevelopmental abnormalities. Sci Rep 8:6851. doi: 10.1038/s41598-018-25205-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lei J, Vermillion MS, Jia B, Xie H, Xie L, McLane MW, Sheffield JS, Pekosz A, Brown A, Klein SL, Burd I. 2019. IL-1 receptor antagonist therapy mitigates placental dysfunction and perinatal injury following Zika virus infection. JCI Insight 4:e122678. doi: 10.1172/jci.insight.122678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Nesheim S, Taylor A, Lampe MA, Kilmarx PH, Fitz Harris L, Whitmore S, Griffith J, Thomas-Proctor M, Fenton K, Mermin J. 2012. A framework for elimination of perinatal transmission of HIV in the United States. Pediatrics 130:738–744. doi: 10.1542/peds.2012-0194. [DOI] [PubMed] [Google Scholar]
  • 120.Cock KMD, Fowler MG, Mercier E, Vincenzi I, Saba J, Hoff E, Alnwick DJ, Rogers M, Shaffer N. 2000. Prevention of mother-to-child HIV transmission in resource-poor countries: translating research into policy and practice. JAMA 283:1175–1182. doi: 10.1001/jama.283.9.1175. [DOI] [PubMed] [Google Scholar]
  • 121.John GC, Nduati RW, Mbori-Ngacha DA, Richardson BA, Panteleeff D, Mwatha A, Overbaugh J, Bwayo J, Ndinya-Achola JO, Kreiss JK. 2001. Correlates of mother-to-child human immunodeficiency virus type 1 (HIV-1) transmission: association with maternal plasma HIV-1 RNA load, genital HIV-1 DNA shedding, and breast infections. J Infect Dis 183:206–212. doi: 10.1086/317918. [DOI] [PubMed] [Google Scholar]
  • 122.Fowler MG, Kourtis Ap Aizire J, Onyango-Makumbi C, Bulterys M. 2012. Breastfeeding and transmission of HIV-1: epidemiology and global magnitude, p 3–25. In Kourtis AP, Bulterys M (ed), Human immunodeficiency virus type 1 (HIV-1) and breastfeeding: science, research advances, and policy. Springer New York, New York, NY. [DOI] [PubMed] [Google Scholar]
  • 123.Lairmore MD, Cuthbert PS, Utley LL, Morgan CJ, Dezzutti CS, Anderson CL, Sedmak DD. 1993. Cellular localization of CD4 in the human for maternal-to-fetal transmission of HIV. J Immunol 151:1673–1681. [PubMed] [Google Scholar]
  • 124.Johnson EL, Chakraborty R. 2012. Placental Hofbauer cells limit HIV-1 replication and potentially offset mother to child transmission (MTCT) by induction of immunoregulatory cytokines. Retrovirology 9:101–101. doi: 10.1186/1742-4690-9-101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Brocklehurst P, French R. 1998. The association between maternal HIV infection and perinatal outcome: a systematic review of the literature and meta-analysis. Br J Obstet Gynaecol 105:836–848. doi: 10.1111/j.1471-0528.1998.tb10227.x. [DOI] [PubMed] [Google Scholar]
  • 126.Lambert JS, Watts DH, Mofenson L, Stiehm ER, Harris DR, Bethel J, Whitehouse J, Jimenez E, Gandia J, Scott G. 2000. Risk factors for preterm birth, low birth weight, and intrauterine growth retardation in infants born to HIV-infected pregnant women receiving zidovudine. Pediatric AIDS Clinical Trials Group 185 Team. AIDS 14:1389–1399. doi: 10.1097/00002030-200007070-00012. [DOI] [PubMed] [Google Scholar]
  • 127.Bodkin C, Klopper H, Langley G. 2006. A comparison of HIV positive and negative pregnant women at a public sector hospital in South Africa. J Clin Nurs 15:735–741. doi: 10.1111/j.1365-2702.2006.01438.x. [DOI] [PubMed] [Google Scholar]
  • 128.Marazzi MC, Palombi L, Nielsen-Saines K, Haswell J, Zimba I, Magid NA, Buonomo E, Scarcella P, Ceffa S, Paturzo G. 2011. Extended antenatal use of triple antiretroviral therapy for prevention of mother-to-child transmission of HIV-1 correlates with favorable pregnancy outcomes. AIDS 25:1611–1618. doi: 10.1097/QAD.0b013e3283493ed0. [DOI] [PubMed] [Google Scholar]
  • 129.Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, Alvarado M, Anderson HR, Anderson LM, Andrews KG, Atkinson C, Baddour LM, Barker-Collo S, Bartels DH, Bell ML, Benjamin EJ, Bennett D, Bhalla K, Bikbov B, Bin Abdulhak A, Birbeck G, Blyth F, Bolliger I, Boufous S, Bucello C, Burch M, Burney P, Carapetis J, Chen H, Chou D, Chugh SS, Coffeng LE, Colan SD, Colquhoun S, Colson KE, Condon J, Connor MD, Cooper LT, Corriere M, Cortinovis M, de Vaccaro KC, Couser W, Cowie BC, Criqui MH, Cross M, Dabhadkar KC, et al. 2012. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380:2095–2128. doi: 10.1016/S0140-6736(12)61728-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Doare KL, Bland R, Newell ML. 2012. Neurodevelopment in children born to HIV-infected mothers by infection and treatment status. Pediatrics 130:e1326–e1344. doi: 10.1542/peds.2012-0405. [DOI] [PubMed] [Google Scholar]
  • 131.Chandwani S, Greco MA, Mittal K, Antoine C, Krasinski K, Borkowsky W. 1991. Pathology and human immunodeficiency virus expression in placentas of seropositive women. J Infect Dis 163:1134–1138. doi: 10.1093/infdis/163.5.1134. [DOI] [PubMed] [Google Scholar]
  • 132.Ackerman W, Kwiek J. 2013. Role of the placenta in adverse perinatal outcomes among HIV-1 seropositive women. J Nippon Med Sch 80:90–94. doi: 10.1272/jnms.80.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Dauby N, Chamekh M, Melin P, Slogrove AL, Goetghebuer T. 2016. Increased risk of group B Streptococcus invasive infection in HIV-exposed but uninfected infants: a review of the evidence and possible mechanisms. Front Immunol 7:505. doi: 10.3389/fimmu.2016.00505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Locks LM, Manji KP, Kupka R, Liu E, Kisenge R, McDonald CM, Aboud S, Wang M, Fawzi WW, Duggan CP. 2017. High burden of morbidity and mortality but not growth failure in infants exposed to but uninfected with human immunodeficiency virus in Tanzania. J Pediatr 180:191–199. doi: 10.1016/j.jpeds.2016.09.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Shapiro RL, Lockman S. 2010. Mortality among HIV-exposed infants: the first and final frontier. Clin Infect Dis 50:445–447. doi: 10.1086/649887. [DOI] [PubMed] [Google Scholar]
  • 136.Slogrove AL, Cotton MF, Esser MM. 2010. Severe infections in HIV-exposed uninfected infants: clinical evidence of immunodeficiency. J Trop Pediatr 56:75–81. doi: 10.1093/tropej/fmp057. [DOI] [PubMed] [Google Scholar]
  • 137.Kuhn L, Kasonde P, Sinkala M, Kankasa C, Semrau K, Scott N, Tsai W-Y, Vermund SH, Aldrovandi GM, Thea DM. 2005. Does severity of HIV disease in HIV-infected mothers affect mortality and morbidity among their uninfected infants? Clin Infect Dis 41:1654–1661. doi: 10.1086/498029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.de Moraes-Pinto MI, Verhoeff F, Chimsuku L, Milligan PJM, Wesumperuma L, Broadhead RL, Brabin BJ, Johnson PM, Hart CA. 1998. Placental antibody transfer: influence of maternal HIV infection and placental malaria. Arch Dis Child Fetal Neonatal Ed 79:F202–F205. doi: 10.1136/fn.79.3.f202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Martinez DR, Fong Y, Li SH, Yang F, Jennewein MF, Weiner JA, Harrell EA, Mangold JF, Goswami R, Seage GR, III, Alter G, Ackerman ME, Peng X, Fouda GG, Permar SR. 2019. Fc characteristics mediate selective placental transfer of IgG in HIV-infected women. Cell 178:190–201. doi: 10.1016/j.cell.2019.05.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Anonymous. . 2000. Effect of breastfeeding on infant and child mortality due to infectious diseases in less developed countries: a pooled analysis. Lancet 355:451–455. doi: 10.1016/S0140-6736(00)82011-5. [DOI] [PubMed] [Google Scholar]
  • 141.Feiterna-Sperling C, Weizsaecker K, Bührer C, Casteleyn S, Loui A, Schmitz T, Wahn V, Obladen M. 2007. Hematologic effects of maternal antiretroviral therapy and transmission prophylaxis in HIV-1-exposed uninfected newborn infants. J Acquired Immune Defic Syndr 45:43–51. doi: 10.1097/QAI.0b013e318042d5e3. [DOI] [PubMed] [Google Scholar]
  • 142.Pacheco SE, McIntosh K, Lu M, Mofenson LM, Diaz C, Foca M, Frederick M, Handelsman E, Hayani K, Shearer WT, Women and Infants Transmission Study. 2006. Effect of perinatal antiretroviral drug exposure on hematologic values in HIV-uninfected children: an analysis of the women and infants transmission study. J Infect Dis 194:1089–1097. doi: 10.1086/507645. [DOI] [PubMed] [Google Scholar]
  • 143.El Beitune P, Duarte G. 2006. Antiretroviral agents during pregnancy: consequences on hematologic parameters in HIV-exposed, uninfected newborn infant. Eur J Obstet Gynecol Reprod Biol 128:59–63. doi: 10.1016/j.ejogrb.2006.01.013. [DOI] [PubMed] [Google Scholar]
  • 144.McNally LM, Jeena PM, Lalloo U, Nyamande K, Gajee K, Sturm AW, Goldblatt D, Tomkins AM, Coovadia HM. 2005. Probable mother to infant transmission of Pneumocystis jiroveci from an HIV-infected woman to her HIV-uninfected infant. AIDS 19:1548–1549. doi: 10.1097/01.aids.0000183941.67730.3a. [DOI] [PubMed] [Google Scholar]
  • 145.Mermin J, Lule J, Ekwaru JP, Downing R, Hughes P, Bunnell R, Malamba S, Ransom R, Kaharuza F, Coutinho A, Kigozi A, Quick R. 2005. Cotrimoxazole prophylaxis by HIV-infected persons in Uganda reduces morbidity and mortality among HIV-uninfected family members. AIDS 19:1035–1042. doi: 10.1097/01.aids.0000174449.32756.c7. [DOI] [PubMed] [Google Scholar]
  • 146.Hladik F, McElrath MJ. 2008. Setting the stage: host invasion by HIV. Nat Rev Immunol 8:447–457. doi: 10.1038/nri2302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ipp H, Zemlin AE, Erasmus RT, Glashoff RH. 2014. Role of inflammation in HIV-1 disease progression and prognosis. Crit Rev Clin Lab Sci 51:98–111. doi: 10.3109/10408363.2013.865702. [DOI] [PubMed] [Google Scholar]
  • 148.Fahey JL, Taylor JM, Manna B, Nishanian P, Aziz N, Giorgi JV, Detels R. 1998. Prognostic significance of plasma markers of immune activation, HIV viral load and CD4 T-cell measurements. AIDS 12:1581–1590. doi: 10.1097/00002030-199813000-00004. [DOI] [PubMed] [Google Scholar]
  • 149.Selvam A, Buhimschi IA, Makin JD, Pattinson RC, Anderson R, Forsyth BW. 2015. Hyperferritinemia and markers of inflammation and oxidative stress in the cord blood of HIV-exposed, uninfected (HEU) infants. HIV Med 16:375–380. doi: 10.1111/hiv.12214. [DOI] [PubMed] [Google Scholar]
  • 150.McDonald CR, Weckman AM, Conroy AL, Olwoch P, Natureeba P, Kamya MR, Havlir DV, Dorsey G, Kain KC. 2019. Systemic inflammation is associated with malaria and preterm birth in women living with HIV on antiretrovirals and co-trimoxazole. Sci Rep 9:6758. doi: 10.1038/s41598-019-43191-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Dirajlal-Fargo S, Mussi-Pinhata MM, Weinberg A, Yu Q, Cohen R, Harris DR, Bowman E, Gabriel J, Kulkarni M, Funderburg N, Chakhtoura N, McComsey GA, Protocol NL. 2019. HIV-exposed-uninfected infants have increased inflammation and monocyte activation. AIDS 33:845–853. doi: 10.1097/QAD.0000000000002128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Girard S, Sebire G. 2016. Transplacental transfer of interleukin-1 receptor agonist and antagonist following maternal immune activation. Am J Reprod Immunol 75:8–12. doi: 10.1111/aji.12444. [DOI] [PubMed] [Google Scholar]
  • 153.Le Campion A, Larouche A, Fauteux-Daniel S, Soudeyns H. 2012. Pathogenesis of hepatitis C during pregnancy and childhood. Viruses 4:3531–3550. doi: 10.3390/v4123531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ejeta E, Dabsu R. 2018. Prevalence of hepatitis C virus and HIV infection among pregnant women attending antenatal care clinic in western Ethiopia. Front Med 5:366. doi: 10.3389/fmed.2018.00366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Polis CB, Shah SN, Johnson KE, Gupta A. 2007. Impact of maternal HIV coinfection on the vertical transmission of hepatitis C virus: a meta-analysis. Clin Infect Dis 44:1123–1131. doi: 10.1086/512815. [DOI] [PubMed] [Google Scholar]
  • 156.Cottrell EB, Chou R, Wasson N, Rahman B, Guise JM. 2013. Reducing risk for mother-to-infant transmission of hepatitis C virus: a systematic review for the U.S. Preventive Services Task Force. Ann Intern Med 158:109–113. doi: 10.7326/0003-4819-158-2-201301150-00575. [DOI] [PubMed] [Google Scholar]
  • 157.Gervais A, Bacq Y, Bernuau J, Martinot M, Auperin A, Boyer N, Kilani A, Erlinger S, Valla D, Marcellin P. 2000. Decrease in serum ALT and increase in serum HCV RNA during pregnancy in women with chronic hepatitis C. J Hepatol 32:293–299. doi: 10.1016/s0168-8278(00)80075-6. [DOI] [PubMed] [Google Scholar]
  • 158.Wejstål R, Widell A, Norkrans G. 1998. HCV-RNA levels increase during pregnancy in women with chronic hepatitis C. Scand J Infect Dis 30:111–113. doi: 10.1080/003655498750003456. [DOI] [PubMed] [Google Scholar]
  • 159.Honegger JR, Kim S, Price AA, Kohout JA, McKnight KL, Prasad MR, Lemon SM, Grakoui A, Walker CM. 2013. Loss of immune escape mutations during persistent HCV infection in pregnancy enhances replication of vertically transmitted viruses. Nat Med 19:1529–1533. doi: 10.1038/nm.3351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Paternoster DM, Belligoli A, Ngaradoumbe NK, Visentin S, Franco R, Fagiuoli S, Boldrin C, Palù G, Baldo V, Floreani A. 2008. Endogenous interferon-alpha level is increased in hepatitis C virus (HCV)-positive pregnant women. J Clin Gastroenterol 42:204–207. doi: 10.1097/01.mcg.0000247991.81591.2e. [DOI] [PubMed] [Google Scholar]
  • 161.Benhammou V, Tubiana R, Matheron S, Sellier P, Mandelbrot L, Chenadec JL, Marel E, Khoshnood B, Warszawski J, ANRS CO1/CO11-EPF French Perinatal Cohort Study Group. 2018. HBV or HCV coinfection in HIV-1-infected pregnant women in France: prevalence and pregnancy outcomes. J Acquir Immune Defic Syndr 77:439–450. doi: 10.1097/QAI.0000000000001618. [DOI] [PubMed] [Google Scholar]
  • 162.Wijarnpreecha K, Thongprayoon C, Sanguankeo A, Upala S, Ungprasert P, Cheungpasitporn W. 2017. Hepatitis C infection and intrahepatic cholestasis of pregnancy: a systematic review and meta-analysis. Clin Res Hepatol Gastroenterol 41:39–45. doi: 10.1016/j.clinre.2016.07.004. [DOI] [PubMed] [Google Scholar]
  • 163.Safir A, Levy A, Sikuler E, Sheiner E. 2010. Maternal hepatitis B virus or hepatitis C virus carrier status as an independent risk factor for adverse perinatal outcome. Liver Int 30:765–770. doi: 10.1111/j.1478-3231.2010.02218.x. [DOI] [PubMed] [Google Scholar]
  • 164.Reddick KL, Jhaveri R, Gandhi M, James AH, Swamy GK. 2011. Pregnancy outcomes associated with viral hepatitis. J Viral Hepat 18:e394–e398. doi: 10.1111/j.1365-2893.2011.01436.x. [DOI] [PubMed] [Google Scholar]
  • 165.Buresi MC, Lee J, Gill S, Kong JM, Money DM, Yoshida EM, Hepatitis C Vertical Transmission Study Group. 2010. The prevalence of gestational diabetes mellitus and glucose abnormalities in pregnant women with hepatitis C virus infection in British Columbia. J Obstet Gynaecol Can 32:935–941. doi: 10.1016/s1701-2163(16)34680-1. [DOI] [PubMed] [Google Scholar]
  • 166.Money D, Boucoiran I, Wagner E, Dobson S, Kennedy A, Lohn Z, Krajden M, Yoshida EM. 2014. Obstetrical and neonatal outcomes among women infected with hepatitis C and their infants. J Obstet Gynaecol Can 36:785–794. doi: 10.1016/S1701-2163(15)30480-1. [DOI] [PubMed] [Google Scholar]
  • 167.Shiraki K, Ohto H, Inaba N, Fujisawa T, Tajiri H, Kanzaki S, Matsui A, Morishima T, Goto K, Kimura A, Hino S. 2008. Guidelines for care of pregnant women carrying hepatitis C virus and their infants. Pediatr Int 50:138–140. doi: 10.1111/j.1442-200X.2007.02518.x. [DOI] [PubMed] [Google Scholar]
  • 168.Compagnone A, Catenazzi P, Riccardi R, Zuppa AA. 2019. Mother-to-child transmission of hepatitis C virus. Minerva Pediatr 71:174–180. doi: 10.23736/S0026-4946.18.04898-3. [DOI] [PubMed] [Google Scholar]
  • 169.Conte D, Fraquelli M, Prati D, Colucci A, Minola E. 2000. Prevalence and clinical course of chronic hepatitis C virus (HCV) infection and rate of HCV vertical transmission in a cohort of 15,250 pregnant women. Hepatology 31:751–755. doi: 10.1002/hep.510310328. [DOI] [PubMed] [Google Scholar]
  • 170.Nie QH, Gao LH, Cheng YQ, Huang XF, Zhang YF, Luo XD, Wang JQ, Wang YY. 2012. Hepatitis C virus infection of human cytotrophoblasts cultured in vitro. J Med Virol 84:1586–1592. doi: 10.1002/jmv.23380. [DOI] [PubMed] [Google Scholar]
  • 171.Azzari C, Moriondo M, Indolfi G, Betti L, Gambineri E, de Martino M, Resti M. 2008. Higher risk of hepatitis C virus perinatal transmission from drug user mothers is mediated by peripheral blood mononuclear cell infection. J Med Virol 80:65–71. doi: 10.1002/jmv.21023. [DOI] [PubMed] [Google Scholar]
  • 172.Babik JM, Cohan D, Monto A, Hartigan-O'Connor DJ, McCune JM. 2011. The human fetal immune response to hepatitis C virus exposure in utero. J Infect Dis 203:196–206. doi: 10.1093/infdis/jiq044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Smith BD, Morgan RL, Beckett GA, Falck-Ytter Y, Holtzman D, Teo CG, Jewett A, Baack B, Rein DB, Patel N, Alter M, Yartel A, Ward JW, CDC. 2012. Recommendations for the identification of chronic hepatitis C virus infection among persons born during 1945–1965. MMWR Recomm Rep 61:1–32. [PubMed] [Google Scholar]
  • 174.Apolinario A, Majano PL, Lorente R, Nunez O, Clemente G, Garcia-Monzon C. 2005. Gene expression profile of T-cell-specific chemokines in human hepatocyte-derived cells: evidence for a synergistic inducer effect of cytokines and hepatitis C virus proteins. J Viral Hepat 12:27–37. doi: 10.1111/j.1365-2893.2005.00540.x. [DOI] [PubMed] [Google Scholar]
  • 175.Zekri AR, Bahnassy AA, Mohamed WS, Alam El-Din HM, Shousha HI, Zayed N, Eldahshan DH, Abdel-Aziz AO. 2013. Dynamic interplay between CXCL levels in chronic hepatitis C patients treated by interferon. Virol J 10:218. doi: 10.1186/1743-422X-10-218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Aroucha DC, do Carmo RF, Moura P, Silva JL, Vasconcelos LR, Cavalcanti MS, Muniz MT, Aroucha ML, Siqueira ER, Cahu GG, Pereira LM, Coelho MR. 2013. High tumor necrosis factor-alpha/interleukin-10 ratio is associated with hepatocellular carcinoma in patients with chronic hepatitis C. Cytokine 62:421–425. doi: 10.1016/j.cyto.2013.03.024. [DOI] [PubMed] [Google Scholar]
  • 177.Knobler H, Schattner A. 2005. TNF-alpha, chronic hepatitis C and diabetes: a novel triad. QJM 98:1–6. doi: 10.1093/qjmed/hci001. [DOI] [PubMed] [Google Scholar]
  • 178.Zampino R, Marrone A, Restivo L, Guerrera B, Sellitto A, Rinaldi L, Romano C, Adinolfi LE. 2013. Chronic HCV infection and inflammation: clinical impact on hepatic and extra-hepatic manifestations. World J Hepatol 5:528–540. doi: 10.4254/wjh.v5.i10.528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Jonsson JR, Barrie HD, O'Rourke P, Clouston AD, Powell EE. 2008. Obesity and steatosis influence serum and hepatic inflammatory markers in chronic hepatitis C. Hepatology 48:80–87. doi: 10.1002/hep.22311. [DOI] [PubMed] [Google Scholar]
  • 180.Falasca K, Ucciferri C, Dalessandro M, Zingariello P, Mancino P, Petrarca C, Pizzigallo E, Conti P, Vecchiet J. 2006. Cytokine patterns correlate with liver damage in patients with chronic hepatitis B and C. Ann Clin Lab Sci 36:144–150. [PubMed] [Google Scholar]
  • 181.Costantini S, Capone F, Guerriero E, Maio P, Colonna G, Castello G. 2010. Serum cytokine levels as putative prognostic markers in the progression of chronic HCV hepatitis to cirrhosis. Eur Cytokine Netw 21:251–256. doi: 10.1684/ecn.2010.0214. [DOI] [PubMed] [Google Scholar]
  • 182.Zeremski M, Dimova R, Brown Q, Jacobson IM, Markatou M, Talal AH. 2009. Peripheral CXCR3-associated chemokines as biomarkers of fibrosis in chronic hepatitis C virus infection. J Infect Dis 200:1774–1780. doi: 10.1086/646614. [DOI] [PubMed] [Google Scholar]
  • 183.Webster WS. 1998. Teratogen update: congenital rubella. Teratology 58:13–23. doi:. [DOI] [PubMed] [Google Scholar]
  • 184.Nguyen TV, Pham VH, Abe K. 2015. Pathogenesis of congenital rubella virus infection in human fetuses: viral infection in the ciliary body could play an important role in cataractogenesis. EBioMedicine 2:59–63. doi: 10.1016/j.ebiom.2014.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Johnson RE, Hall AP. 1958. Rubella arthritis; report of cases studied by latex tests. N Engl J Med 258:743–745. doi: 10.1056/NEJM195804102581506. [DOI] [PubMed] [Google Scholar]
  • 186.Pugachev KV, Frey TK. 1998. Rubella virus induces apoptosis in culture cells. Virology 250:359–370. doi: 10.1006/viro.1998.9395. [DOI] [PubMed] [Google Scholar]
  • 187.Adamo P, Asis L, Silveyra P, Cuffini C, Pedranti M, Zapata M. 2004. Rubella virus does not induce apoptosis in primary human embryo fibroblast cultures: a possible way of viral persistence in congenital infection. Viral Immunol 17:87–100. doi: 10.1089/088282404322875485. [DOI] [PubMed] [Google Scholar]
  • 188.Megyeri K, Berencsi K, Halazonetis TD, Prendergast GC, Gri G, Plotkin SA, Rovera G, Gönczöl E. 1999. Involvement of a p53-dependent pathway in rubella virus-induced apoptosis. Virology 259:74–84. doi: 10.1006/viro.1999.9757. [DOI] [PubMed] [Google Scholar]
  • 189.George S, Viswanathan R, Sapkal GN. 2019. Molecular aspects of the teratogenesis of rubella virus. Biol Res 52:47. doi: 10.1186/s40659-019-0254-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Hemphill ML, Forng RY, Abernathy ES, Frey TK. 1988. Time course of virus-specific macromolecular synthesis during rubella virus infection in Vero cells. Virology 162:65–75. doi: 10.1016/0042-6822(88)90395-9. [DOI] [PubMed] [Google Scholar]
  • 191.Geyer H, Bauer M, Neumann J, Ludde A, Rennert P, Friedrich N, Claus C, Perelygina L, Mankertz A. 2016. Gene expression profiling of rubella virus infected primary endothelial cells of fetal and adult origin. Virol J 13:21. doi: 10.1186/s12985-016-0475-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Potter JE, Banatvala JE, Best JM. 1973. Interferon studies with Japanese and U.S. rubella virus strains. Br Med J 1:197–199. doi: 10.1136/bmj.1.5847.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Dhiman N, Haralambieva IH, Vierkant RA, Pankratz VS, Ryan JE, Jacobson RM, Ovsyannikova IG, Poland GA. 2010. Predominant inflammatory cytokine secretion pattern in response to two doses of live rubella vaccine in healthy vaccinees. Cytokine 50:24–29. doi: 10.1016/j.cyto.2009.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Elshafeey F, Magdi R, Hindi N, Elshebiny M, Farrag N, Mahdy S, Sabbour M, Gebril S, Nasser M, Kamel M, Amir A, Emara MM, Nabhan A. 2020. A systematic scoping review of COVID-19 during pregnancy and childbirth. Int J Gynaecol Obstet 150:47–52. doi: 10.1002/ijgo.13182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, Wang B, Xiang H, Cheng Z, Xiong Y, Zhao Y, Li Y, Wang X, Peng Z. 2020. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA 323:1061. doi: 10.1001/jama.2020.1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Yang Z, Wang M, Zhu Z, Liu Y. 30 April 2020. Coronavirus disease 2019 (COVID-19) and pregnancy: a systematic review. J Matern Fetal Neonatal Med doi: 10.1080/14767058.2020.1759541. [DOI] [PubMed] [Google Scholar]
  • 197.Qiancheng X, Jian S, Lingling P, Lei H, Xiaogan J, Weihua L, Gang Y, Shirong L, Zhen W, GuoPing X, Lei Z, Sixth Batch of Anhui Medical Team Aiding Wuhan for COVID-19. 2020. Coronavirus disease 2019 in pregnancy. Int J Infect Dis 95:376–383. doi: 10.1016/j.ijid.2020.04.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Della Gatta AN, Rizzo R, Pilu G, Simonazzi G. 2020. COVID19 during pregnancy: a systematic review of reported cases. Am J Obstet Gynecol 223:36–41. doi: 10.1016/j.ajog.2020.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Di Mascio D, Khalil A, Saccone G, Rizzo G, Buca D, Liberati M, Vecchiet J, Nappi L, Scambia G, Berghella V, D’Antonio F. 2020. Outcome of coronavirus spectrum infections (SARS, MERS, COVID 1–19) during pregnancy: a systematic review and meta-analysis. Am J Obstet Gynecol MFM 2:100107. doi: 10.1016/j.ajogmf.2020.100107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Panahi L, Amiri M, Pouy S. 2020. Risks of novel coronavirus disease (COVID-19) in pregnancy; a narrative review. Arch Acad Emerg Med 8:e34. [PMC free article] [PubMed] [Google Scholar]
  • 201.CDC COVID-19 Response Team. 2020. Severe outcomes among patients with coronavirus disease 2019 (COVID-19)—United States, February 12-March 16, 2020. MMWR Morb Mortal Wkly Rep 69:343–346. doi: 10.15585/mmwr.mm6912e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Garg S, Kim L, Whitaker M, O'Halloran A, Cummings C, Holstein R, Prill M, Chai SJ, Kirley PD, Alden NB, Kawasaki B, Yousey-Hindes K, Niccolai L, Anderson EJ, Openo KP, Weigel A, Monroe ML, Ryan P, Henderson J, Kim S, Como-Sabetti K, Lynfield R, Sosin D, Torres S, Muse A, Bennett NM, Billing L, Sutton M, West N, Schaffner W, Talbot HK, Aquino C, George A, Budd A, Brammer L, Langley G, Hall AJ, Fry A. 2020. Hospitalization rates and characteristics of patients hospitalized with laboratory-confirmed coronavirus disease 2019–COVID-NET, 14 States, March 1–30, 2020. MMWR Morb Mortal Wkly Rep 69:458–464. doi: 10.15585/mmwr.mm6915e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, Cheng Z, Yu T, Xia J, Wei Y, Wu W, Xie X, Yin W, Li H, Liu M, Xiao Y, Gao H, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B. 2020. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395:497–506. doi: 10.1016/S0140-6736(20)30183-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ, HLH Across Speciality Collaboration UK. 2020. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 395:1033–1034. doi: 10.1016/S0140-6736(20)30628-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Yan J, Guo J, Fan C, Juan J, Yu X, Li J, Feng L, Li C, Chen H, Qiao Y, Lei D, Wang C, Xiong G, Xiao F, He W, Pang Q, Hu X, Wang S, Chen D, Zhang Y, Poon LC, Yang H. 2020. Coronavirus disease 2019 (COVID-19) in pregnant women: a report based on 116 cases. Am J Obstet Gynecol 223:111.e1–111.e14. doi: 10.1016/j.ajog.2020.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Karami P, Naghavi M, Feyzi A, Aghamohammadi M, Novin MS, Mobaien A, Qorbanisani M, Karami A, Norooznezhad AH. 2020. Mortality of a pregnant patient diagnosed with COVID-19: a case report with clinical, radiological, and histopathological findings. Travel Med Infect Dis 2020:101665. doi: 10.1016/j.tmaid.2020.101665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Mullins E, Evans D, Viner RM, O'Brien P, Morris E. 2020. Coronavirus in pregnancy and delivery: rapid review. Ultrasound Obstet Gynecol 55:586–592. doi: 10.1002/uog.22014. [DOI] [PubMed] [Google Scholar]
