Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Neuroimmunomodulation ; 28(1): 1-21, 2021.
Article in English | MEDLINE | ID: covidwho-1206095

ABSTRACT

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) has devastating effects on the population worldwide. Given this scenario, the extent of the impact of the disease on more vulnerable individuals, such as pregnant women, is of great concern. Although pregnancy may be a risk factor in respiratory virus infections, there are no considerable differences regarding COVID-19 severity observed between pregnant and nonpregnant women. In these circumstances, an emergent concern is the possibility of neurodevelopmental and neuropsychiatric harm for the offspring of infected mothers. Currently, there is no stronger evidence indicating vertical transmission of SARS-CoV-2; however, the exacerbated inflammatory response observed in the disease could lead to several impairments in the offspring's brain. Furthermore, in the face of historical knowledge on possible long-term consequences for the progeny's brain after infection by viruses, we must consider that this might be another deleterious facet of COVID-19. In light of neuroimmune interactions at the maternal-fetal interface, we review here the possible harmful outcomes to the offspring brains of mothers infected by SARS-CoV-2.


Subject(s)
COVID-19/immunology , Neurodevelopmental Disorders/physiopathology , Neuroimmunomodulation/immunology , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/physiopathology , COVID-19/metabolism , COVID-19/physiopathology , Cytokine Release Syndrome/immunology , Decidua/immunology , Female , Humans , Immune Tolerance/immunology , Infectious Disease Transmission, Vertical , Neuroimmunomodulation/physiology , Placenta/immunology , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/physiopathology , SARS-CoV-2 , Umbilical Cord/immunology
2.
Mucosal Immunol ; 14(5): 991-1005, 2021 09.
Article in English | MEDLINE | ID: covidwho-1203411

ABSTRACT

Many maternal immune cells populate the decidua, which is the mucosal lining of the uterus transformed during pregnancy. Here, abundant natural killer (NK) cells and macrophages help the uterine vasculature adapt to fetal demands for gas and nutrients, thereby supporting fetal growth. Fetal trophoblast cells budding off the forming placenta and invading deep into maternal tissues come into contact with these and other immune cells. Besides their homeostatic functions, decidual NK cells can respond to pathogens during infection, but in doing so, they may become conflicted between destroying the invader and sustaining fetoplacental growth. We review how maternal NK cells balance their double duty both in the local microenvironment of the uterus and systemically, during toxoplasmosis, influenza, cytomegalovirus, malaria and other infections that threat pregnancy. We also discuss recent developments in the understanding of NK-cell responses to SARS-Cov-2 infection and the possible dangers of COVID-19 during pregnancy.


Subject(s)
Decidua/immunology , Immunity, Mucosal , Killer Cells, Natural/immunology , Pregnancy Complications, Infectious/immunology , COVID-19/immunology , Cytokines/immunology , Female , Humans , Pregnancy
3.
J Immunol ; 205(11): 3083-3094, 2020 12 01.
Article in English | MEDLINE | ID: covidwho-902734

ABSTRACT

Vertical transmission of the Zika virus (ZIKV) causes severe fetal defects, but the exact pathogenic mechanism is unclear. We identified up to a 10,480-fold higher expression of viral attachment factors AXL, GAS6, and PROS1 and a 3880-fold increase in ZIKV infectiousness/propagation in human term decidual stromal cells versus trophoblasts. Moreover, levels of viral attachment factors and ZIKV are significantly increased, whereas expression of innate immune response genes are significantly decreased, in human first trimester versus term decidual cells. ZIKV-infected decidual cell supernatants increased cytotrophoblasts infection up to 252-fold compared with directly infected cytotrophoblasts. Tizoxanide treatment efficiently inhibited Zika infection in both maternal and fetal cells. We conclude that ZIKV permissiveness, as well as innate immune responsiveness of human decidual cells, are gestational age dependent, and decidual cells augment ZIKV infection of primary human cytotrophoblast cultures, which are otherwise ZIKV resistant. Human decidual cells may act as reservoirs for trimester-dependent placental transmission of ZIKV, accounting for the higher Zika infection susceptibility and more severe fetal sequelae observed in early versus late pregnancy. Moreover, tizoxanide is a promising agent in preventing perinatal Zika transmission as well as other RNA viruses such as coronavirus.


Subject(s)
Decidua , Gestational Age , Immunity, Innate , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious , Zika Virus Infection , Zika Virus/immunology , Animals , Chlorocebus aethiops , Decidua/immunology , Decidua/pathology , Decidua/virology , Female , Humans , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/pathology , Trophoblasts , Vero Cells , Zika Virus Infection/immunology , Zika Virus Infection/pathology , Zika Virus Infection/transmission
4.
Front Immunol ; 11: 572567, 2020.
Article in English | MEDLINE | ID: covidwho-886164

ABSTRACT

Immunological adaptations in pregnancy allow maternal tolerance of the semi-allogeneic fetus but also increase maternal susceptibility to infection. At implantation, the endometrial stroma, glands, arteries and immune cells undergo anatomical and functional transformation to create the decidua, the specialized secretory endometrium of pregnancy. The maternal decidua and the invading fetal trophoblast constitute a dynamic junction that facilitates a complex immunological dialogue between the two. The decidual and peripheral immune systems together assume a pivotal role in regulating the critical balance between tolerance and defense against infection. Throughout pregnancy, this equilibrium is repeatedly subjected to microbial challenge. Acute viral infection in pregnancy is associated with a wide spectrum of adverse consequences for both mother and fetus. Vertical transmission from mother to fetus can cause developmental anomalies, growth restriction, preterm birth and stillbirth, while the mother is predisposed to heightened morbidity and maternal death. A rapid, effective response to invasive pathogens is therefore essential in order to avoid overwhelming maternal infection and consequent fetal compromise. This sentinel response is mediated by the innate immune system: a heritable, highly evolutionarily conserved system comprising physical barriers, antimicrobial peptides (AMP) and a variety of immune cells-principally neutrophils, macrophages, dendritic cells, and natural killer cells-which express pattern-receptors that detect invariant molecular signatures unique to pathogenic micro-organisms. Recognition of these signatures during acute infection triggers signaling cascades that enhance antimicrobial properties such as phagocytosis, secretion of pro-inflammatory cytokines and activation of the complement system. As well as coordinating the initial immune response, macrophages and dendritic cells present microbial antigens to lymphocytes, initiating and influencing the development of specific, long-lasting adaptive immunity. Despite extensive progress in unraveling the immunological adaptations of pregnancy, pregnant women remain particularly susceptible to certain acute viral infections and continue to experience mortality rates equivalent to those observed in pandemics several decades ago. Here, we focus specifically on the pregnancy-induced vulnerabilities in innate immunity that contribute to the disproportionately high maternal mortality observed in the following acute viral infections: Lassa fever, Ebola virus disease (EVD), dengue fever, hepatitis E, influenza, and novel coronavirus infections.


Subject(s)
Decidua/immunology , Placenta/immunology , Virus Diseases/immunology , Adaptive Immunity/immunology , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Dengue/immunology , Dengue/pathology , Female , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/pathology , Hepatitis E/immunology , Hepatitis E/pathology , Humans , Immune Tolerance/immunology , Immunity, Innate/immunology , Influenza, Human/immunology , Influenza, Human/pathology , Lassa Fever/immunology , Lassa Fever/pathology , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL