Skip to main content
Reproductive Sciences logoLink to Reproductive Sciences
. 2014 Dec;21(12):1434–1451. doi: 10.1177/1933719114537720

Influenza, Immune System, and Pregnancy

Renju S Raj 1, Elizabeth A Bonney 1, Mark Phillippe 2,
PMCID: PMC4231130  PMID: 24899469

Abstract

Influenza is a major health problem worldwide. Both seasonal influenza and pandemics take a major toll on the health and economy of our country. The present review focuses on the virology and complex immunology of this RNA virus in general and in relation to pregnancy. The goal is to attempt to explain the increased morbidity and mortality seen in infection during pregnancy. We discuss elements of innate and adaptive immunity as well as placental cellular responses to infection. In addition, we delineate findings in animal models as well as human disease. Increased knowledge of maternal and fetal immunologic responses to influenza is needed. However, enhanced understanding of nonimmune, pregnancy-specific factors influencing direct interaction of the virus with host cells is also important for the development of more effective prevention and treatment options in the future.

Keywords: influenza, pregnancy, innate immunity, adaptive immunity

Introduction

Influenza epidemics and pandemics pose a serious health problem worldwide. Understanding the illness and its effects on the immune system will aid in better disease management, including effective prevention and improved treatment strategies. The fact that certain segments of the population are more susceptible to severe infections and experience increased morbidity and mortality adds to the complexity of the disease. The goal of this review is to explore literature on the influenza virus and its immunology in humans and animals in an attempt to understand the increased morbidity and mortality rates associated with influenza during pregnancy.

Epidemiology

Viral Characteristics

Influenza virus is a single-stranded RNA (ssRNA) virus belonging to the Orthomyxoviridae family.1 There are 3 major types of influenza virus—types A, B, and C—of which types A and B are responsible for most of the infections globally observed in humans. The genome for the influenza A virus (IAV) is composed of 8 negative-sense RNA strands that encode at least 13 viral genes. Of these, 2 major surface glycoproteins, hemagglutinin (HA; with 16 different subtypes) and neuraminidase (NA; with 9 different subtypes), are the major antigenic sites for the influenza virus.2,3 The main natural reservoir for influenza viruses is the intestinal tract of aquatic birds and possibly the source of all human pandemic IAV strains.4 Before the 2009 pandemic, there were 2 IAV subtypes prevalent (eg, the seasonal H1N1 and the H3N2 viruses) and 1 influenza B virus responsible for annual epidemics. In contrast, the influenza type C is antigenically stable and causes only mild illness in immunocompetent individuals.

Pandemics occur when an influenza virus strain enters the population and is antigenically different from seasonal virus strains. The generation of antigenically novel viruses occurs through point mutations in genes encoding HA and NA (antigenic drift) or through viral genome reassortment of subtypes (antigenic shift). The reassortment events occur during interspecies transmission followed by the introduction of the novel influenza strains into the human population.3,5 The H1N1 strain of IAV, which caused the pandemic in 2009, contained genetic elements from human, avian, and swine viruses.6 In contrast, the influenza pandemic of 1918 (Spanish flu) was attributed to a highly pathogenic strain of IAV. This H1N1 strain is thought to have spread from birds to humans after undergoing point mutations resulting in genomic adaptation.7 The other 20th century pandemics occurred because of viral reassortment events: the 1957 pandemic (Asian flu) was caused by the generation of an H2N2 viral strain and the 1967 pandemic (Hong Kong flu) was caused by the generation of an H3N2 strain.5,8 During the last decade, observed outbreaks of localized influenza infections mainly in Southeast Asia involved novel influenza viral strains. A highly pathogenic H5N1 influenza virus, first identified in 1997, is typically transmitted directly from birds (especially chickens) to humans; however, there is concern that with further adaptation, infection with this virus could evolve into a major pandemic.9 More recently, the emergence of an H7N9 virus in 2013 raised public health concerns around the pandemic potential of this highly virulent strain of influenza virus.10

Influenza Infection and Pregnancy

Although the 2009 H1N1 IAV has generally been characterized as a self-limited, uncomplicated infection, severe illnesses and deaths have been reported among several high-risk patient groups. Groups at risk of seasonal influenza complications are age specific (individuals less than 2 and more than 65 years of age) and characterized by individuals with chronic illnesses (eg, chronic lung disease, neurological disorders, and diabetes).11,12 Reports indicate that pregnant women with severe influenza infection are also at increased risk of having pregnancy-related complications.1316 An analysis of the pandemic 2009 IAV epidemic among pregnant women in the United States showed a disproportionately high risk of mortality in this group. There were a total of 280 intensive care unit admissions and 56 deaths among the 788 reported cases of pregnant women in the first 8 months of the pandemic. Pregnant women accounted for 5% of the deaths in the United States, although they only represented approximately 1% of the population. Pregnant women in all 3 trimesters were at increased risk of influenza-associated complications, especially when early antiviral treatment was not instituted within the first 48 hours after symptom onset. Among the deaths, 7.1% occurred in the first trimester, 26.8% in the second, and a remarkable 64.3% in the third.13 Medical reports from the 1918 influenza pandemic also noted high mortality rates in pregnant women with influenza infection.17,18 Besides adverse maternal outcomes, influenza virus infection during pregnancy is associated with preterm delivery, stillbirth, and abortion.1921

Pathogenesis

Virus–Host Interaction

Research on the interaction between virus and immune system in the host provides insight into the pathogenesis and immunogenicity of influenza. Both human and animal studies have been fruitful, suggesting that the pathogenesis of a complex disease like influenza likely involves a combination of the direct effects of the virus and an imbalance between the beneficial and the harmful effects of the mediators released by immune cells.22

The influenza virus attaches to respiratory epithelial cell surface sialic acid residues linked to surface glycoproteins through binding to the HA molecules on the viral surface. The HA protein is critical for the binding of virus and cellular receptor, fusion of the viral and cellular membranes, and subsequent endocytosis. Viral RNA replication and transcription are carried out by 3 polymerase subunits (PB2, PB1m, and PA) and nucleoprotein. Newly synthesized viral ribonucleoprotein complexes are exported from the nucleus to the cytoplasm by nuclear export protein (formerly called “NS2”) and matrix protein M1 and are assembled into virions at the plasma membrane. The NA protein facilitates the release of mature viral particles from infected cells by cleaving sialic acid bonds between cell surface glycoproteins and viral HA proteins.2,4 The resulting progeny virions then spread to adjacent cells, where the replicative cycle is repeated. Viral NA also decreases the viscosity of mucous film in the respiratory tract, exposing cell surface glycoprotein receptors and promoting the spread of virus-containing fluid to the lower respiratory tract. Through this process, within a short time, many cells in the respiratory tract become infected and are eventually killed directly by the virus. Reportedly, the most significant and life-threatening pathology after IAV infection occurs in the lower respiratory tract.23

Influenza viral infection is also known to enhance host susceptibility to secondary bacterial infections.12,2426 Viral damage to the epithelium of the respiratory tract lowers epithelial resistance to secondary bacterial invaders, especially staphylococci, streptococci, and Haemophilus influenzae. Influenza infection has also been demonstrated to enhance the production of the anti-inflammatory cytokine interleukin (IL) 10 (IL-10), thereby suppressing the overall immune response and increasing susceptibility to secondary infections.26 Pneumonia complicating influenza infections can be viral, secondary bacterial, or both12 and is attributed to loss of ciliary clearance, dysfunction of phagocytic cells, and provision of a rich bacterial growth medium by alveolar exudate. Edema and mononuclear infiltrations in response to cell death and desquamation as a result of viral replication likely account for local symptoms (cough and sore throat), whereas prominent systemic symptoms associated with influenza (eg, headache, feverishness, chills, myalgia, and malaise) probably reflect the production of cytokines. An infectious virus is very rarely recovered from blood; however, viremia may occur in severe infections or with highly pathogenic IAV strains (eg, H5N1).27

Viral Interaction With the Fetoplacental Unit

During pregnancy, there are several systems protecting the fetus. These systems include mechanical defenses (amniotic fluid, fetal membranes, the placenta, and maternal decidua) and immunological defenses (including humoral and complement systems, and cell-mediated immunity).28 The IAV has been isolated from the placenta and amniotic fluid in both fatal29,30 and nonfatal cases,31 although direct infection of the fetus has also been uncommonly reported.32 Placentitis induced by influenza virus infection has been characterized by hyperplasia and degeneration of amniotic cells, placental trophoblasts, decidual cells, and vascular endothelial cells. Viral antigens have been detected in affected cells and in lymphoid cell infiltrates.33,34 Human decidua seems to provide a more favorable environment for viral replication than placental tissues.35 These studies suggest that the influenza virus is capable of spreading from the maternal blood stream to the maternal decidua in order to replicate within the tissue and then infect the fetal chorion and amnion. Other studies have suggested that viral infection and replication have a direct cytopathic effect by inducing apoptosis in chorion cells, a process that may contribute to influenza-associated pregnancy loss.3639

Immune Responses to Viral Infection

Innate and adaptive immune responses form the mainstay of the host defense against microbes. The innate immune system is genetically programmed to detect the invariant features of invading microbes, conferring rapid and early protection against an infection. Neutrophils, macrophages, and dendritic cells (DCs), among others, play a primary role in the innate immune response to influenza.40 Host microbial sensors called “pattern-recognition receptors” (PRRs) recognize viral components, such as IAV ssRNA, and induce antiviral innate immune responses. The PRRs are expressed in multiple cell types, especially in innate immune cells such as DCs.4143 The DCs represent the most effective antigen-presenting cells capable of inducing robust CD4+ and CD8+ T-cell immunity in vivo, thereby regulating subsequent adaptive immune responses.40,44,45

Function of Toll-Like Receptors

Toll-like receptors function as PRRs that principally sense conserved molecular motifs called “pathogen-associated molecular patterns” (PAMPs), which are found in a variety of pathogens.46,47 In mice, 13 TLRs have been described, among which only TLRs 1 to 9 and 11 to 13 are functional but TLR10 (thought to arise from a retrovirus insertion) is not.48 Interestingly, in humans only 10 functional TLR molecules have been identified, whereas TLRs 11 to 13 appear to have been lost from the human genome (Kawai and Akira48). The TLRs can be divided into 2 groups depending on their cellular localization and respective PAMP ligand. One group includes TLR1, 2, 4 to 6, and 11, which are expressed on cell surfaces and mainly recognize microbial membrane components such as lipids, lipoproteins, and proteins; the other group comprises TLR3, 7, and 9, which are expressed within intracellular structures—such as the endoplasmic reticulum (ER), endosomes, lysosomes, and endolysosomes—where they respond to microbial nucleic acids.48 These intracellular TLRs play an important role in antiviral immunity.

Toll-like receptor 3 exclusively signals through a myeloid differentiation factor 88 (MyD88)-independent pathway that uses toll–IL-1 receptor [TIR] domain-containing adaptor-inducing interferon [IFN] β (TRIF) as an intracellular adaptor protein and uses alternate pathways that lead to the activation of transcription factors (IFN-regulatory factor [IRF] 3 [IRF3], nuclear factor kappa B (NFκ-B), and mitogen-activated protein [MAP] kinases) to induce inflammatory cytokines.48,49 The TLR3 and its signaling-associated adaptor molecule TRIF play a key role in the immune response of respiratory epithelial cells to IAV in humans.50 Moreover, the importance of TLR3 signaling in antiviral immunity has been demonstrated in a study using TLR3 knockout mice,22 which compared the time course of several parameters, including animal survival, respiratory distress, viral clearance, and inflammation in infected control wild-type versus TLR3-deficient mice. The TLR3-deficient mice displayed significantly reduced inflammatory mediators as well as fewer CD8+ T lymphocytes in the bronchoalveolar airspace.22 Polyriboinosinic–polyribocytidylic acid (poly[I: C]) is a synthetic ligand for TLR3 that plays an immunomodulating role in prophylaxis and therapy in influenza infections. This is described in more detail in the Immunotherapy section of this review.

There are several TLRs expressed on plasmacytoid DCs (pDCs), also known as “interferon-producing cells.” The pDCs recognize viral components such as genomic DNA and RNA and secrete copious amounts of IFNs, especially IFN-α.51 Myeloid differentiation factor 88 is an adapter protein in IL-1 signaling and is involved in most TLR signaling except TLR3.41,48,52 Through activation of IL-1 signalining, MyD88 promotes the downstream transcription of various proinflammatory cytokines and acute-phase reactants through NFκ-B/activating protein 1.5355 Myeloid differentiation factor 88 plays an important role during pDC recognition of ssRNA viruses, which involves TLR7.48 The endocytic location of viral RNA serves as a molecular recognition signature for RNA viruses, and the strategic localization of TLR7 within the lysosome is important in this pathway of viral detection. By rapidly producing high levels of type-1 IFNs, pDCs rarely become infected with viruses taken up through endocytic pathways, and this resistance enhances their ability to participate in antiviral immunity.56

Toll-like receptors 7 and 9 represent a structurally related subfamily that responds to nucleic acids by eliciting type-1 IFN production and are implicated in the response to HIV and influenza viral infections. Toll-like receptors 7 and 9 are exclusively sequestered in the ER in unstimulated cells and rapidly traffic to endolysosomes after ligand stimulation. This translocation is regulated by the ER-localizing protein UNC93B1.57 This is a 12 membrane-spanning protein, mutation of which has defects in cytokine production and upregulation of costimulatory molecules in response to TLR7 and 9, as well as TLR3 ligands, making them highly susceptible to viral and bacterial infection.48,58 Production of IFN-α by pDCs in TLR7-deficient mice is impaired after infection with the influenza virus.59 The TLR9 recognizes unmethylated 2′-deoxyribo cytidine-phosphate-guanosine (CpG) DNA motifs present in bacterial and viral DNA but are rare in mammalian cells. Synthetic CpG oligodeoxynucleotides function as TLR9 ligands directly activating DCs, macrophages, and B cells, thus driving strong T helper 1 (Th1) responses.48,49

Additionally, epithelial cells can express intracellular RNA helicases that function as PRRs for actively replicating viruses and help establish antiviral immunity by triggering type-1 IFN responses.41 The influenza virus encodes the NS1 protein, which binds to double-stranded RNA and inhibits various antiviral pathways.60 However, pDCs can still secrete high levels of IFN-α, despite infection with the influenza virus (even in the presence of NS1),61,62 whereas classical DCs can only do so when NS1 is deleted.63

During pregnancy, DCs infiltrate the decidua around invading trophoblasts.64 Invading trophoblasts (ie, nonimmune syncytiotrophoblast cells) also express TLRs 3 to 6 and 9 intermittently during pregnancy. This expression is temporal rather than constant, with certain TLRs on syncytiotrophoblasts upregulated only in the third trimester.6567 Placental expression of TLRs may play a role in pathologic states including preeclampsia and preterm labor as well as infectious morbidity during pregnancy.6871

In human pregnancy, messenger RNA (mRNA) levels of TLRs 2 and 4 in the maternal neutrophils do not significantly change in the maternal circulation longitudinally during pregnancy, compared to nonpregnant controls.72 Although there is a significant reduction in the percentage of circulating pDCs compared to the nonpregnant state,73 longitudinal studies have demonstrated increases in protein levels of TLRs 1, 7, and 9 expressed by pDCs throughout pregnancy.74 It was also noted that these changes in TLR expression were associated with significantly elevated expression of IL-6 and IL-12 and tumor necrosis factor (TNF) α, of which only IL-12 remained elevated in the postpartum period. Moreover, the altered pDC phenotype during pregnancy includes increased expression of activation and antigen-presenting molecules and paradoxically, inhibitory ligands.73 It could be hypothesized that altered pDC phenotype and its decreased circulating numbers, as opposed to alterations in other inflammatory cells, may affect systemic viral clearance and the exaggerated inflammatory responses to infection during pregnancy. This pregnancy-specific phenomenon needs confirmation in future studies.

Cytokines as the Initial Line of Defense

Influenza infection initially triggers an array of host immune responses in an attempt to limit virus replication and spread. Acute-phase response is defined as the dose-dependent behavioral and physiological response of host organisms to infections and is regarded as a nonspecific host defense response.75 The acute-phase response includes fevers of different magnitudes, changes in food and water intake in mice as well as decreases in activity and body temperature, which again have been correlated to cytokine activity after influenza virus infection.75

On recognition of viral components, PRRs initiate production of a variety of cytokines, mainly type-1 INFs (IFN-β and -α), and induce innate and adaptive immune responses.40,42,43,46,76,77 It has been shown that type-3 IFNs, which include IFN-λ, contribute to the initial immune response to a viral respiratory infection.78 Type-1 IFNs induce maturation of DCs by increasing expression of costimulatory molecules such as CD80, CD86, and CD40 as well as antigen presentation through major histocompatibility complex (MHC) class I.43 Type-1 IFNs also mediate induction of chemokines that cause stimulation and recruitment of lymphocytes and monocytes to inflamed sites. In addition, type-1 IFNs upregulate effector molecules that directly influence protein synthesis and cell growth and survival in the process of establishing an antiviral response. Both IRF3 and IRF7 in particular are activated in response to viral infection and are primarily involved in inducing type-1 IFNs.79 Apart from respiratory epithelial cells, human peripheral blood mononucleated cells (PBMCs) also produce type-1 and -3 IFNs in response to viral infection.78 It has been reported that PBMCs from nonvaccinated pregnant women have an attenuated antiviral immune response as evidenced by the production of significantly less IFN-α and -λ compared with nonpregnant women.80

Early in the infection, low levels of TNF-α may mediate inflammation through limiting viral replication and direct virus-induced injury. As the infection resolves, TNF-α may also support the tissue repair process, stimulating the growth of fibroblasts and endothelial cells. If viral replication is not inhibited at an early stage, inflammation and infection become widespread, resulting in significant tissue damage.81,82 When IAV infection progresses to pneumonia, TNF-α can enhance pulmonary inflammation without playing a significant antiviral role.83 Dysregulation of these innate immune responses involving TNF-α has also been linked to severe influenza infection in the pediatric population.84

Natural Killer Cells

Natural killer (NK) cells are a key component of innate immunity. The NK cells have both cytotoxic and cytokine-producing functions controlled by a complex panel of activating and inhibitory receptors. These receptors play a central role in the regulation of NK cell function.85 The NK cells may play a more significant role than activated effector CD8+ T lymphocytes in controlling viral burden when the host is infected with a new influenza subtype. Among patients severely ill after H1N1/09 infection, reduction in NK cells, but preservation of T lymphocytes, was observed.86 The NK cells are present in maternal decidua in large numbers where they can destroy virus-infected cells via a perforin-dependent mechanism, leading to apoptosis induction.36

Role of Adaptive Immunity

Adaptive immune responses to influenza infection can be homotypic and heterosubtypic based on the mechanism and the cell types involved. In what is called “homotypic immunity,” protection against influenza infection may arise from previous exposure to influenza of the same serotype and is dependent on circulating neutralizing antibodies, mainly immunoglobulin (Ig) subclass G (IgG).87,88 By comparison, in heterotypic immunity, the protection against severe disease arises from previous infection with an influenza virus of a different serotype.89 Heterotypic immunity is thought to involve multiple components, including CD8+ T cells, B cells, and CD4+ T cells, which play a role individually and synergistically to provide antiviral immunity. There are variations in the level of these responses in primed individuals based on antigen-specific clonal diversity, age, and infection with other pathogens and the length of time between exposures to the influenza virus. Influenza-specific memory CD4+ T cells can act as helper cells in generating optimal B- and T-cell responses, as regulators of innate immunity, and as direct effectors of protection.90 They can support protective heterotypic responses in both early and later stages of infection. Although the role of heterotypic immunity in human influenza is controversial, recent pandemic strain outbreaks have demonstrated that seasonal vaccines can provide a degree of protection against an emergent pandemic strain, especially against severe disease. A retrospective analysis of the 1918 epidemic showed the significant impact of heterosubtypic immunity on antiviral response.91 It is likely that memory CD4+ T cells play a role in this heterosubtypic protection, and these effects merit further investigation.90

CD8+ T cells form an integral part of the adaptive immune system and assist host response to viral and specific intracellular bacterial infections. A recent study examined PBMCs from individuals naive to the 2009 pandemic H1N1 virus to assess the role of cross-reactive CD8+ T cells. The PBMCs were stimulated in vitro with a panel of live viruses and the study found that most patients exhibited cytokine-positive CD8+ cells in response to pandemic H1N1 infection.92 Cytokine-producing cells were predominantly single positive (IL-2, IFN-γ, or TNF-α), and polyfunctional triple cytokine-producing cells are relatively rare. These findings suggested that these cells were functionally limited, possibly because the patients had been recently exposed to either seasonal or pandemic influenza strains. The majority of circulating memory T cells specific for influenza were in a quiescent state with restricted functionality in the absence of a prior priming effect.92 Recently, a study of the in vitro effects of H1N1/09 infection on PBMCs collected from healthy nonpregnant and pregnant women suggested that pregnancy may enhance phenotypic and functional exhaustion in CD8+ T cells following H1N1/09 infection.73

A study of a cohort of pregnant patients infected with the 2009 H1N1 pandemic influenza virus showed that there were significantly lower levels of Igs of the G2 subclass in the infected group compared to healthy pregnant controls.93 This infection-related reduction in IgG2 level in pregnancy may explain the increased severity of H1N1 infection in some but not all pregnant patients with infection. These findings were confirmed in another recently published study from China, where the pregnant H1N1-infected population was shown to have an imbalance of anti-H1N1 Ig subclasses (IgG1, IgG2, IgG3, and IgG4) and dysregulated cytokines, when compared to nonpregnant as well as uninfected pregnant controls.94 The anti-H1N1 IgG2 constituent proportion was decreased and IgG4 was increased in the pregnant H1N1 group. This was also associated with a reduction in CD4+ and CD8+ T-cell percentages and a low CD4+–CD8+ ratio in the same population. Both IL-10 and IFN-γ were noted to be significantly elevated in the infected pregnant group compared to pregnant controls and thus thought to potentially contribute to the increased severity of infection in this population.

Immune Responses at the Maternal–Fetal Interface

Cytokines play an important role in the regulation of intrauterine functions, including parturition and defense against various infections. They regulate the production of prostaglandins and matrix metalloproteinases, which are postulated to facilitate parturition by increasing myometrial contraction and collagen degradation in fetal membranes.95 A current thinking suggests that the maternal–fetal interface is in a controlled state of inflammation early during implantation and later during parturition.9698 Consistent with this model is a recent longitudinal study where the measured peripheral blood cytokine levels during pregnancy, compared with postpartum, showed significant pregnancy-related alterations in proinflammatory and chemotactic cytokines, including IFN-γ (↓), TNF-α (↑), granulocyte colony-stimulating factor (G-CSF; ↑), and monocyte chemotactic protein 1 (MCP-1; ↓). These changes were especially dramatic in the second and third trimesters.99 Thus, pregnancy may enhance systemic inflammatory response to influenza infection.

Viremia can result from IAV infection during pregnancy, leading to decidual and placental infection.29,31,100,101 The preferred environment for viral replication is human decidua (vs placental tissue), and from there, the virus spreads to fetal membranes35,36 and is likely the basis of adverse pregnancy outcomes related to IAV infection.37 The IAV infection induces apoptosis and gene expression of proinflammatory cytokines IL-1β, IL-6, and TNF-α in human fetal membranes in culture and may facilitate premature rupture of membranes by collapsing the amniotic epithelial cell layer.102 In vitro IAV infection of chorion cells induces apoptotic cell death and mRNA expression of proinflammatory cytokines such as IL-1β, IL-6, TNF-α, IFN-β and -γ, and GM-CSF. The IAV affects cultured chorion cells from human fetal membrane tissues directly both cytopathically (such as detachment and cell rounding) and by cellular degradation (eg, oligonucleosomal DNA fragmentation and lactate dehydrogenase leakage in chorion cells), which are characteristics of cells undergoing apoptosis.37,103 These apoptotic effects were not seen in infected, cultured amnion cells, which in turn induces persistent infection.37 Synthesis of specific viral macromolecules at an early stage of infection plays a critical role in inducing apoptosis.104,105 Further, it was apoptotic chorionic cells, not amnion cells, that induced secretion of bioactive IL-6 and TNF-α in vitro.106 Thus, IAV infection of the fetal chorionic membrane results in a cellular proinflammatory cytokine response with associated apoptosis.

As with choriodecidual cells, macrophages also significantly participate in local innate inflammatory response to influenza infection. Human monocytic leukemia THP-1 cells differentiate into mature macrophages with adhesion and phagocytic abilities when the cells were incubated with heat-treated culture supernatants of influenza virus-infected chorion cells undergoing apoptosis.38 Further studies suggested that these apoptotic chorion cells secreted heat-stable monocyte differentiation-inducing (MDI) factors which potentially induce the differentiation of maternal monocytes in decidua tissue into well-matured macrophages.39 These activated macrophages phagocytize influenza-infected apoptotic cells in a process mediated by a scavenger receptor (scavenger receptor A [SR-A]) on the cell surface of macrophages, which identifies cells undergoing apoptosis. Treatment with MDI factors induced SR-A mRNA expression in THP-1 cells.

Following macrophage phagocytosis, there is an abrupt onset of superoxide production known as “oxidative burst,” catalyzed by a reduced nicotinamide adenine dinucleotide phosphate oxidase enzyme complex.107 The controlled production of superoxide anion by macrophages is known to be necessary for remodeling tissues damaged by infectious agents, and excessive production may be implicated in the pathogenic effects of influenza virus infection. The MDI factors seem to play a critical role in the molecular pathogenesis of influenza virus infection through differentiation induction of monocytes into macrophages capable of phagocytosis and superoxide anion production. Studies have tried to further characterize MDI factors.39 Interleukin 6 has been shown to induce the differentiation of human monocytic leukemia cell lines, including THP-1 cells, into macrophages contributing to MDI activity derived from influenza virus-infected chorion cells. This IL-6 effect is synergistically enhanced in combination with either TNF-α or IFN-γ. Influenza virus infection induces the secretion of MDI factors containing IL-6 from organ-cultured human amniochorion tissues, which in turn induces the differentiation of maternal decidual monocytes into mature macrophages with superoxide anion production. Interleukin 6 is partly responsible for MDI activity by associating with its receptor α- (gp80) and β-chains (gp130).39

Studies involving several known inhibitors of cytokine production suggest that the cellular oxidation process and peroxisome proliferator-activated receptor, MAP kinase, and NFκ-B regulate the induction of proinflammatory cytokine gene expression in fetal membranes during infection.108110 This raises the possibility that balanced levels of proinflammatory (IL-1β, IL-6, 8, and TNF-α) and anti-inflammatory (IL-10 and activin A) cytokines are needed to control various intrauterine functions in infectious states, including influenza, during pregnancy.36

Fetal membrane tissues produce specific chemotactic cytokines (eg, IL-8, MCP-1, IFN-γ inducible protein 10 [IP-10; IFN-γ inducible protein], and regulated upon activation and normal T-cell expressed and secreted [RANTES]) upon infection with IAV.111,112 Elevated mRNA expression of chemotactic cytokines (such as macrophage inflammatory protein 1β [MIP-1β] and pulmonary and activation-regulated chemokine [PARC]) are present in the amnion and chorion tissues of patients with chorioamnionitis after preterm labor as compared with nonchorioamnionitis patients.113 During infection, chemokines may prompt monocyte migration from maternal blood to fetal chorioamnionic tissue.

Other Factors Affecting the Immune Response to Influenza Infection

Adipocytes secrete factors called “adipokines” that can significantly alter inflammatory cell and immune function. Adiponectin is an adipokine that reduces macrophage activity and proinflammatory cytokine production114 and thus may alter the inflammatory response to IAV. Adiponectin plasma levels and their correlation with H1N1 infection have been studied. Individuals with low adiponectin level (which include those who are obese, pregnant, or have metabolic syndrome) are more likely to mount a more pronounced innate immune response when infected with H1N1.115 Proinflammatory mediators are more elevated in groups with decreased adiponectin levels, likely resulting in more severe disease.

Stress and depression predict exaggerated responses to biological challenges in humans and animals.116118 Studies on the macrophage migration inhibitory factor (MIF) levels in pregnant women vaccinated against the influenza virus link depression and sensitization of the inflammatory immune response.119 In these studies, depressive symptoms predicted exaggerated MIF production following vaccination. Thus, depression may enhance the pathogenic effect of IAV infection during pregnancy, and modulation of depression or its biological mediators could present a novel therapeutic avenue.

Animal Studies

Many animal models have been used to study different aspects of mammalian influenza infection including mice, cotton rats, Syrian hamsters, guinea pigs, ferrets, dogs, cats, domestic swine, and nonhuman primates such as rhesus, pigtailed, and cynomolgus macaques, and most recently marmosets.120125 Studies on animal models of influenza infection in pregnancy are limited. Here, we focus on studies in mice and nonhuman primate (cynomolgus macaque).

Studies With Recombinant Influenza Virus

Studies conducted with reconstructed 1918 (H1N1) influenza virus (r1918) examined the virulence and pathogenesis of the virus responsible for the pandemic.126128 BALB/c mice infected with r1918 had more severe weight loss, an earlier mean time of death, and viral titers of lung tissue 10-fold higher on days 1 and 3 compared to mice infected with other viral strains.129 Gene expression studies confirmed that the virulence correlated with the early and increased expression of immune response-related genes in r1918 infection. Characterizing the functional consequences of this gene expression data by pathway analysis showed the most significant activation of death receptor, IL-6, IF-1, and TLR response pathways. There was also r1918 infection-induced expression of mRNAs for Fas and caspase 8 and -9, which play a central role in apoptosis induction through the death receptor and mitochondrial apoptosis pathways.129 Increased mortality of r1918-infected mice was accompanied by increased (more than 200-fold) viral replication, a greater influx of neutrophils into the lungs, an increased number of alveolar macrophages, and increased protein expression of cytokines and chemokines in lung tissues compared to controls.130 Human influenza H1N1 virus with 1918 HA and NA glycoproteins can induce severe lung inflammatory infiltrates consisting of alveolar macrophages and neutrophils, which play a role in controlling the replication and spread of the r1918 virus after intranasal infection of mice.

The pathogenic potential of the r1918 virus in primates was demonstrated by studies of respiratory infection in a cynomolgus macaque model that resulted in acute respiratory distress and a fatal outcome.131 The animals demonstrated substantial increase in the serum levels of IL-6 and IL-8, MCP-1 (CC chemokine ligand 2), and RANTES, with no significant changes in IL-2 or -4, IFN-γ, or TNF-α. This study supports the theory that atypical host innate immune responses characterized by dysregulation of the antiviral response (which is insufficient for protection) may contribute to lethality. The ability of influenza viruses to modulate host immune responses was also found to be applicable in cases of infection with H5N1 strains, demonstrating that this may be a shared feature among highly pathogenic influenza viruses. Marked elevation in chemokines and proinflammatory cytokines (including IL-6, TNF-α, and MCP-1) is referred to as a “cytokine storm” and hypothesized as the main cause of mortality in H5N1 influenza viral infections.132138 Mice deficient in these important cytokines succumb to infection with H5N1 virus as do wild-type mice treated with glucocorticoids for cytokine suppression, which indicates that cytokine inhibition alone cannot sufficiently reduce the morbidity and mortality of a highly pathogenic H5N1 infection.139

Role of Cytokines/Chemokines and Their Receptors

Influenza A virus replicates in lung epithelial cells and leukocytes, resulting in the production of chemokines and cytokines that favor the recruitment of mononuclear cells to the site of infection. Influenza infection leads to the synthesis of major inflammatory mediators, including IL-6, G-CSF, IL-12p40/p70, MCPs, MIPs, and RANTES. Interleukin 6 exhibits multifunctional activities that are largely proinflammatory and their release has been correlated with the same clinical symptoms and signs related to influenza in mice.75 Interferon γ inducible protein is a chemokine that is being studied for its role in the severity of lung damage after IAV infection. BALB/c mice infected with H5N1 after administration of IP-10 presented with more fulminant and necrotizing diffuse alveolar and bronchiolar damage with lymphocyte infiltration compared to infected mice without IP-10 administration.140 This chemokine needs further research in IAV infection in pregnancy for its potential role in worsening the lung pathology and thereby increasing the severity.

Chemokine receptor (CCR) 5 and CCR2 (CCRs for MIP-1α and MCP-1, respectively) are both expressed on activated macrophages and T cells, and their role in immune response against IAV has been studied.141 When infected with IAV, CCR5-deficient mice displayed increased mortality rates associated with acute, severe pneumonitis, and enhanced expression of MCP-1 and RANTES. In contrast, CCR2-deficient mice were protected from early pathological manifestations of influenza possibly because of defective macrophage recruitment. Chemokine receptor 2 deficiency also resulted in high pulmonary viral titers early in infection in CCR2-deficient mice which correlated with a relative delay in T-cell migration. Mice deficient in both CCR2 and MIP-1α paradoxically had the best survival and the highest viral titers among all groups in the study.141 Absence of CCR2 blocks most of the early pulmonary macrophage accumulation, whereas MIP-1α has been shown to be a major regulator of T-cell trafficking into lymph nodes and specific tissues in response to a variety of antigenic stimuli, including IAV.142,143 These studies suggest that MCP-1 and MIP-1α make distinct and additive contributions to the pulmonary inflammation induced by influenza virus infection.

As in humans, mice are highly susceptible to secondary bacterial infections after recovery from airway infection from an influenza virus. Mice with postinfluenza pneumonia express strikingly elevated pulmonary concentrations of IL-10 in comparison to mice with primary pneumococcal pneumonia.26 Interleukin 10 inhibits the production of proinflammatory cytokines and chemokines by potently inhibiting neutrophil functions, including degranulation.144 Thus elevated levels of IL-10 in mice with postinfluenza pneumonia may be responsible for the relatively reduced neutrophil function observed, resulting in increased susceptibility to secondary bacterial pneumonia. Therefore, therapies aimed at neutralization of IL-10 could play a role in prevention of postinfluenza pneumococcal pneumonia.26

Adaptive Immune Responses in Animal Studies

Published research on adaptive immune responses in animal models of influenza infection in pregnancy is very limited. Some of the studies described here outside of pregnancy can provide insights into further research in pregnancy and also in the area of vaccination. In mouse studies, memory CD4+ T lymphocytes impact various stages of antiviral immune response. Studies have shown that virus-specific memory CD4+ T cells may directly regulate innate inflammation within 48 hours after heterotypic challenge, independent of PAMP recognition.145 In studies on heterosubtypic immunity in mice, clinical outcomes and histopathological changes in the lungs were compared between mice with heterosubtypic immunity and the naive group after lethal influenza virus infection.146 Weight loss up to the first 4 days after infection followed by weight gain and accelerated clearance of the virus from the lungs were observed in mice with prior exposure to the influenza virus. The reduced clinical manifestations and accelerated viral clearance are correlated with anamnestic cytotoxic T-lymphocyte (CTL) responses in mice with prior priming infection. It has also been shown that memory CTLs can reside in the lungs which would allow an immediate recall response upon secondary infection.147149

Another important role CD4+ T cells fulfill when responding to a primary influenza challenge is actively supporting the generation of long-lasting functional memory CD8+ T cells, thereby aiding the magnitude of response and function of CD8+ T-cell effectors.150,151 Interleukin 6 is critical to inducing naive and memory CD4+ T-cell IL-21 production upon T-cell receptor stimulation.152 CD4+ T-cell IL-21 production is required for IL-6 to promote B-cell antibody production in vitro. Moreover, administration of IL-6 with an inactive influenza virus enhances virus-specific antibody production. Thus, IL-6, through increased IL-21 production, promotes antibody production by supporting the B-cell helper capabilities of CD4+ T cells.152

CD8+ CTLs are important effectors responsible for the clearance of viral infections and consequently a valuable population to induce by vaccination. It was noted that proinflammatory cytokine production by effector CD8+ T cells was largely restricted by T cells localized to the pulmonary interstitium in mice with sublethal influenza virus infection.153 T-cell immunity induced after infection with human IAV may be cross-reactive with avian influenza viruses and could provide some degree of heterotypic protection against these highly pathogenic viruses by involving these virus-specific CD8+ CTLs. Induction of these cross-reactive CTLs may aid in the development of universal vaccines against viral infections.146

Although CD8+ T cells are associated with protective responses to IAV infection, it is becoming increasingly evident that they can also be associated with the development of influenza-related immunopathological sequelae.154 A dysregulated CD8+ T cell response to IAV infection might lead to exacerbation of lung pathology and sustained lung injury.155 This TLR3-mediated T-cell response has been studied in TLR3-deficient mice, where the excessive IFN-γ recovered from lungs was thought to contribute to a decrease in CD8+ T-cell number and limit T cell-mediated inflammation compared to wild type.22 Prolonged survival and also increased pulmonary IAV load seen in TLR3-deficient mice after IAV infection were also attributed to this negative feedback regulation of CD8+ T cells by IFN-γ. Dysregulation of CD8+ T cells was also noted in studies of mouse pregnancy complicated by influenza infection. Such dysregulation was thought to be related to altered lung DC function and impaired IFN signaling.156 It was further hypothesized that estrogen may play a role in this process.

Influenza viruses with HA of the H2 subtype are potent inducers of T cell-independent murine B-cell proliferation, and induction of this response is affected by the expression of cell surface I-E molecules (class II MHC glycoprotein) on B cells.157 The inactivated H2-bearing influenza virus and purified HA of the H2 subtype induce B-cell proliferation and upregulate costimulatory molecules through a MyD88-dependent pathway independent of any known TLR.158 Only members of the TLR/IL-1R receptor family have been reported to signal via MyD88-dependent pathways, thus suggesting activation of these innate responses in B cells by influenza HA may involve an unidentified TLR.158

Pregnancy Model

In one of the initial mouse models used to study the effects of IAV infection during pregnancy, infections with the WSN strain during the first and third weeks of gestation were found to have a detrimental effect on neonatal growth and development.159,160 Infections in the third week resulted in significantly increased postnatal mortality in the first 8 weeks of life, with a similar trend observed in mice born to mothers infected in the first week of gestation. C3H inbred and Prince Henry outbred pregnant mice, infected with WSN and MEL strains in the third week of gestation, exhibited a 3-fold higher maternal mortality rate when compared with mice in the first week or with nonpregnant mice.159 These findings are similar to the higher maternal mortality rates seen in human pregnancies during past influenza pandemics.15,16

The effects of wild-type and a mutant 2009 H1N1 influenza virus strain have been compared in pregnant and nonpregnant BALB/c mice.161 The mutant strain contained a glutamate–glycine substitution at position 222 on viral HA and was found more frequently in patients who had severe pandemic infection. When infected at day 12 to 14 of gestation, mice experienced higher viral titers in the lungs, more severe histologic evidence of pneumonia, and significantly increased mortality rates compared to nonpregnant mice, especially in response to infection with the mutant virus. Moreover, pregnant infected mice expressed higher levels of proinflammatory cytokines and chemokines in lung homogenates. Only IFN-γ was significantly lower in pregnant mice. In addition, CD3+/CD4+ and CD3+/CD8+ peripheral T lymphocytes and specific antiviral serum antibody counts were lower in pregnant mice, leading to the conclusion that both innate and adaptive immune responses to influenza are suppressed during pregnancy.161

A similar study recently demonstrated significantly increased maternal mortality and fetal absorptions as a result of pandemic H1N1 infection. In this study, pregnant BALB/c mice had higher viral titers and elevated levels of inflammatory cytokines and chemokines (eg, IL-1α, IL-6, G-CSF, RANTES, and MCP-1) when infected with the pandemic H1N1 virus versus seasonal H1N1.162 The study attributed fatality from pandemic H1N1 to its high pathogenic characteristics rather than a lack of preexisting immunity against the virus. Another study showed that the increased mortality rate in pregnant mice due to pandemic H1N1 infection was related to reduced regeneration of the respiratory epithelium, suggesting impaired lung repair and not related to the absolute viral burden in the nasal cavities/lungs.163 This lethal influenza infection during pregnancy was associated with elevated pulmonary chemoattracants (such as MCP-1), enhanced numbers of macrophages and neutrophils, and increased nitrite (a nitric oxide metabolite) in the lungs. These mediators were associated with decreased regeneration of the pulmonary epithelial lining, indicating an immunopathology-based mechanism of lung injury mediated by elevated cellular recruitment in severe pandemic H1N1 infection in pregnant mice.163

Another study in CD-1 mice addressed the immune system in pregnant and nonpregnant states and its contribution to increased maternal mortality in systemic illnesses, such as influenza and pyelonephritis.164 Using a mouse model of systemic and localized inflammation induced by lipopolysaccharide injection (intraperitoneal or intrauterine) derived from Escherichia coli 055: B5, it was shown that the maternal immune response appeared to be dynamic and functionally different in pregnancy compared to nonpregnant state. The response to both systemic and local inflammation during pregnancy was functionally altered with increased serum concentrations of IL-10, IL-6, IL-12, and TNF-α in the pregnant state, correlating with decreased immune reactivity and increased susceptibility to infections. Moreover, response to local infection differed according to gestational age. Significantly lower concentrations of cytokines were observed in at-term versus preterm mice.164 These results to some extent explain the increased morbidity from influenza infections during pregnancy as well as the associated risk of preterm birth.

Pregnancy Hormones and the Immune Response to Influenza

Progesterone and glucocorticoids, which increase during pregnancy, can have an anti-inflammatory effect.165 This would explain the increase in severity of infectious agents such as influenza, which require prompt inflammatory responses for the initial control and clearance of pathogens.15,166,167

In addition, elevated levels of progesterone during pregnancy can stimulate the synthesis of progesterone-induced binding factor that promotes CD4+ T cell/helper T cell type 2 (Th2) differentiation, with increased serum concentrations of Th2 cytokines, including IL-4, -5, and -10.168170 The observed promotion of Th2 responses during pregnancy corresponds with a reduction in Th1 responses both systemically and at the maternal–fetal interface in animal models as well as in humans.166,171175 Peripheral regulatory T cells (a subset of CD4+ T cells) are thought to promote immune tolerance in pregnancy by upregulating transforming growth factor β and IL-10 through hemeoxygenase 1.176178 It is thought that the apparently tolerant microenvironment of the placenta is supported by these pregnancy-specific alterations in T cell immunity. Further research into the role of regulatory T cells and T helper subset regulation specific to influenza infection during pregnancy is needed.176 Apart from Th responses, the direct role of progesterone in disease susceptibility and severity, in the context of influenza infection, also requires further investigation.

The effects of estrogens on the severity of influenza infection are more complex, wherein elevated levels imposed on nonpregnant mice are protective; in contrast, elevated levels during pregnancy are not.179 Estrogen appears to have both anti- and proinflammatory effects depending on the level expressed,156,179 and this may explain the differences in the severity observed. Additionally, estradiol, through its receptors, has been shown to activate the alveolar epithelial sodium channels promoting alveolar fluid clearance.180 This mechanism seems to be challenged during influenza infection.181,182 Influenza infection seems to induce a hypoestrogenic state that affects these sodium channels, decreasing clearance of alveolar fluid, and thereby increasing susceptibility to pneumonia.180 Thus, estrogen can affect the severity of disease through mechanisms unrelated to immune system modulation.

Behavioral Implications of Influenza Infection

Evidence from animal and human studies has indicated that influenza infection during pregnancy is a risk factor for neuropsychiatric diseases, such as schizophrenia, in offspring.183186 This has been further elucidated in a recent study conducted on rhesus monkeys, where influenza infection affected fetal neural development with a reduction in gray matter throughout the cortex and decreased white matter in the parietal cortex.187 These changes are thought to increase the likelihood of behavioral impairments later in life. The nature and extent of brain volume reductions seen in monkey bore most in common with the structural abnormalities found frequently in schizophrenia. Another interesting observation is that species differences in the extent of behavioral findings reflect the virulence of the influenza strain. The exact mechanism of abnormal brain development is not known. Proinflammatory cytokines generated by a maternal immune response transferred to amniotic fluid entering into the fetal circulation and permeating the immature blood–brain barrier and central nervous system has been implicated rather than direct exposure of the fetus to the virus.187 There is also recent evidence from a nested case–control study of a population-based birth cohort, showing a significant 4-fold increase in the risk of bipolar disorder in adult offspring when exposed to influenza in utero.188 An increase in slow-wave sleep accompanies influenza infection in certain strains of mice and has been shown to be related to the levels of the expression of the If-1 gene.189 The If-1 gene codes for high and low production of IFN, which is considered a putative sleep-inducing cytokine. If-1 has thus been identified as a genetically determined factor that elicits sleep after viral challenge.

Immunization

Vaccination

Immunization is the mainstay of influenza prophylaxis. There are 2 types of influenza vaccine: parenterally administered multivalent inactivated vaccine and intranasal live-attenuated virus vaccine.190 The split-virus vaccine is the most used inactivated influenza formulation because it induces protective levels of antibody and has few side effects.191 Whole-virus vaccine is considered more immunogenic than split virus192; however, whole-virus vaccine has been associated with adverse reactions and thus it is currently not widely used.193

The American College of Obstetricians and Gynecologists and the Centers for Disease Control and Prevention recommend that all pregnant women and women planning a pregnancy occurring during the influenza season should be vaccinated and state that the multivalent inactivated vaccination is safe in all trimesters of pregnancy.194196 The immunization of women during pregnancy with an inactivated trivalent influenza vaccine (seasonal H1N1, H3N2, and influenza B virus) has also been recommended by the World Health Organization since 2005.197 Population-based studies have not shown any adverse pregnancy effects of vaccination even with receiving the vaccine in the first trimester of pregnancy.198 A recent study showed significantly decreased low birth weight and preterm birth (both decreased by nearly 25%) in the vaccinated compared to nonvaccinated women.199 In contrast, other studies did not show any association of vaccination with spontaneous abortion, preterm birth, birth defects, or small for gestational age babies.200,201 In the recent years, significant improvements in influenza vaccination coverage among pregnant women have been observed, from 11% during the 2001 to 2002 influenza season to 38% to 50% during the 2010 to 2011 influenza season, depending on the survey tool used.202 Despite these improvements, efforts need to be directed toward identifying the key barriers to immunization and further increasing the acceptance rate of vaccination among pregnant women to reach the 80% vaccination goal established for 2020.203

In a randomized study of mothers who received the multivalent inactivated influenza vaccine, the illness caused by influenza virus decreased by 63% in infants, and the rate of respiratory illness with fever decreased by 36% in the mothers.204 A follow-up study investigated the prevalence of mothers and infants with protective antibodies postvaccination in the third trimester and noticed significant antibody titers against the influenza A subtypes used to create the vaccine in a high proportion of mothers and their newborns, with newborns protected during first 6 months of childhood.205 A study evaluating the efficiency of influenza vaccination in the second and third trimesters of pregnancy measured the Th1/Th2 balance, maintenance of antibody titers, and transplacental transfer of antibodies to the fetus after vaccination.206 It was reported that the maintenance of the level of antibody depends on the time elapsed after vaccination, and titers decreased with time irrespective of gestational age. The presence of higher antibody titers in fetal blood may be related to the mechanism of active transfer. The successful immunization rate in the study of approximately 90% was independent of both the Th1/Th2 balance and the stage of gestation. Vaccination at any gestational age yielded sufficient antibody titers to provide protection against infection, showing that influenza vaccination can be administered at any stage of pregnancy.206 In a study of a nonlethal murine challenge model, 2 doses of split-virus vaccine were found to be most effective in reducing viral shedding and were associated with high concentrations of prechallenge vaccine-induced serum IgG. Also, irrespective of IgG subclass, high concentrations of serum IgG induced by immunization were an important indicator of the efficacy of the vaccine.193

Significant protection from lethal challenge of H5N1 influenza viruses was reported in mothers and fetuses in studies conducted on pregnant mice immunized against H5N1 with 1- and 2-dose immunizations.207 Newborn mice born to immunized mothers were also protected, and the complete protective immunity lasted for as long as 4 weeks after delivery. Protective immunity against the H5N1 influenza virus in newborn mice was closely related to the presence of IgG2a antibody subtype. This study concluded that maternal vaccination was effective and critical in protecting pregnant females, their fetuses, and their newborns from highly pathogenic viruses like H5N1.207

Immunotherapy

Although still under investigation, immunotherapy may be beneficial for the prevention and treatment of influenza. It remains uncertain whether administration of Igs to IgG2-deficient patients is likely to be therapeutically beneficial, although some studies report an IgG2-subclass deficiency in H1N1 infections during pregnancy.93,94 Interestingly, convalescent blood products were administered during the 1918 Spanish influenza pandemic with a reduction in mortality.208 A vaccination strategy providing broad protection against diverse influenza isolates could eliminate the need for annual vaccines and protect against emergent pandemic strains. One approach towards this goal could involve vaccines that generate strong memory T cells against multiple internal core proteins.90 The CTL peptides (lipopeptide immunogens), although not significantly superior in inducing primary effector CD8+ T cells, do elicit a much more effective memory cell population, the recall of which may account for their superiority in inducing pulmonary protection after viral challenge, making them candidates for future vaccine incorporation.150

The use of TLR7 agonists as an adjuvant for antiviral vaccines and immunological interventions to clear existing viral infections are promising approaches in the search for new antiviral treatments.56 Synthetic TLR7/8 agonist (3M-011) has been studied as an immunomodulator in a rat model of influenza infection and was noted to be beneficial for populations at risk of influenza infection. The advantage of a small molecule (less than 400 kDa) to act locally via intranasal preparations to induce an antiviral state without causing adverse systemic effects seems to be feasible. Apart from TLR7/8 agonists, TLR3, TLR4, and TLR9 agonists also have been investigated as standalone immunomodulators in murine models of influenza infection.209 Studies in mice have revealed that poly(I: C), a synthetic TLR3 ligand, can modulate the immune responses including IFN induction and activation of NK cells. These responses provide nonspecific antiviral defense against several viral infections and may therefore provide a broad-spectrum antiviral effect against influenza viruses regardless of the strain or subtype involved.210 Caution is needed with this approach because multiple high doses of poly(I: C) administered intravenously have been found to produce toxic reactions in humans.211 In addition, poly(I: C) has been observed to stimulate preterm delivery in pregnant mice71,212; therefore, its therapeutic utility during pregnancy is uncertain.

Summary, Conclusion, and Future Directions

During pregnancy, the immune system undergoes important adaptations to accommodate the immune tolerance required by the mother to carry the allogeneic fetus; however, these adaptions induce a potentially dysfunctional response to infections, when compared to the nonpregnant state. Further, these adaptations may account for the increased maternal/fetal morbidity and mortality observed in certain infections, including influenza. The underlying mechanisms contributing to these effects are summarized in Table 1. Existing evidence suggests that there occurs complex regulation of the maternal immune system, including innate and adaptive responses. This regulation occurs at various levels (locally in the lung, systemically, and at the maternal–fetal interface) and appears to differ according to gestational age. Regarding infection, such regulation is activated at various time points after exposure and is influenced by the extent of preexisting (prepregnancy) immunity. Expanded research concentrating on the maternal–fetal interface in influenza infection will likely lead to a better understanding of the mechanisms underlying pregnancy-related complications that occur as a result of infection. Ongoing research addressing the effects of gender and sex hormones on immunity as well as the direct effects of these hormones on the growth, adaptability, and virulence of infectious agents will provide further insights into this apparent altered immune status and susceptibility to infections.

Table 1.

Influenza Infection-Related Pathology and the Immune Alterations in Pregnancy.a

Site Pathology/Sequelae Pregnancy-Specific Immune Alterations
Lung Inflammation-induced lung damage ↑ IL-6, IL-8, RANTES, IP-10
Impaired lung repair ↑ MCP-1, neutrophils, macrophages, nitrites
Secondary bacterial infection Infection induced hypoestrogenic state → decrease alveolar fluid clearance and ↑ risk for pneumonia and secondary infection
Circulation Viremia
 ↑ severity of infection ↑ Expression of TLRs 1, 3, 6, 7, 9
↑ IL-10, IFN-γ (↑ susceptibility to secondary infection)
↑ IL-6, 12
 ↓ viral clearance Altered pDC phenotype/number
↓ IFN-α, λ
Altered proinflammatory state ↓MCP-1, IFN-γ, ↑TNF-α
Progesterone and glucocorticoids induced ↑ Th2 response
Altered CD8 T-cell phenotype
↓IgG2 levels
Maternal–fetal interface Inflammation of placenta and membranes ↑ NK cell activation in maternal decidua
Labor ↑ Prostaglandin release and ↑ MMP activity
Apoptosis of chorion with rupture of membranes ↑ IL-6, 8, IL-1β, TNF-α, IFN-β, γ
↑ chemotactic cytokines (IP-10, MDI factors)
↑ MDI factors including IL-6 → recruitment of macrophages, ↑MCP-1, RANTES, MIP-1β

Abbreviations: IFN, interferon; IL, interleukin; IP-10; IFN-γ inducible protein 10; MDI, monocyte differentiation-inducing; MIP, macrophage inflammatory protein; NK, natural killer; RANTES, regulated upon activation and normal T-cell expressed and secreted; th2, helper T cell type 2; TLR, toll-like receptor; TNF, tumor necrosis factor; MCP, monocyte chemotactic protein 1; pDCs, plasmacytoid dendritic cells; Ig, immunoglobulin; MMP, matrix metalloproteinase.

aSummary of the pathology at different levels (lung, systemic circulation, and maternal fetal interface) in relation to influenza infection in pregnancy and the likely immune mechanisms contributing to these changes.

Development of better animal models to more closely replicate the morbidity and mortality observed with human infections is also necessary to advance the study of the effect of infectious diseases during human pregnancy. For example, there is evidence that host genetic variations increase susceptibility to influenza infection.213 Animal models may help define the role of genetic variability during influenza infection, especially by identifying key polymorphisms. Gene-specific animal models will facilitate identification of at-risk populations and new targets for therapeutic interventions and vaccines.214 This is an area of growing interest.

In addition to immune system regulation during pregnancy, another important aspect of increased susceptibility in a pregnant host may be pregnancy-related adaptations in other organ systems. Pregnancy-related changes in the lung may be an important example with regard to influenza infection, as it has been reported that progenitor cells present in the bronchiolar epithelium are capable of regeneration after H1N1 infection.215 Influenza immunopathogenicity during pregnancy is related to impaired lung repair in mice.163 If confirmed in humans, this could open up new avenues for stem cell therapies in acute and chronic lung injury following severe infections, facilitating faster recovery.

Although influenza infection during pregnancy can result in spontaneous abortions and preterm delivery, the fetus appears to be rarely infected directly. However, the emerging literature on schizophrenia and bipolar disorder in offspring of mothers exposed to IAV during pregnancy, and the contribution of the immune system to these indirect effects of infection, is worth further exploration.

Vaccination remains the mainstay for prevention of infection, and multivalent inactivated influenza vaccines are recommended during pregnancy irrespective of trimester. Another potentially fruitful area of research will be defining strategies to recognize the key barriers to immunization during pregnancy and improving vaccination rates in this high-risk group.

Influenza infection during pregnancy continues to be a complex and clinically important problem. Continuing efforts to understand the interactions between the mother, the virus, and the immune system, both systemically and locally, will lead to improved strategies for prevention and treatment.

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) received the following financial support for the research, authorship, and/or publication of this article: Supported by NIHK12HD063082 (all authors), NIHP20 RR021905 (EAB, through the Vermont Center for Immunology and Infectious Disease), The Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine (RSR, EAB), and the Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard School of Medicine(MP).

References

  • 1. Palese PSM. Orthomyxoviridae: the viruses and their replication. In: Knipe DM HP, ed. Fields Virology. Vol 1 Philadelphia: Lippincott, Williams & Wilkins; 2007:1647–1690. [Google Scholar]
  • 2. Neumann G, Noda T, Kawaoka Y. Emergence and pandemic potential of swine-origin H1N1 influenza virus. Nature. 2009;459 (7249):931–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Taubenberger JK, Kash JC. Influenza virus evolution, host adaptation, and pandemic formation. Cell Host Microbe. 2010;7 (6):440–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Webster RG, Bean WJ, Gorman OT, Chambers TM, Kawaoka Y. Evolution and ecology of influenza A viruses. Microbiol Rev. 1992;56 (1):152–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Khiabanian H, Trifonov V, Rabadan R. Reassortment patterns in Swine influenza viruses. PLoS Curr. 2009;1:RRN1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Trifonov V, Khiabanian H, Greenbaum B, Rabadan R. The origin of the recent swine influenza A(H1N1) virus infecting humans. Euro Surveill. 2009;14 (17). [PubMed] [Google Scholar]
  • 7. Watanabe T, Kawaoka Y. Pathogenesis of the 1918 pandemic influenza virus. PLoS Pathog. 2011;7 (1):e1001218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Scholtissek C, Rohde W, Von Hoyningen V, Rott R. On the origin of the human influenza virus subtypes H2N2 and H3N2. Virology. 1978;87 (1):13–20. [DOI] [PubMed] [Google Scholar]
  • 9. Abdel-Ghafar AN, Chotpitayasunondh T, Gao Z, et al. Update on avian influenza A (H5N1) virus infection in humans. N Engl J Med. 2008;358 (3):261–273. [DOI] [PubMed] [Google Scholar]
  • 10. Gao R, Cao B, Hu Y, et al. Human infection with a novel avian-origin influenza A (H7N9) virus. N Engl J Med. 2013;368 (20):1888–1897. [DOI] [PubMed] [Google Scholar]
  • 11. Simonsen L. The global impact of influenza on morbidity and mortality. Vaccine. 1999;17 (suppl 1):S3–S10. [DOI] [PubMed] [Google Scholar]
  • 12. Wright PF NG, Kawaoka Y. Orthomyxoviruses. In: Howley DMKaPM, ed. Fields Virology. Philadelphia: Lippincott, Williams & Wilkins; 2007:1691–1740. [Google Scholar]
  • 13. Siston AM, Rasmussen SA, Honein MA, et al. Pandemic 2009 influenza A(H1N1) virus illness among pregnant women in the United States. JAMA. 2010;303 (15):1517–1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Ellington SR, Hartman LK, Acosta M, et al. Pandemic 2009 influenza A (H1N1) in 71 critically ill pregnant women in California. Am J Obstet Gynecol. 2011;204 (6 suppl 1):S21–S30. [DOI] [PubMed] [Google Scholar]
  • 15. Jamieson DJ, Honein MA, Rasmussen SA, et al. H1N1 2009 influenza virus infection during pregnancy in the USA. Lancet. 2009;374 (9688):451–458. [DOI] [PubMed] [Google Scholar]
  • 16. Louie JK, Acosta M, Jamieson DJ, Honein MA. Severe 2009 H1N1 influenza in pregnant and postpartum women in California. N Engl J Med. 2010;362 (1):27–35. [DOI] [PubMed] [Google Scholar]
  • 17. Woolston WJ, Conley DO. Epidemic pneumonia (spanish influenza) in pregnancy: effect in one hundred and one cases. J Am Med Assoc. 1918;71 (23):1898–1899. [Google Scholar]
  • 18. Bland PB. Influenza in its relation to pregnancy and labor. Am J Obstetr Dis Women Child. 1919;79(2):184–197. [Google Scholar]
  • 19. Harris JW. Influenza occurring in pregnant women: a statistical study of thirteen hundred and fifty cases. J Am Med Assoc. 1919;72 (14):978–980. [Google Scholar]
  • 20. Hardy JM, Azarowicz EN, Mannini A, Medearis DN, Jr, , Cooke RE. The effect of Asian influenza on the outcome of pregnancy, Baltimore, 1957-1958. Am J Public Health Nations Health. 1961;51:1182–1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Stanwell-Smith R, Parker AM, Chakraverty P, Soltanpoor N, Simpson CN. Possible association of influenza A with fetal loss: investigation of a cluster of spontaneous abortions and stillbirths. Commun Dis Rep CDR Rev. 1994;4 (3):R28–R32. [PubMed] [Google Scholar]
  • 22. Le Goffic R, Balloy V, Lagranderie M, et al. Detrimental contribution of the Toll-like receptor (TLR)3 to influenza A virus-induced acute pneumonia. PLoS Pathog. 2006;2 (6):e53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Walsh JJ, Dietlein LF, Low FN, Burch GE, Mogabgab WJ. Bronchotracheal response in human influenza. Type A, Asian strain, as studied by light and electron microscopic examination of bronchoscopic biopsies. Arch Intern Med. 1961;108:376–388. [DOI] [PubMed] [Google Scholar]
  • 24. Louria DB, Blumenfeld HL, Ellis JT, Kilbourne ED, Rogers DE. Studies on influenza in the pandemic of 1957-1958. II. Pulmonary complications of influenza. J Clin Invest. 1959;38 (1 pt 2):213–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. O'Brien KL, Walters MI, Sellman J, et al. Severe pneumococcal pneumonia in previously healthy children: the role of preceding influenza infection. Clin Infect Dis. 2000;30 (5):784–789. [DOI] [PubMed] [Google Scholar]
  • 26. van der Sluijs KF, van Elden LJ, Nijhuis M, et al. IL-10 is an important mediator of the enhanced susceptibility to pneumococcal pneumonia after influenza infection. J Immunol. 2004;172 (12):7603–7609. [DOI] [PubMed] [Google Scholar]
  • 27. Brooks GF CK, Butel JS, Morse SA, Mietzner TA. Jawetz, Melnick, & Adelberg’s Medical Microbiology. 25 ed: The McGraw-Hill Companies, Inc; 2010. [Google Scholar]
  • 28. Grossman JH DL. Infections affecting the placenta. In: JP L, ed. The Human Placenta. Maryland: Aspen Publishers; 1987:131–154. [Google Scholar]
  • 29. Yawn DH, Pyeatte JC, Joseph JM, Eichler SL, Garcia-Bunuel R. Transplacental transfer of influenza virus. JAMA. 1971;216 (6):1022–1023. [PubMed] [Google Scholar]
  • 30. Jewett JF. Influenza pneumonia at term. N Engl J Med. 1974;291 (5):256–257. [DOI] [PubMed] [Google Scholar]
  • 31. McGregor JA, Burns JC, Levin MJ, Burlington B, Meiklejohn G. Transplacental passage of influenza A/Bangkok (H3N2) mimicking amniotic fluid infection syndrome. Am J Obstet Gynecol. 1984;149 (8):856–859. [DOI] [PubMed] [Google Scholar]
  • 32. Gu J, Xie Z, Gao Z, et al. H5N1 infection of the respiratory tract and beyond: a molecular pathology study. Lancet. 2007;370 (9593):1137–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Mel'nikova VF, Tsinzerling VA, Aksenov OA, Tsinzerling AV. [Chronic course of influenza with extrapulmonary involvement]. Arkh Patol. 1994;56 (1):33–38. [PubMed] [Google Scholar]
  • 34. Mel'nikova VF, Tsinzerling AV, Aksenov OA, Vydumkina SP, Kalinina NA. [Involvement of the afterbirth in influenza]. Arkh Patol. 1987;49 (9):19–25. [PubMed] [Google Scholar]
  • 35. Rosztoczy I, Sweet C, Toms GL, Smith H. Replication of influenza virus in organ cultures of human and simian urogenital tissues and human foetal tissues. Br J Exp Pathol. 1975;56 (4):322–328. [PMC free article] [PubMed] [Google Scholar]
  • 36. Uchide N, Ohyama K, Bessho T, Toyoda H. Induction of pro-inflammatory cytokine gene expression and apoptosis in human chorion cells of fetal membranes by influenza virus infection: possible implications for maintenance and interruption of pregnancy during infection. Med Sci Monit. 2005;11 (1):RA7–R16. [PubMed] [Google Scholar]
  • 37. Uchide N, Ohyama K, Bessho T, Yuan B, Yamakawa T. Apoptosis in cultured human fetal membrane cells infected with influenza virus. Biol Pharm Bull. 2002;25 (1):109–114. [DOI] [PubMed] [Google Scholar]
  • 38. Uchide N, Ohyama K, Yuan B, Bessho T, Yamakawa T. Differentiation of monocytes to macrophages induced by influenza virus-infected apoptotic cells. J Gen Virol. 2002;83 (pt 4):747–751. [DOI] [PubMed] [Google Scholar]
  • 39. Uchide N, Tadera C, Sarai H, Ohyama K, Bessho T, Toyoda H. Characterization of monocyte differentiation-inducing (MDI) factors derived from human fetal membrane chorion cells undergoing apoptosis after influenza virus infection. Int J Biochem Cell Biol. 2006;38 (11):1926–1938. [DOI] [PubMed] [Google Scholar]
  • 40. Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science. 2010;327 (5963):291–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Kawai T, Akira S. Innate immune recognition of viral infection. Nat Immunol. 2006;7 (2):131–137. [DOI] [PubMed] [Google Scholar]
  • 42. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004;5 (10):987–995. [DOI] [PubMed] [Google Scholar]
  • 43. Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I interferons (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol. 2005;23:307–336. [DOI] [PubMed] [Google Scholar]
  • 44. Sato A, Iwasaki A. Induction of antiviral immunity requires Toll-like receptor signaling in both stromal and dendritic cell compartments. Proc Natl Acad Sci U S A. 2004;101 (46):16274–16279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392 (6673):245–252. [DOI] [PubMed] [Google Scholar]
  • 46. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001;2 (8):675–680. [DOI] [PubMed] [Google Scholar]
  • 47. Janeway CA, Jr, , Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216. [DOI] [PubMed] [Google Scholar]
  • 48. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11 (5):373–384. [DOI] [PubMed] [Google Scholar]
  • 49. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124 (4):783–801. [DOI] [PubMed] [Google Scholar]
  • 50. Guillot L, Le Goffic R, Bloch S, et al. Involvement of toll-like receptor 3 in the immune response of lung epithelial cells to double-stranded RNA and influenza A virus. J Biol Chem. 2005;280 (7):5571–5580. [DOI] [PubMed] [Google Scholar]
  • 51. Perussia B, Fanning V, Trinchieri G. A leukocyte subset bearing HLA-DR antigens is responsible for in vitro alpha interferon production in response to viruses. Nat Immu Cell Growth Regul. 1985;4 (3):120–137. [PubMed] [Google Scholar]
  • 52. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4 (7):499–511. [DOI] [PubMed] [Google Scholar]
  • 53. Dinarello CA. Biologic basis for interleukin-1 in disease. Blood. 1996;87 (6):2095–2147. [PubMed] [Google Scholar]
  • 54. Adachi O, Kawai T, Takeda K, et al. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity. 1998;9 (1):143–150. [DOI] [PubMed] [Google Scholar]
  • 55. Seo SU, Kwon HJ, Song JH, et al. MyD88 signaling is indispensable for primary influenza A virus infection but dispensable for secondary infection. J Virol. 2010;84 (24):12713–12722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Lund JM, Alexopoulou L, Sato A, et al. Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci U S A. 2004;101 (15):5598–5603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Kim YM, Brinkmann MM, Paquet ME, Ploegh HL. UNC93B1 delivers nucleotide-sensing toll-like receptors to endolysosomes. Nature. 2008;452 (7184):234–238. [DOI] [PubMed] [Google Scholar]
  • 58. Tabeta K, Hoebe K, Janssen EM, et al. The Unc93b1 mutation 3d disrupts exogenous antigen presentation and signaling via Toll-like receptors 3, 7 and 9. Nat Immunol. 2006;7 (2):156–164. [DOI] [PubMed] [Google Scholar]
  • 59. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004;303 (5663):1529–1531. [DOI] [PubMed] [Google Scholar]
  • 60. Garcia-Sastre A. Mechanisms of inhibition of the host interferon alpha/beta-mediated antiviral responses by viruses. Microbes Infect. 2002;4 (6):647–655. [DOI] [PubMed] [Google Scholar]
  • 61. Cella M, Facchetti F, Lanzavecchia A, Colonna M. Plasmacytoid dendritic cells activated by influenza virus and CD40 L drive a potent TH1 polarization. Nat Immunol. 2000;1 (4):305–310. [DOI] [PubMed] [Google Scholar]
  • 62. Asselin-Paturel C, Boonstra A, Dalod M, et al. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nat Immunol. 2001;2 (12):1144–1150. [DOI] [PubMed] [Google Scholar]
  • 63. Diebold SS, Montoya M, Unger H, et al. Viral infection switches non-plasmacytoid dendritic cells into high interferon producers. Nature. 2003;424 (6946):324–328. [DOI] [PubMed] [Google Scholar]
  • 64. Koga K, Mor G. Toll-like receptors at the maternal-fetal interface in normal pregnancy and pregnancy disorders. Am J Reprod Immunol. 2010;63 (6):587–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Beijar EC, Mallard C, Powell TL. Expression and subcellular localization of TLR-4 in term and first trimester human placenta. Placenta. 2006;27 (2-3):322–326. [DOI] [PubMed] [Google Scholar]
  • 66. Mitsunari M, Yoshida S, Shoji T, et al. Macrophage-activating lipopeptide-2 induces cyclooxygenase-2 and prostaglandin E(2) via toll-like receptor 2 in human placental trophoblast cells. J Reprod Immunol. 2006;72 (1-2):46–59. [DOI] [PubMed] [Google Scholar]
  • 67. Abrahams VM, Bole-Aldo P, Kim YM, et al. Divergent trophoblast responses to bacterial products mediated by TLRs. J Immunol. 2004;173 (7):4286–4296. [DOI] [PubMed] [Google Scholar]
  • 68. Tinsley JH, Chiasson VL, Mahajan A, Young KJ, Mitchell BM. Toll-like receptor 3 activation during pregnancy elicits preeclampsia-like symptoms in rats. Am J Hypertens. 2009;22 (12):1314–1319. [DOI] [PubMed] [Google Scholar]
  • 69. Panda B, Panda A, Ueda I, et al. Dendritic cells in the circulation of women with preeclampsia demonstrate a pro-inflammatory bias secondary to dysregulation of TLR receptors. J Reprod Immunol. 2012;94 (2):210–215. [DOI] [PubMed] [Google Scholar]
  • 70. Thaxton JE, Romero R, Sharma S. TLR9 activation coupled to IL-10 deficiency induces adverse pregnancy outcomes. J Immunol. 2009;183 (2):1144–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Ilievski V, Lu SJ, Hirsch E. Activation of toll-like receptors 2 or 3 and preterm delivery in the mouse. Reprod Sci. 2007;14 (4):315–320. [DOI] [PubMed] [Google Scholar]
  • 72. Nitsche JF, Jiang SW, Brost BC. Toll-like receptor-2 and toll-like receptor-4 expression on maternal neutrophils during pregnancy. Am J Reprod Immunol. 2010;64 (6):427–434. [DOI] [PubMed] [Google Scholar]
  • 73. Vanders RL, Gibson PG, Murphy VE, Wark PA. Plasmacytoid dendritic cells and CD8 T cells from pregnant women show altered phenotype and function following H1N1/09 infection. J Infect Dis. 2013;208 (7):1062–1070. [DOI] [PubMed] [Google Scholar]
  • 74. Young BC, Stanic AK, Panda B, Rueda BR, Panda A. Longitudinal expression of Toll-like receptors on dendritic cells in uncomplicated pregnancy and postpartum. Am J Obstet Gynecol. 2014;210 (5):445.e1–e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Conn CA, McClellan JL, Maassab HF, Smitka CW, Majde JA, Kluger MJ. Cytokines and the acute phase response to influenza virus in mice. Am J Physiol. 1995;268 (1 pt 2):R78–R84. [DOI] [PubMed] [Google Scholar]
  • 76. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003;21:335–376. [DOI] [PubMed] [Google Scholar]
  • 77. Choudhary S, Gao J, Leaman DW, De BP. Interferon action against human parainfluenza virus type 3: involvement of a novel antiviral pathway in the inhibition of transcription. J Virol. 2001;75 (10):4823–4831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Khaitov MR, Laza-Stanca V, Edwards MR, et al. Respiratory virus induction of alpha-, beta- and lambda-interferons in bronchial epithelial cells and peripheral blood mononuclear cells. Allergy. 2009;64 (3):375–386. [DOI] [PubMed] [Google Scholar]
  • 79. Honda K, Yanai H, Takaoka A, Taniguchi T. Regulation of the type I IFN induction: a current view. Int Immunol. 2005;17 (11):1367–1378. [DOI] [PubMed] [Google Scholar]
  • 80. Forbes RL, Wark PA, Murphy VE, Gibson PG. Pregnant women have attenuated innate interferon responses to 2009 pandemic influenza A virus subtype H1N1. J Infect Dis. 2012;206 (5):646–653. [DOI] [PubMed] [Google Scholar]
  • 81. Plata-Salaman CR. Immunoregulators in the nervous system. Neurosci Biobehav Rev. 1991;15 (2):185–215. [DOI] [PubMed] [Google Scholar]
  • 82. Tracey KJ, Cerami A. Tumor necrosis factor: an updated review of its biology. Crit Care Med. 1993;21 (10 suppl):S415–S422. [PubMed] [Google Scholar]
  • 83. Peper RL, Van Campen H. Tumor necrosis factor as a mediator of inflammation in influenza A viral pneumonia. Microb Pathog. 1995;19 (3):175–183. [DOI] [PubMed] [Google Scholar]
  • 84. Heltzer ML, Coffin SE, Maurer K, et al. Immune dysregulation in severe influenza. J Leukoc Biol. 2009;85 (6):1036–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9 (5):503–510. [DOI] [PubMed] [Google Scholar]
  • 86. Denney L, Aitken C, Li CK, et al. Reduction of natural killer but not effector CD8 T lymphocytes in three consecutive cases of severe/lethal H1N1/09 influenza A virus infection. PLoS One. 2010;5 (5):e10675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Gerhard W, Mozdzanowska K, Furchner M, Washko G, Maiese K. Role of the B-cell response in recovery of mice from primary influenza virus infection. Immunol Rev. 1997;159:95–103. [DOI] [PubMed] [Google Scholar]
  • 88. Virelizier JL. Host defenses against influenza virus: the role of anti-hemagglutinin antibody. J Immunol. 1975;115 (2):434–439. [PubMed] [Google Scholar]
  • 89. Schulman JL, Kilbourne ED. Induction of Partial Specific Heterotypic Immunity in Mice by a Single Infection with Influenza a Virus. J Bacteriol. 1965;89:170–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. McKinstry KK, Strutt TM, Swain SL. Hallmarks of CD4 T cell immunity against influenza. J Intern Med. 2011;269 (5):507–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Mathews JD, McBryde ES, McVernon J, Pallaghy PK, McCaw JM. Prior immunity helps to explain wave-like behaviour of pandemic influenza in 1918-9. BMC Infect Dis. 2010;10:128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Scheible K, Zhang G, Baer J, et al. CD8+ T cell immunity to 2009 pandemic and seasonal H1N1 influenza viruses. Vaccine. 2011;29 (11):2159–2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Gordon CL, Johnson PD, Permezel M, et al. Association between severe pandemic 2009 influenza A (H1N1) virus infection and immunoglobulin G(2) subclass deficiency. Clin Infect Dis. 2010;50 (5):672–678. [DOI] [PubMed] [Google Scholar]
  • 94. Zheng R, Qin X, Li Y, et al. Imbalanced anti-H1N1 immunoglobulin subclasses and dysregulated cytokines in hospitalized pregnant women with 2009 H1N1 influenza and pneumonia in Shenyang, China. Hum Immunol. 2012;73 (9):906–911. [DOI] [PubMed] [Google Scholar]
  • 95. Parry S, Strauss JF., III Premature rupture of the fetal membranes. N Engl J Med. 1998;338 (10):663–670. [DOI] [PubMed] [Google Scholar]
  • 96. Sargent IL, Borzychowski AM, Redman CW. NK cells and human pregnancy--an inflammatory view. Trends Immunol. 2006;27 (9):399–404. [DOI] [PubMed] [Google Scholar]
  • 97. Christiaens I, Zaragoza DB, Guilbert L, Robertson SA, Mitchell BF, Olson DM. Inflammatory processes in preterm and term parturition. J Reprod Immunol. 2008;79 (1):50–57. [DOI] [PubMed] [Google Scholar]
  • 98. Smarason AK, Gunnarsson A, Alfredsson JH, Valdimarsson H. Monocytosis and monocytic infiltration of decidua in early pregnancy. J Clin Lab Immunol. 1986;21 (1):1–5. [PubMed] [Google Scholar]
  • 99. Kraus TA, Sperling RS, Engel SM, et al. Peripheral blood cytokine profiling during pregnancy and post-partum periods. Am J Reprod Immunol. 2010;64 (6):411–426. [DOI] [PubMed] [Google Scholar]
  • 100. Khakpour M, Saidi A, Naficy K. Proved viraemia in Asian influenza (Hong Kong variant) during incubation period. Br Med J. 1969;4 (5677):208–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Naficy K. Human influenza infection with proved viremia. Report of a case. N Engl J Med. 1963;269:964–966. [DOI] [PubMed] [Google Scholar]
  • 102. Uchide N, Ohyama K, Yuan B, Sano T, Bessho T, Yamakawa T. Differential mRNA expression of inflammatory cytokines in cultured human fetal membrane cells responding to influenza virus infection. Biol Pharm Bull. 2002;25 (2):239–243. [DOI] [PubMed] [Google Scholar]
  • 103. Wyllie AH, Kerr JF, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol. 1980;68:251–306. [DOI] [PubMed] [Google Scholar]
  • 104. Uchide N, Ohyama K. Antiviral function of pyrrolidine dithiocarbamate against influenza virus: the inhibition of viral gene replication and transcription. J Antimicrob Chemother. 2003;52 (1):8–10. [DOI] [PubMed] [Google Scholar]
  • 105. Uchide N, Ohyama K, Bessho T, Yuan B, Yamakawa T. Effect of antioxidants on apoptosis induced by influenza virus infection: inhibition of viral gene replication and transcription with pyrrolidine dithiocarbamate. Antiviral Res. 2002;56 (3):207–217. [DOI] [PubMed] [Google Scholar]
  • 106. Uchide N, Suzuki A, Ohyama K, Bessho T, Toyoda H. Secretion of bioactive interleukin-6 and tumor necrosis factor-alpha proteins from primary cultured human fetal membrane chorion cells infected with influenza virus. Placenta. 2006;27 (6-7):678–690. [DOI] [PubMed] [Google Scholar]
  • 107. Park JB. Phagocytosis induces superoxide formation and apoptosis in macrophages. Exp Mol Med. 2003;35 (5):325–335. [DOI] [PubMed] [Google Scholar]
  • 108. Lappas M, Permezel M, Georgiou HM, Rice GE. Regulation of proinflammatory cytokines in human gestational tissues by peroxisome proliferator-activated receptor-gamma: effect of 15-deoxy-Delta(12,14)-PGJ(2) and troglitazone. J Clin Endocrinol Metab. 2002;87 (10):4667–4672. [DOI] [PubMed] [Google Scholar]
  • 109. Sullivan MH, Alvi SA, Brown NL, Elder MG, Bennett PR. The effects of a cytokine suppressive anti-inflammatory drug on the output of prostaglandin E(2) and interleukin-1 beta from human fetal membranes. Mol Hum Reprod. 2002;8 (3):281–285. [DOI] [PubMed] [Google Scholar]
  • 110. Lappas M, Permezel M, Georgiou HM, Rice GE. Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro. Biol Reprod. 2002;67 (2):668–673. [DOI] [PubMed] [Google Scholar]
  • 111. Denison FC, Kelly RW, Calder AA, Riley SC. Cytokine secretion by human fetal membranes, decidua and placenta at term. Hum Reprod. 1998;13 (12):3560–3565. [DOI] [PubMed] [Google Scholar]
  • 112. Uchide N, Ohyama K, Bessho T, Takeichi M, Toyoda H. Possible roles of proinflammatory and chemoattractive cytokines produced by human fetal membrane cells in the pathology of adverse pregnancy outcomes associated with influenza virus infection. Mediators Inflamm. 2012;2012:270670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Marvin KW, Keelan JA, Eykholt RL, Sato TA, Mitchell MD. Use of cDNA arrays to generate differential expression profiles for inflammatory genes in human gestational membranes delivered at term and preterm. Mol Hum Reprod. 2002;8 (4):399–408. [DOI] [PubMed] [Google Scholar]
  • 114. Yokota T, Oritani K, Takahashi I, et al. Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood. 2000;96 (5):1723–1732. [PubMed] [Google Scholar]
  • 115. Tsatsanis C, Margioris AN, Kontoyiannis DP. Association between H1N1 infection severity and obesity-adiponectin as a potential etiologic factor. J Infect Dis. 2010;202 (3):459–460. [DOI] [PubMed] [Google Scholar]
  • 116. Avitsur R, Hunzeker J, Sheridan JF. Role of early stress in the individual differences in host response to viral infection. Brain Behav Immun. 2006;20 (4):339–348. [DOI] [PubMed] [Google Scholar]
  • 117. Avitsur R, Kavelaars A, Heijnen C, Sheridan JF. Social stress and the regulation of tumor necrosis factor-alpha secretion. Brain Behav Immun. 2005;19 (4):311–317. [DOI] [PubMed] [Google Scholar]
  • 118. Johnson JD, O'Connor KA, Deak T, Stark M, Watkins LR, Maier SF. Prior stressor exposure sensitizes LPS-induced cytokine production. Brain Behav Immun. 2002;16 (4):461–476. [DOI] [PubMed] [Google Scholar]
  • 119. Christian LM, Franco A, Iams JD, Sheridan J, Glaser R. Depressive symptoms predict exaggerated inflammatory responses to an in vivo immune challenge among pregnant women. Brain Behav Immun. 2010;24 (1):49–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Thangavel RR, Bouvier NM. Animal models for influenza virus pathogenesis, transmission, and immunology [published online April 4, 2014.] J Immunol Methods. 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Moncla LH, Ross TM, Dinis JM, et al. A novel nonhuman primate model for influenza transmission. PLoS One. 2013;8 (11):e78750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Bouvier NM, Lowen AC. Animal models for influenza virus pathogenesis and transmission. Viruses. 2010;2 (8):1530–1563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Eichelberger MC, Green MD. Animal models to assess the toxicity, immunogenicity and effectiveness of candidate influenza vaccines. Expert Opinion Drug Metabol Toxicol. 2011;7 (9):1117–1127. [DOI] [PubMed] [Google Scholar]
  • 124. Tripp RA, Tompkins SM. Animal models for evaluation of influenza vaccines. Curr Top Microbiol Immunol. 2009;333:397–412. [DOI] [PubMed] [Google Scholar]
  • 125. Barnard DL. Animal models for the study of influenza pathogenesis and therapy. Antiviral Res. 2009;82 (2):A110–A122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Kash JC, Basler CF, Garcia-Sastre A, et al. 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. 2004;78 (17):9499–9511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Kobasa D, Takada A, Shinya K, et al. Enhanced virulence of influenza A viruses with the haemagglutinin of the 1918 pandemic virus. Nature. 2004;431 (7009):703–707. [DOI] [PubMed] [Google Scholar]
  • 128. Tumpey TM, Garcia-Sastre A, Taubenberger JK, Palese P, Swayne DE, Basler CF. Pathogenicity and immunogenicity of influenza viruses with genes from the 1918 pandemic virus. Proc Natl Acad Sci U S A. 2004;101 (9):3166–3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Kash JC, Tumpey TM, Proll SC, et al. Genomic analysis of increased host immune and cell death responses induced by 1918 influenza virus. Nature. 2006;443 (7111):578–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Tumpey TM, Garcia-Sastre A, Taubenberger JK, et al. 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. 2005;79 (23):14933–14944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Kobasa D, Jones SM, Shinya K, et al. Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus. Nature. 2007;445 (7125):319–323. [DOI] [PubMed] [Google Scholar]
  • 132. Cheung CY, Poon LL, Lau AS, et al. Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease? Lancet. 2002;360 (9348):1831–1837. [DOI] [PubMed] [Google Scholar]
  • 133. Lee DC, Cheung CY, Law AH, Mok CK, Peiris M, Lau AS. p38 mitogen-activated protein kinase-dependent hyperinduction of tumor necrosis factor alpha expression in response to avian influenza virus H5N1. J Virol. 2005;79 (16):10147–10154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Chan MC, Cheung CY, Chui WH, et al. Proinflammatory cytokine responses induced by influenza A (H5N1) viruses in primary human alveolar and bronchial epithelial cells. Respiratory research. 2005;6:135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Xu T, Qiao J, Zhao L, et al. Acute respiratory distress syndrome induced by avian influenza A (H5N1) virus in mice. Am J Respir Crit Care Med. 2006;174 (9):1011–1017. [DOI] [PubMed] [Google Scholar]
  • 136. de Jong MD, Bach VC, Phan TQ, et al. Fatal avian influenza A (H5N1) in a child presenting with diarrhea followed by coma. N Engl J Med. 2005;352 (7):686–691. [DOI] [PubMed] [Google Scholar]
  • 137. Szretter KJ, Gangappa S, Lu X, et al. Role of host cytokine responses in the pathogenesis of avian H5N1 influenza viruses in mice. J Virol. 2007;81 (6):2736–2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Hui KP, Lee SM, Cheung CY, et al. Induction of proinflammatory cytokines in primary human macrophages by influenza A virus (H5N1) is selectively regulated by IFN regulatory factor 3 and p38 MAPK. J Immunol. 2009;182 (2):1088–1098. [DOI] [PubMed] [Google Scholar]
  • 139. Salomon R, Hoffmann E, Webster RG. Inhibition of the cytokine response does not protect against lethal H5N1 influenza infection. Proc Natl Acad Sci U S A. 2007;104 (30):12479–12481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Luo H, Wang D, Che HL, Zhao Y, Jin H. Pathological observations of lung inflammation after administration of IP-10 in influenza virus- and respiratory syncytial virus-infected mice. Exp Ther Med. 2012;3 (1):76–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Dawson TC, Beck MA, Kuziel WA, Henderson F, Maeda N. Contrasting effects of CCR5 and CCR2 deficiency in the pulmonary inflammatory response to influenza A virus. Am J Pathol. 2000;156 (6):1951–1959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Cook DN, Beck MA, Coffman TM, et al. Requirement of MIP-1 alpha for an inflammatory response to viral infection. Science. 1995;269 (5230):1583–1585. [DOI] [PubMed] [Google Scholar]
  • 143. Tedla N, Wang HW, McNeil HP, et al. Regulation of T lymphocyte trafficking into lymph nodes during an immune response by the chemokines macrophage inflammatory protein (MIP)-1 alpha and MIP-1 beta. J Immunol. 1998;161 (10):5663–5672. [PubMed] [Google Scholar]
  • 144. Opal SM, Wherry JC, Grint P. Interleukin-10: potential benefits and possible risks in clinical infectious diseases. Clin Infect Dis. 1998;27 (6):1497–1507. [DOI] [PubMed] [Google Scholar]
  • 145. Strutt TM, McKinstry KK, Dibble JP, et al. Memory CD4+ T cells induce innate responses independently of pathogen. Nat Med. 2010;16 (5):558–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Kreijtz JH, Bodewes R, van Amerongen G, et al. Primary influenza A virus infection induces cross-protective immunity against a lethal infection with a heterosubtypic virus strain in mice. Vaccine. 2007;25 (4):612–620. [DOI] [PubMed] [Google Scholar]
  • 147. Hikono H, Kohlmeier JE, Ely KH, et al. T-cell memory and recall responses to respiratory virus infections. Immunol Rev. 2006;211:119–132. [DOI] [PubMed] [Google Scholar]
  • 148. Woodland DL, Scott I. T cell memory in the lung airways. Proc Am Thorac Soc. 2005;2 (2):126–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Seo SH, Peiris M, Webster RG. Protective cross-reactive cellular immunity to lethal A/Goose/Guangdong/1/96-like H5N1 influenza virus is correlated with the proportion of pulmonary CD8(+) T cells expressing gamma interferon. J Virol. 2002;76 (10):4886–4890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Deliyannis G, Jackson DC, Ede NJ, et al. Induction of long-term memory CD8(+) T cells for recall of viral clearing responses against influenza virus. J Virol. 2002;76 (9):4212–4221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Belz GT, Wodarz D, Diaz G, Nowak MA, Doherty PC. Compromised influenza virus-specific CD8(+)-T-cell memory in CD4(+)-T-cell-deficient mice. J Virol. 2002;76 (23):12388–12393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Dienz O, Eaton SM, Bond JP, et al. The induction of antibody production by IL-6 is indirectly mediated by IL-21 produced by CD4+ T cells. J Exp Med. 2009;206 (1):69–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Hufford MM, Kim TS, Sun J, Braciale TJ. Antiviral CD8+ T cell effector activities in situ are regulated by target cell type. J Exp Med. 2011;208 (1):167–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Wiley JA, Cerwenka A, Harkema JR, Dutton RW, Harmsen AG. Production of interferon-gamma by influenza hemagglutinin-specific CD8 effector T cells influences the development of pulmonary immunopathology. Am J Pathol. 2001;158 (1):119–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Moskophidis D, Kioussis D. Contribution of virus-specific CD8+ cytotoxic T cells to virus clearance or pathologic manifestations of influenza virus infection in a T cell receptor transgenic mouse model. J Exp Med. 1998;188 (2):223–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Pazos MA, Kraus TA, Munoz-Fontela C, Moran TM. Estrogen mediates innate and adaptive immune alterations to influenza infection in pregnant mice. PLoS One. 2012;7 (7):e40502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Scalzo AA, Anders EM. Influenza viruses as lymphocyte mitogens. I. B cell mitogenesis by influenza A viruses of the H2 and H6 subtypes is controlled by the I-E/C subregion of the major histocompatibility complex. J Immunol. 1985;134 (2):757–760. [PubMed] [Google Scholar]
  • 158. Marshall-Clarke S, Tasker L, Buchatska O, et al. Influenza H2 haemagglutinin activates B cells via a MyD88-dependent pathway. Eur J Immunol. 2006;36 (1):95–106. [DOI] [PubMed] [Google Scholar]
  • 159. Williams K, Mackenzie JS. Influenza infections during pregnancy in the mouse. J Hyg (Lond). 1977;79 (2):249–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Mackenzie JS, Williams K, Papadimitriou J. Influenza A virus and its influence on the outcome of pregnancy in the mouse. Dev Biol Stand. 1977;39:489–496. [PubMed] [Google Scholar]
  • 161. Chan KH, Zhang AJ, To KK, et al. Wild type and mutant 2009 pandemic influenza A (H1N1) viruses cause more severe disease and higher mortality in pregnant BALB/c mice. PLoS One. 2010;5 (10):e13757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Kim HM, Kang YM, Song BM, Kim HS, Seo SH. The 2009 pandemic H1N1 influenza virus is more pathogenic in pregnant mice than seasonal H1N1 influenza virus. Viral Immunol. 2012;25 (5):402–410. [DOI] [PubMed] [Google Scholar]
  • 163. Marcelin G, Aldridge JR, Duan S, et al. 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. 2011;85 (21):11208–11219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Gonzalez JM, Ofori E, Burd I, Chai J, Scholler N, Elovitz MA. Maternal mortality from systemic illness: unraveling the contribution of the immune response. Am J Obstet Gynecol. 2009;200 (4):430. e431–438. [DOI] [PubMed] [Google Scholar]
  • 165. Robinson DP, Klein SL. Pregnancy and pregnancy-associated hormones alter immune responses and disease pathogenesis. Horm Behav. 2012;62 (3):263–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Krishnan L, Guilbert LJ, Russell AS, Wegmann TG, Mosmann TR, Belosevic M. Pregnancy impairs resistance of C57BL/6 mice to Leishmania major infection and causes decreased antigen-specific IFN-gamma response and increased production of T helper 2 cytokines. J Immunol. 1996;156 (2):644–652. [PubMed] [Google Scholar]
  • 167. Luft BJ, Remington JS. Effect of pregnancy on resistance to Listeria monocytogenes and Toxoplasma gondii infections in mice. Infect Immun. 1982;38 (3):1164–1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Szekeres-Bartho J, Polgar B. PIBF: the double edged sword. Pregnancy and tumor. Am J Reprod Immunol. 2010;64(2):77–86. [DOI] [PubMed] [Google Scholar]
  • 169. Szekeres-Bartho J, Wegmann TG. A progesterone-dependent immunomodulatory protein alters the Th1/Th2 balance. J Reprod Immunol. 1996;31 (1-2):81–95. [DOI] [PubMed] [Google Scholar]
  • 170. Szekeres-Bartho J, Faust Z, Varga P, Szereday L, Kelemen K. The immunological pregnancy protective effect of progesterone is manifested via controlling cytokine production. Am J Reprod Immunol. 1996;35 (4):348–351. [DOI] [PubMed] [Google Scholar]
  • 171. Veenstra van Nieuwenhoven AL, Bouman A, Moes H, et al. Cytokine production in natural killer cells and lymphocytes in pregnant women compared with women in the follicular phase of the ovarian cycle. Fertil Steril. 2002;77 (5):1032–1037. [DOI] [PubMed] [Google Scholar]
  • 172. Sacks GP, Clover LM, Bainbridge DR, Redman CW, Sargent IL. Flow cytometric measurement of intracellular Th1 and Th2 cytokine production by human villous and extravillous cytotrophoblast. Placenta. 2001;22 (6):550–559. [DOI] [PubMed] [Google Scholar]
  • 173. Ostensen M. Sex hormones and pregnancy in rheumatoid arthritis and systemic lupus erythematosus. Ann N Y Acad Sci. 1999;876:131–143; discussion 144. [DOI] [PubMed] [Google Scholar]
  • 174. Marzi M, Vigano A, Trabattoni D, et al. Characterization of type 1 and type 2 cytokine production profile in physiologic and pathologic human pregnancy. Clin Exp Immunol. 1996;106 (1):127–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Lin H, Mosmann TR, Guilbert L, Tuntipopipat S, Wegmann TG. Synthesis of T helper 2-type cytokines at the maternal-fetal interface. J Immunol. 1993;151 (9):4562–4573. [PubMed] [Google Scholar]
  • 176. Alijotas-Reig J, Llurba E, Gris JM. Potentiating maternal immune tolerance in pregnancy: a new challenging role for regulatory T cells. Placenta. 2014;35 (4):241–248. [DOI] [PubMed] [Google Scholar]
  • 177. Zenclussen AC, Gerlof K, Zenclussen ML, et al. Abnormal T-cell reactivity against paternal antigens in spontaneous abortion: adoptive transfer of pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in a murine abortion model. Am J Pathol. 2005;166 (3):811–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Zenclussen AC, Fest S, Busse P, Joachim R, Klapp BF, Arck PC. Questioning the Th1/Th2 paradigm in reproduction: peripheral levels of IL-12 are down-regulated in miscarriage patients. Am J Reprod Immunol. 2002;48 (4):245–251. [DOI] [PubMed] [Google Scholar]
  • 179. Robinson DP, Lorenzo ME, Jian W, Klein SL. Elevated 17beta-estradiol protects females from influenza A virus pathogenesis by suppressing inflammatory responses. PLoS Pathog. 2011;7 (7):e1002149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Greenlee MM, Mitzelfelt JD, Yu L, et al. Estradiol activates epithelial sodium channels in rat alveolar cells through the G protein-coupled estrogen receptor. Am J Physiol Lung Cell Mol Physiol. 2013;305 (11):L878–L889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Gu X, Li P, Liu H, Li N, Li S, Sakuma T. The effect of influenza virus A on th1/th2 balance and alveolar fluid clearance in pregnant rats. Exp Lung Res. 2011;37 (7):445–451. [DOI] [PubMed] [Google Scholar]
  • 182. Chen XJ, Seth S, Yue G, et al. Influenza virus inhibits ENaC and lung fluid clearance. Am J Physiol Lung Cell Mol Physiol. 2004;287 (2):L366–L373. [DOI] [PubMed] [Google Scholar]
  • 183. Brown AS. Prenatal infection as a risk factor for schizophrenia. Schizophr Bull. 2006;32 (2):200–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Gilmore JH, Fredrik Jarskog L, Vadlamudi S, Lauder JM. Prenatal infection and risk for schizophrenia: IL-1beta, IL-6, and TNFalpha inhibit cortical neuron dendrite development. Neuropsychopharmacology. 2004;29 (7):1221–1229. [DOI] [PubMed] [Google Scholar]
  • 185. Mednick SA, Machon RA, Huttunen MO, Bonett D. Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch Gen Psychiatry. 1988;45 (2):189–192. [DOI] [PubMed] [Google Scholar]
  • 186. Meyer U, Feldon J, Schedlowski M, Yee BK. Towards an immuno-precipitated neurodevelopmental animal model of schizophrenia. Neurosci Biobehav Rev. 2005;29 (6):913–947. [DOI] [PubMed] [Google Scholar]
  • 187. Short SJ, Lubach GR, Karasin AI, et al. Maternal influenza infection during pregnancy impacts postnatal brain development in the rhesus monkey. Biol Psychiatry. 2010;67 (10):965–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Parboosing R, Bao Y, Shen L, Schaefer CA, Brown AS. Gestational influenza and bipolar disorder in adult offspring. JAMA Psychiatry (Chicago, Ill.). 2013;70 (7):677–685. [DOI] [PubMed] [Google Scholar]
  • 189. Toth LA. Strain differences in the somnogenic effects of interferon inducers in mice. J Interferon Cytokine Res. 1996;16 (12):1065–1072. [DOI] [PubMed] [Google Scholar]
  • 190. Cox RJ, Brokstad KA, Ogra P. Influenza virus: immunity and vaccination strategies. Comparison of the immune response to inactivated and live, attenuated influenza vaccines. Scand J Immunol. 2004;59 (1):1–15. [DOI] [PubMed] [Google Scholar]
  • 191. Gross PA, Ennis FA, Gaerlan PF, Denson LJ, Denning CR, Schiffman D. A controlled double-blind comparison of reactogenicity, immunogenicity, and protective efficacy of whole-virus and split-product influenza vaccines in children. J Infect Dis. 1977;136 (5):623–632. [DOI] [PubMed] [Google Scholar]
  • 192. Barry DW, Mayner RE, Staton E, et al. Comparative trial of influenza vaccines. I. Immunogenicity of whole virus and split product vaccines in man. Am J Epidemiol. 1976;104(1):34–46. [DOI] [PubMed] [Google Scholar]
  • 193. Hovden AO, Cox RJ, Madhun A, Haaheim LR. Two doses of parenterally administered split influenza virus vaccine elicited high serum IgG concentrations which effectively limited viral shedding upon challenge in mice. Scand J Immunol. 2005;62 (4):342–352. [DOI] [PubMed] [Google Scholar]
  • 194. American College of O, Gynecologists. ACOG committee opinion no. 558: Integrating immunizations into practice. Obstet Gynecol. 2013;121 (4):897–903. [DOI] [PubMed] [Google Scholar]
  • 195. Prevention and control of seasonal influenza with vaccines. Recommendations of the Advisory Committee on Immunization Practices--United States, 2013-2014. MMWR Recomm Rep. 2013;62 (RR-07):1–43. [PubMed] [Google Scholar]
  • 196. Bednarczyk RA, Adjaye-Gbewonyo D, Omer SB. Safety of influenza immunization during pregnancy for the fetus and the neonate. Am J Obstet Gynecol. 2012;207 (3):S38–S46. [DOI] [PubMed] [Google Scholar]
  • 197. Smith NM, Bresee JS, Shay DK, Uyeki TM, Cox NJ, Strikas RA. Prevention and Control of Influenza: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. 2006;55 (RR-10):1–42. [PubMed] [Google Scholar]
  • 198. Nordin JD, Kharbanda EO, Vazquez Benitez G, et al. Maternal influenza vaccine and risks for preterm or small for gestational age birth. J Pediatr. 2014;164 (5):1051–1057.e2. [DOI] [PubMed] [Google Scholar]
  • 199. Legge A, Dodds L, Macdonald NE, Scott J, McNeil S. Rates and determinants of seasonal influenza vaccination in pregnancy and association with neonatal outcomes. CMAJ. 2014;186 (4):E157–E164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Chambers CD, Johnson D, Xu R, et al. Risks and safety of pandemic h1n1 influenza vaccine in pregnancy: birth defects, spontaneous abortion, preterm delivery, and small for gestational age infants. Vaccine. 2013;31 (44):5026–5032. [DOI] [PubMed] [Google Scholar]
  • 201. Moro PL, Museru OI, Broder K, et al. Safety of influenza A (H1N1) 2009 live attenuated monovalent vaccine in pregnant women. Obstet Gynecol. 2013;122 (6):1271–1278. [DOI] [PubMed] [Google Scholar]
  • 202. Kennedy ED, Ahluwalia IB, Ding H, Lu P-J, Singleton JA, Bridges CB. Monitoring seasonal influenza vaccination coverage among pregnant women in the United States. Am J Obstet Gynecol. 2012;207 (3 suppl):S9–S16. [DOI] [PubMed] [Google Scholar]
  • 203. Rasmussen SA, Watson AK, Kennedy ED, Broder KR, Jamieson DJ. Vaccines and pregnancy: past, present, and future [published online December 16, 2013]. Semin Fetal Neonatal Medicine . Dec 16 2013. [DOI] [PubMed] [Google Scholar]
  • 204. Zaman K, Roy E, Arifeen SE, et al. Effectiveness of maternal influenza immunization in mothers and infants. N Engl J Med. 2008;359 (15):1555–1564. [DOI] [PubMed] [Google Scholar]
  • 205. Steinhoff MC, Omer SB, Roy E, et al. Influenza immunization in pregnancy--antibody responses in mothers and infants. N Engl J Med. 2010;362 (17):1644–1646. [DOI] [PubMed] [Google Scholar]
  • 206. Yamaguchi K, Hisano M, Isojima S, et al. Relationship of Th1/Th2 cell balance with the immune response to influenza vaccine during pregnancy. J Med Virol. 2009;81 (11):1923–1928. [DOI] [PubMed] [Google Scholar]
  • 207. Hwang SD, Shin JS, Ku KB, Kim HS, Cho SW, Seo SH. Protection of pregnant mice, fetuses and neonates from lethality of H5N1 influenza viruses by maternal vaccination. Vaccine. 2010;28 (17):2957–2964. [DOI] [PubMed] [Google Scholar]
  • 208. Luke TC, Kilbane EM, Jackson JL, Hoffman SL. Meta-analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment? Ann Intern Med. 2006;145 (8):599–609. [DOI] [PubMed] [Google Scholar]
  • 209. Hammerbeck DM, Burleson GR, Schuller CJ, et al. Administration of a dual toll-like receptor 7 and toll-like receptor 8 agonist protects against influenza in rats. Antiviral Res. 2007;73 (1):1–11. [DOI] [PubMed] [Google Scholar]
  • 210. Wong JP, Saravolac EG, Sabuda D, Levy HB, Kende M. Prophylactic and therapeutic efficacies of poly(IC.LC) against respiratory influenza A virus infection in mice. Antimicrob Agents Chemother. 1995;39 (11):2574–2576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. McFarlin DE, Bever CT, Salazar AM, Levy HB. A preliminary trial of poly(I, C)-LC in multiple sclerosis. J Biol Response Mod. 1985;4 (5):544–548. [PubMed] [Google Scholar]
  • 212. Koga K, Cardenas I, Aldo P, et al. Activation of TLR3 in the trophoblast is associated with preterm delivery. Am J Reprod Immunol. 2009;61 (3):196–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Liu Y, Li S, Zhang G, et al. Genetic variants in IL1A and IL1B contribute to the susceptibility to 2009 pandemic H1N1 influenza A virus. BMC Immunol. 2013;14:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Juno J, Fowke KR, Keynan Y. Immunogenetic factors associated with severe respiratory illness caused by zoonotic H1N1 and H5N1 influenza viruses. Clin Dev Immunol. 2012;2012:797180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Kumar PA, Hu Y, Yamamoto Y, et al. Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell. 2011;147 (3):525–538. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Reproductive Sciences are provided here courtesy of Society for Reproductive Investigation

RESOURCES