  • 208.Levy A, Yagil Y, Bursztyn M, Barkalifa R, Scharf S, Yagil C. 2008. ACE2 expression and activity are enhanced during pregnancy. Am J Physiol Regul Integr Comp Physiol 295:R1953–R1961. doi: 10.1152/ajpregu.90592.2008. [DOI] [PubMed] [Google Scholar]
  • 209.Hirano T, Murakami M. 2020. COVID-19: a new virus, but a familiar receptor and cytokine release syndrome. Immunity 52:731–733. doi: 10.1016/j.immuni.2020.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Wang W, Xu Y, Gao R, Lu R, Han K, Wu G, Tan W. 2020. Detection of SARS-CoV-2 in different types of clinical specimens. JAMA 323:1843–1844. doi: 10.1001/jama.2020.3786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Schwartz DA. 17 March 2020. An analysis of 38 pregnant women with COVID-19, their newborn infants, and maternal-fetal transmission of SARS-CoV-2: maternal coronavirus infections and pregnancy outcomes. Arch Pathol Lab Med doi: 10.5858/arpa.2020-0901-SA. [DOI] [PubMed] [Google Scholar]
  • 212.Alzamora MC, Paredes T, Caceres D, Webb CM, Valdez LM, La Rosa M. 2020. Severe COVID-19 during pregnancy and possible vertical transmission. Am J Perinatol 37:861–865. doi: 10.1055/s-0040-1710050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Zeng H, Xu C, Fan J, Tang Y, Deng Q, Zhang W, Long X. 2020. Antibodies in infants born to mothers with COVID-19 pneumonia. JAMA 323:1848–1849. doi: 10.1001/jama.2020.4861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Dong L, Tian J, He S, Zhu C, Wang J, Liu C, Yang J. 2020. Possible vertical transmission of SARS-CoV-2 from an infected mother to her newborn. JAMA 323:1846–1848. doi: 10.1001/jama.2020.4621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Zeng L, Xia S, Yuan W, Yan K, Xiao F, Shao J, Zhou W. 2020. Neonatal early-onset infection with SARS-CoV-2 in 33 neonates born to mothers with COVID-19 in Wuhan, China. JAMA Pediatr 174:722. doi: 10.1001/jamapediatrics.2020.0878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Kimberlin DW, Stagno S. 2020. Can SARS-CoV-2 infection be acquired in utero? More definitive evidence is needed. JAMA 323:1788–1789. doi: 10.1001/jama.2020.4868. [DOI] [PubMed] [Google Scholar]
  • 217.de Wit E, van Doremalen N, Falzarano D, Munster VJ. 2016. SARS and MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol 14:523–534. doi: 10.1038/nrmicro.2016.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Ortiz-Martinez Y. 2020. Tocilizumab: a new opportunity in the possible therapeutic arsenal against COVID-19. Travel Med Infect Dis 37:101678. doi: 10.1016/j.tmaid.2020.101678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Zhang C, Wu Z, Li JW, Zhao H, Wang GQ. 2020. The cytokine release syndrome (CRS) of severe COVID-19 and Interleukin-6 receptor (IL-6R) antagonist tocilizumab may be the key to reduce the mortality. Int J Antimicrob Agents 55:105954. doi: 10.1016/j.ijantimicag.2020.105954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Ruan Q, Yang K, Wang W, Jiang L, Song J. 2020. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med 46:846–848. doi: 10.1007/s00134-020-05991-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Chen H, Guo J, Wang C, Luo F, Yu X, Zhang W, Li J, Zhao D, Xu D, Gong Q, Liao J, Yang H, Hou W, Zhang Y. 2020. Clinical characteristics and intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a retrospective review of medical records. Lancet 395:809–815. doi: 10.1016/S0140-6736(20)30360-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Liu W, Li H. 2020. COVID-19: attacks the 1-beta chain of hemoglobin and captures the porphyrin to inhibit heme metabolism. ChemRxiv doi: 10.26434/chemrxiv.11938173.v9. [DOI]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES