Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 306
Filtrar
1.
Med Ultrason ; 25(2): 233-235, 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-36191249

RESUMO

The effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on placental tissue is unclear. We present a case of symptomatic first trimester SARS-CoV-2 infection in which longitudinal ultrasound images demonstrated diffuse areas of echogenic foci. Her 39-week delivery, following an elective induction of labor, was uncomplicated, and placental pathol-ogy evaluation noted extensive calcifications. Such findings are sometimes seen in late and post-term pregnancies and those complicated by smoking, hypertensive disorders, diabetes, and viral infections. In this case, no other potential etiology was identified. Thus, we conclude that placental calcifications may be associated with SAR-CoV-2 infection in early pregnancy.


Assuntos
COVID-19 , Calcinose , Complicações Infecciosas na Gravidez , Humanos , Gravidez , Feminino , COVID-19/complicações , COVID-19/diagnóstico por imagem , Placenta/diagnóstico por imagem , SARS-CoV-2 , Primeiro Trimestre da Gravidez , Complicações Infecciosas na Gravidez/diagnóstico por imagem , Complicações Infecciosas na Gravidez/patologia , Calcinose/complicações , Calcinose/diagnóstico por imagem , Calcinose/patologia
2.
Am J Surg Pathol ; 46(8): 1036-1047, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35319524

RESUMO

Coronavirus disease 2019 (COVID-19) infection in pregnancy has been associated with preterm delivery and preeclampsia. A less frequent and underrecognized complication is extensive placental infection which is associated with high rates of perinatal morbidity and mortality. The frequency, early pathogenesis, and range of lesions associated with this infection are poorly understood. We conducted a population-based study of placental pathology from all mothers with COVID-19 (n=271) over an 18-month period delivering within our health system. The overall prevalence of diffuse severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) placentitis, as defined by typical histology and immunohistochemical (IHC) staining for SARS-CoV-2 spike protein, was 14.8/1000, but increased to 59/1000 in preterm births. We also identified 3 cases with isolated small foci of localized SARS-CoV-2 placentitis, characterized by focal perivillous fibrin and intervillositis, which illustrate the early pathogenesis and suggest that infection may be contained in some cases. Two other placental lesions were more common in mothers with COVID-19, high-grade maternal vascular malperfusion in preterm deliveries and high-grade chronic villitis at term (5/5 cases tested of the latter were negative by IHC for SARS-CoV-2). Additional investigation of diffuse and localized SARS-CoV-2 placentitis by IHC showed loss of BCL-2, C4d staining in surrounding villi, and an early neutrophil-predominant intervillous infiltrate that later became dominated by monocyte-macrophages. We propose a model of focal infection of syncytiotrophoblast by virally infected maternal leukocytes leading to loss of BCL-2 and apoptosis. Infection is then either contained by surrounding fibrinoid (localized) or initiates waves of aponecrosis and immune activation that spread throughout the villous parenchyma (diffuse).


Assuntos
COVID-19 , Complicações Infecciosas na Gravidez , COVID-19/complicações , Feminino , Humanos , Recém-Nascido , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/epidemiologia , Complicações Infecciosas na Gravidez/patologia , Prevalência , Proteínas Proto-Oncogênicas c-bcl-2 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus
3.
J Reprod Immunol ; 147: 103368, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34461555

RESUMO

Recent studies suggest that women with high exposures to dibutyl phthalate (DBP) are at increased risk for preterm birth, a condition associated with aberrant inflammation in the placenta often caused by subclinical infections. Placental inflammation is also a risk factor for neurodevelopmental disorders whose risk may also be enhanced by DBP. It is unclear, however, if DBP enhances placental inflammation. Therefore, we studied the effects of DBP on the production of biomarkers of placental inflammation and neurodevelopment under basal conditions and a setting of mild infection. Placental explant cultures established from women undergoing elective caesarean delivery were treated with DBP with and without co-stimulation by 107 CFU/mL heat-killed E. coli for 24 h at 37 °C. Conditioned medium was harvested and concentrations of IL-1ß, TNF-α, IL-10, HO-1 and BDNF, a biomarker for neurodevelopment, were quantified. DBP significantly enhanced IL-6 production in basal cultures but had no significant on the other biomarkers quantified. Both TNF-α and IL-1ß production was enhanced by DBP for cultures co-stimulated with E. coli. Although marginal enhancement of IL-6, and IL-10 were observed for bacteria co-treated cultures, results were either non-monotonic or only approached statistical significance. HO-1 production tended to be reduced at the highest concentration of DBP tested and BDNF production was reduced by DBP in a dose-dependent manner for bacteria-stimulated cultures. These results suggest that DBP enhances basal IL-6 production but has little or no effect on other biomarkers studied. However, DBP enhances IL-1ß and TNF-α production but reduces BDNF production by bacteria-stimulated cultures.


Assuntos
Dibutilftalato/toxicidade , Poluentes Ambientais/toxicidade , Placenta/imunologia , Complicações Infecciosas na Gravidez/induzido quimicamente , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Placenta/efeitos dos fármacos , Placenta/metabolismo , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/patologia , Cultura Primária de Células/métodos , Fator de Necrose Tumoral alfa/metabolismo
4.
Biomolecules ; 11(8)2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34439868

RESUMO

The term 'cytokine storm' (CS) applies to a pathological autoimmune reaction when the interactions that lead to cytokine production are destabilised and may even lead to death. CS may be induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In this study, we present our analysis of certain pathological processes that induce a CS in pregnant and postpartum women. We draw our attention to the similarities between the severe course of Coronavirus Disease 2019 (COVID-19) and haemophagocytic lymphohistiocytosis (HLH). It is noteworthy that many of the criteria used to diagnose HLH are described as COVID-19 mortality predictors. Cytokine storms are considered to be an important cause of death in patients with the severe course of SARS-CoV-2 infection. Due to the fact that pregnant women are in an immunosuppressive state, viral pulmonary infections are more perilous for them-possible risks include miscarriage, intrauterine growth restriction or birth before the term; sometimes ventilation support is needed. HLH should be considered in pregnant and puerperal women suffering from moderately severe to severe COVID-19 and presenting with: fever unresponsive to antibiotic therapy, cytopenia, hepatitis and hyperferritinaemia. The HLH disorder is rare and difficult to diagnose; however, its early detection could reduce patient mortality.


Assuntos
COVID-19/patologia , Síndrome da Liberação de Citocina/patologia , Linfo-Histiocitose Hemofagocítica/patologia , Complicações Infecciosas na Gravidez/patologia , COVID-19/complicações , COVID-19/imunologia , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/imunologia , Feminino , Humanos , Linfo-Histiocitose Hemofagocítica/etiologia , Linfo-Histiocitose Hemofagocítica/imunologia , Período Pós-Parto , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/virologia
5.
Arch Pathol Lab Med ; 145(11): 1328-1340, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34297794

RESUMO

CONTEXT.­: SARS-CoV-2 can undergo maternal-fetal transmission, heightening interest in the placental pathology findings from this infection. Transplacental SARS-CoV-2 transmission is typically accompanied by chronic histiocytic intervillositis together with necrosis and positivity of syncytiotrophoblast for SARS-CoV-2. Hofbauer cells are placental macrophages that have been involved in viral diseases, including HIV and Zika virus, but their involvement in SARS-CoV-2 is unknown. OBJECTIVE.­: To determine whether SARS-CoV-2 can extend beyond the syncytiotrophoblast to enter Hofbauer cells, endothelium, and other villous stromal cells in infected placentas of liveborn and stillborn infants. DESIGN.­: Case-based retrospective analysis by 29 perinatal and molecular pathology specialists of placental findings from a preselected cohort of 22 SARS-CoV-2-infected placentas delivered to pregnant women testing positive for SARS-CoV-2 from 7 countries. Molecular pathology methods were used to investigate viral involvement of Hofbauer cells, villous capillary endothelium, syncytiotrophoblast, and other fetal-derived cells. RESULTS.­: Chronic histiocytic intervillositis and trophoblast necrosis were present in all 22 placentas (100%). SARS-CoV-2 was identified in Hofbauer cells from 4 of 22 placentas (18.2%). Villous capillary endothelial staining was positive in 2 of 22 cases (9.1%), both of which also had viral positivity in Hofbauer cells. Syncytiotrophoblast staining occurred in 21 of 22 placentas (95.5%). Hofbauer cell hyperplasia was present in 3 of 22 placentas (13.6%). In the 7 cases having documented transplacental infection of the fetus, 2 (28.6%) occurred in placentas with Hofbauer cell staining positive for SARS-CoV-2. CONCLUSIONS.­: SARS-CoV-2 can extend beyond the trophoblast into the villous stroma, involving Hofbauer cells and capillary endothelial cells, in a small number of infected placentas. Most cases of SARS-CoV-2 transplacental fetal infection occur without Hofbauer cell involvement.


Assuntos
COVID-19/transmissão , COVID-19/virologia , Transmissão Vertical de Doenças Infecciosas , Macrófagos/virologia , Placenta/virologia , Complicações Infecciosas na Gravidez/virologia , SARS-CoV-2/patogenicidade , Adulto , COVID-19/imunologia , COVID-19/patologia , Proliferação de Células , Endotélio/patologia , Endotélio/virologia , Feminino , Humanos , Hiperplasia/patologia , Hiperplasia/virologia , Recém-Nascido , Macrófagos/patologia , Macrófagos/fisiologia , Masculino , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/patologia , Estudos Retrospectivos , SARS-CoV-2/imunologia , Natimorto , Trofoblastos/patologia , Trofoblastos/virologia
6.
J Med Life ; 14(2): 165-169, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34104238

RESUMO

Tuberculosis (TB) in pregnancy is not only a matter of the past; it is also a current problem. These days, TB appears through mass migration and tourism in countries where it was believed that this condition is eradicated. Adequate knowledge about the medical history of patients, risk factors, diagnosis and treatment of tuberculosis should be part of the armamentarium of each physician involved in clinical practice. TB is mainly found in urban and socially deprived areas. Due to the length of the treatment, there is an increased risk of drug resistance in partially treated patients. Strong knowledge about the history, risk factors, diagnosis and treatment of TB should be part of the armamentarium of each physician. Many practitioners are reluctant to request a chest X-ray in pregnancy due to the fear of harming the fetus. Bypassing a diagnosis can have a devastating effect on the mother and fetus, as well as their family and medical staff. This article discusses the matters of diagnosis and treatment of asymptomatic infection and active TB in pregnancy.


Assuntos
Complicações Infecciosas na Gravidez/patologia , Tuberculose/patologia , Farmacorresistência Bacteriana , Feminino , Humanos , Programas de Rastreamento , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico , Complicações Infecciosas na Gravidez/terapia , Fatores de Risco , Tuberculose/diagnóstico , Tuberculose/tratamento farmacológico , Tuberculose/terapia
7.
Rev. bras. ginecol. obstet ; 43(6): 474-479, June 2021. tab
Artigo em Inglês | LILACS | ID: biblio-1341139

RESUMO

Abstract Placental pathophysiology in SARS-CoV-2 infection can help researchers understand more about the infection and its impact on thematernal/neonatal outcomes. This brief review provides an overview about some aspects of the placental pathology in SARSCoV- 2 infection. In total, 11 papers were included. The current literature suggests that there are no specific histopathological characteristics in the placenta related to SARSCoV- 2 infection, but placentas frominfected women aremore likely to show findings of maternal and/or fetal malperfusion. The most common findings in placentas from infected women were fibrin deposition and intense recruitment of inflammatory infiltrates. The transplacental transmission of this virus is unlikely to occur, probably due to low expression of the receptor for SARS-CoV-2 in placental cell types. Further studies are needed to improve our knowledge about the interaction between the virus and the mother-fetus dyad and the impact on maternal and neonatal/fetal outcomes.


Resumo A fisiopatologia da placenta na infecção por SARS-CoV-2 pode ajudar os pesquisadores a entender mais sobre a infecção e seu impacto nos resultados maternos/neonatais. Esta revisão breve fornece uma visão geral sobre alguns aspectos da patologia placentária na infecção por SARS-CoV-2. Ao todo, 11 artigos foram incluídos. A literatura atual sugere que não há características histopatológicas específicas nas placentas relacionadas à infecção por SARS-CoV-2, mas as placentas de mulheres infectadas têm maior probabilidade de apresentar achados de má perfusão materna e/ou fetal. Os achados mais comuns em placentas de mulheres infectadas foram deposição de fibrina e intenso recrutamento de infiltrado inflamatório. A transmissão transplacentária deste vírus é improvável, devido à baixa expressão do receptor para SARS-CoV-2 em tipos de células da placenta. Mais estudos são necessários para melhorar nosso conhecimento sobre a interação entre o vírus e a díade mãe-feto e o impacto nos resultados maternos e neonatais/fetais.


Assuntos
Humanos , Feminino , Gravidez , Placenta/patologia , Complicações Infecciosas na Gravidez/patologia , COVID-19/patologia , Placenta/fisiopatologia , Placenta/irrigação sanguínea , Placenta/virologia , Complicações Infecciosas na Gravidez/fisiopatologia , Complicações Infecciosas na Gravidez/virologia , Transmissão Vertical de Doenças Infecciosas , COVID-19/fisiopatologia , COVID-19/virologia
8.
BMC Pregnancy Childbirth ; 21(1): 250, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33765949

RESUMO

BACKGROUND: Chorioamnionitis may cause serious perinatal and neonatal adverse outcomes, and group B streptococcus (GBS) is one of the most common bacteria isolated from human chorioamnionitis. The present study analyzed the impact of GBS infection and histological chorioamnionitis (HCA) on pregnancy outcomes and the diagnostic value of various biomarkers. METHODS: Pregnant women were grouped according to GBS infection and HCA detection. Perinatal and neonatal adverse outcomes were recorded with a follow-up period of 6 weeks. The white blood cell count (WBC), neutrophil ratio, and C-reactive protein (CRP) level from peripheral blood and soluble intercellular adhesion molecule-1 (sICAM-1), interleukin 8 (IL-8), and tumor necrosis factor α (TNF-α) levels from cord blood were assessed. RESULTS: A total of 371 pregnant women were included. Pregnant women with GBS infection or HCA had a higher risk of pathological jaundice and premature rupture of membranes and higher levels of sICAM-1, IL-8, and TNF-α in umbilical cord blood. Univariate and multivariate regression analysis revealed that sICMA-1, IL-8, TNF-α, WBC, and CRP were significantly related to an increased HCA risk. For all included pregnant women, TNF-α had the largest receiver operating characteristic (ROC) area (area: 0.841; 95% CI: 0.778-0.904) of the biomarkers analyzed. TNF-α still had the largest area under the ROC curve (area: 0.898; 95% CI: 0.814-0.982) for non-GBS-infected pregnant women, who also exhibited a higher neutrophil ratio (area: 0.815; 95% CI: 0.645-0.985) and WBC (area: 0.849; 95% CI: 0.72-0.978), but all biomarkers had lower value in the diagnosis of HCA in GBS-infected pregnant women. CONCLUSION: GBS infection and HCA correlated with several perinatal and neonatal adverse outcomes. TNF-α in cord blood and WBCs in peripheral blood had diagnostic value for HCA in non-GBS-infected pregnant women but not GBS-infected pregnant women.


Assuntos
Corioamnionite/diagnóstico , Complicações Infecciosas na Gravidez/diagnóstico , Nascimento Prematuro/epidemiologia , Infecções Estreptocócicas/diagnóstico , Streptococcus agalactiae/isolamento & purificação , Adulto , Biomarcadores/sangue , Estudos de Casos e Controles , Corioamnionite/sangue , Corioamnionite/microbiologia , Corioamnionite/patologia , Feminino , Sangue Fetal/química , Seguimentos , Humanos , Recém-Nascido , Contagem de Leucócitos , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/sangue , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/patologia , Resultado da Gravidez , Curva ROC , Medição de Risco/métodos , Infecções Estreptocócicas/sangue , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Fator de Necrose Tumoral alfa/sangue , Cordão Umbilical/patologia , Adulto Jovem
9.
Microbiol Mol Biol Rev ; 85(1)2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33568459

RESUMO

Bacteria in the genus Brucella are important human and veterinary pathogens. The abortion and infertility they cause in food animals produce economic hardships in areas where the disease has not been controlled, and human brucellosis is one of the world's most common zoonoses. Brucella strains have also been isolated from wildlife, but we know much less about the pathobiology and epidemiology of these infections than we do about brucellosis in domestic animals. The brucellae maintain predominantly an intracellular lifestyle in their mammalian hosts, and their ability to subvert the host immune response and survive and replicate in macrophages and placental trophoblasts underlies their success as pathogens. We are just beginning to understand how these bacteria evolved from a progenitor alphaproteobacterium with an environmental niche and diverged to become highly host-adapted and host-specific pathogens. Two important virulence determinants played critical roles in this evolution: (i) a type IV secretion system that secretes effector molecules into the host cell cytoplasm that direct the intracellular trafficking of the brucellae and modulate host immune responses and (ii) a lipopolysaccharide moiety which poorly stimulates host inflammatory responses. This review highlights what we presently know about how these and other virulence determinants contribute to Brucella pathogenesis. Gaining a better understanding of how the brucellae produce disease will provide us with information that can be used to design better strategies for preventing brucellosis in animals and for preventing and treating this disease in humans.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella/patogenicidade , Brucelose/patologia , Lipopolissacarídeos/imunologia , Sistemas de Secreção Tipo IV/metabolismo , Adaptação Fisiológica , Animais , Proteínas da Membrana Bacteriana Externa/genética , Brucella/genética , Brucella/imunologia , Feminino , Especificidade de Hospedeiro/genética , Humanos , Macrófagos/microbiologia , Gravidez , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/patologia , Trofoblastos/microbiologia , Sistemas de Secreção Tipo IV/genética , Virulência , Fatores de Virulência
10.
FASEB J ; 35(2): e21272, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33423320

RESUMO

Interleukin 33 (IL33) signaling has been implicated in the establishment and maintenance of pregnancy and in pregnancy disorders. The goal of this project was to evaluate the role of IL33 signaling in rat pregnancy. The rat possesses hemochorial placentation with deep intrauterine trophoblast invasion; features also characteristic of human placentation. We generated and characterized a germline mutant rat model for IL33 using CRISPR/Cas9 genome editing. IL33 deficient rats exhibited deficits in lung responses to an inflammatory stimulus (Sephadex G-200) and to estrogen-induced uterine eosinophilia. Female rats deficient in IL33 were fertile and exhibited pregnancy outcomes (gestation length and litter size) similar to wild-type rats. Placental weight was adversely affected by the disruption of IL33 signaling. A difference in pregnancy-dependent adaptations to lipopolysaccharide (LPS) exposure was observed between wild-type and IL33 deficient pregnancies. Pregnancy in wild-type rats treated with LPS did not differ significantly from pregnancy in vehicle-treated wild-type rats. In contrast, LPS treatment decreased fetal survival rate, fetal and placental weights, and increased fetal growth restriction in IL33 deficient rats. In summary, a new rat model for investigating IL33 signaling has been established. IL33 signaling participates in the regulation of placental development and protection against LPS-induced fetal and placental growth restriction.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Interleucina-33/metabolismo , Doenças Placentárias/metabolismo , Complicações Infecciosas na Gravidez/metabolismo , Transdução de Sinais , Animais , Feminino , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/patologia , Interleucina-33/genética , Lipopolissacarídeos/toxicidade , Mutação , Doenças Placentárias/etiologia , Doenças Placentárias/patologia , Gravidez , Complicações Infecciosas na Gravidez/etiologia , Complicações Infecciosas na Gravidez/patologia , Resultado da Gravidez , Ratos , Ratos Sprague-Dawley
11.
Mol Hum Reprod ; 27(2)2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33275139

RESUMO

Slit proteins have been reported to act as axonal repellents in Drosophila; however, their role in the placental microenvironment has not been explored. In this study, we found that human placental multipotent mesenchymal stromal cells (hPMSCs) constitutively express Slit2. Therefore, we hypothesized that Slit2 expressed by hPMSCs could be involved in macrophage migration during placental inflammation through membrane cognate Roundabout (Robo) receptor signaling. In order to develop a preclinical in vitro mouse model of hPMSCs in treatment of perinatal infection, RAW 264.7 cells were used in this study. Slit2 interacted with Robo4 that was highly expressed in RAW 264.7 macrophages: their interaction increased the adhesive ability of RAW 264.7 cells and inhibited migration. Lipopolysaccharide (LPS)-induced CD11bCD18 expression could be inhibited by Slit2 and by hPMSC-conditioned medium (CM). LPS-induced activation of p38 and Rap1 was also attenuated by Slit2 and by hPMSC-CM. Noticeably, these inhibitory effects of hPMSC-CM decreased after depletion of Slit2 from the CM. Furthermore, we found that p38 siRNA inhibited LPS-induced Rap1 expression in RAW 264.7 cells, indicating that Rap1 functions downstream of p38 signaling. p38 siRNA increased cell adhesion and inhibited migration through reducing LPS-stimulated CD11bCD18 expression in RAW 264.7 cells. Thus, hPMSC-derived Slit2 may inhibit LPS-induced CD11bCD18 expression to decrease cell migration and increase adhesion through modulating the activity and motility of inflammatory macrophages in placenta. This may represent a novel mechanism for LPS-induced placental infection.


Assuntos
Movimento Celular , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Placenta/metabolismo , Complicações Infecciosas na Gravidez/metabolismo , Animais , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Antígenos CD18/genética , Antígenos CD18/metabolismo , Adesão Celular , Movimento Celular/efeitos dos fármacos , Técnicas de Cocultura , Feminino , Humanos , Lipopolissacarídeos/toxicidade , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Camundongos , Comunicação Parácrina , Placenta/imunologia , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/patologia , Células RAW 264.7 , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rap1 de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/metabolismo
12.
Placenta ; 103: 94-103, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33120051

RESUMO

Placental macrophages are a heterogenous population of immune cells present throughout pregnancy. They are essential for maintenance of the homeostatic placenta environment and host defense against infections. The characterization of placental macrophages as well as their activation have been limited for a long time by the lack of convenient tools. The emergence of unbiased methods makes it possible to reappraise the study of placental macrophages. In this review, we discuss the diversity and the functions of placental macrophages to better understand their dysfunctions during placental infections.


Assuntos
Macrófagos/fisiologia , Placenta/citologia , Complicações Infecciosas na Gravidez/imunologia , Animais , Feminino , Humanos , Ativação de Macrófagos/fisiologia , Macrófagos/citologia , Macrófagos/patologia , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/patologia
13.
Probl Endokrinol (Mosk) ; 66(3): 56-61, 2020 09 16.
Artigo em Russo | MEDLINE | ID: mdl-33351339

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a virus that causes coronavirus disease in 2019 (COVID-19). Currently, there is no evidence that pregnant women are more vulnerable to COVID-19. All concerns and anticipated risks are related to the potential impact of COVID-19 on perinatal outcomes, so pregnant women require special attention in relation to the preventive measures, diagnosis and treatment of a new coronavirus disease. Women with gestational diabetes mellitus (GDM) belong to the group of high perinatal risk and need timely medical assistance. During the COVID-19 pandemic, there is a necessity in temporary changes of approaches to diagnosing GSD and pregnancy care before and after delivery in women with GSD. The purpose of our review is to present and analyze all available GSD screening recommendations, updated and published in various countries in response to the coronavirus pandemic, at the time of publication of this article. It seems that there is no single universal strategy to achieve a reasonable balance. In this regard, it is necessary to develop new national algorithms for GSD screening, taking into account both demographic factors and the features and capabilities of our health system. We believe that the knowledge and experience achieved as a result of these changes will lead to the revision and improvement of national and international recommendations.


Assuntos
COVID-19/diagnóstico , Diabetes Gestacional/diagnóstico , Pandemias , Complicações Infecciosas na Gravidez/diagnóstico , COVID-19/complicações , COVID-19/virologia , Diabetes Gestacional/patologia , Diabetes Gestacional/virologia , Feminino , Humanos , Programas de Rastreamento , Gravidez , Complicações Infecciosas na Gravidez/patologia , Complicações Infecciosas na Gravidez/virologia , SARS-CoV-2/patogenicidade
14.
J Clin Invest ; 130(11): 6171-6186, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33074247

RESUMO

Human T cell leukemia virus type 1 (HTLV-1) is mainly transmitted vertically through breast milk. The rate of mother-to-child transmission (MTCT) through formula feeding, although significantly lower than through breastfeeding, is approximately 2.4%-3.6%, suggesting the possibility of alternative transmission routes. MTCT of HTLV-1 might occur through the uterus, birth canal, or placental tissues; the latter is known as transplacental transmission. Here, we found that HTLV-1 proviral DNA was present in the placental villous tissues of the fetuses of nearly half of pregnant carriers and in a small number of cord blood samples. An RNA ISH assay showed that HTLV-1-expressing cells were present in nearly all subjects with HTLV-1-positive placental villous tissues, and their frequency was significantly higher in subjects with HTLV-1-positive cord blood samples. Furthermore, placental villous trophoblasts expressed HTLV-1 receptors and showed increased susceptibility to HTLV-1 infection. In addition, HTLV-1-infected trophoblasts expressed high levels of viral antigens and promoted the de novo infection of target T cells in a humanized mouse model. In summary, during pregnancy of HTLV-1 carriers, HTLV-1 was highly expressed in placental villous tissues, and villous trophoblasts showed high HTLV-1 sensitivity, suggesting that MTCT of HTLV-1 occurs through the placenta.


Assuntos
Infecções por HTLV-I/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Transmissão Vertical de Doenças Infecciosas , Complicações Infecciosas na Gravidez/metabolismo , Trofoblastos/metabolismo , Adulto , Células Cultivadas , Feminino , Infecções por HTLV-I/patologia , Infecções por HTLV-I/transmissão , Humanos , Gravidez , Complicações Infecciosas na Gravidez/patologia , Complicações Infecciosas na Gravidez/virologia , Trofoblastos/patologia , Trofoblastos/virologia
15.
AAPS J ; 22(6): 138, 2020 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-33099683

RESUMO

Treatment guidelines recommend continuation of combination antiretroviral therapy (cART) throughout pregnancy for all women living with human immunodeficiency virus (HIV). Many of these drugs are substrates of transporters expressed in the placenta and therefore play a role in fetal exposure. As placental transporters can be impacted by both HIV infection and drug therapy, our objective was to explore the impact of HIV infection and cART on transporter expression. Drug transporter expression was examined in human placental samples collected from women with HIV (n = 25) and from healthy HIV(-) controls (n = 23). The effect of exposure to drugs commonly used in cART during pregnancy was examined in vitro in placental villous explants obtained from healthy women. Gene expression was measured via qRT-PCR. Several ABC (ABCG2, ABCC1,2,4) and SLC (SLC21A9, SLC22A1,3,11) transporters were significantly downregulated in placentas isolated from HIV(+) women as compared with HIV(-) controls (p < 0.05-0.001), while ABCB1 and SLC21A12 were significantly upregulated (p < 0.001). Twenty-four to 48-h exposure of human placental explants to agents used in cART resulted in significant upregulation of ABCB1 and downregulation of SLC22A11. Our findings suggest that transplacental transport may be compromised during HIV infection due to altered expression of clinically important transporters. Furthermore, in vitro results indicate that cART imposes significant alterations in placental transporters but not all changes are consistent with findings in the placenta from HIV(+) women, indicating disease effects. As this may impact in utero-fetal exposure to clinically used medications, further studies are needed to determine the overall impact on maternal-fetal transfer.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Fármacos Anti-HIV/farmacocinética , Infecções por HIV/tratamento farmacológico , Transportadores de Ânions Orgânicos/metabolismo , Placenta/metabolismo , Complicações Infecciosas na Gravidez/tratamento farmacológico , Adulto , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/efeitos adversos , Regulação para Baixo , Quimioterapia Combinada , Feminino , Perfilação da Expressão Gênica , Infecções por HIV/diagnóstico , Infecções por HIV/patologia , Infecções por HIV/transmissão , HIV-1/isolamento & purificação , Humanos , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Masculino , Idade Materna , Troca Materno-Fetal , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico , Complicações Infecciosas na Gravidez/patologia , Técnicas de Cultura de Tecidos , Resultado do Tratamento , Regulação para Cima , Carga Viral
16.
Pediatr Ann ; 49(7): e305-e312, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32674168

RESUMO

Mycoplasma species (spp.) can be commensals or opportunistic pathogens of the urogenital tract, and they can be commonly isolated from amniotic fluid, placenta, and fetal/neonatal tissue or blood in mothers delivering prematurely or their preterm infants. Although the presence of Mycoplasma spp. has been associated with adverse maternal-fetal outcomes such as preterm birth and maternal chorioamnionitis, it is less clear whether vertical transmission to the neonate results in colonization or active infection/inflammation. Moreover, the presence of Mycoplasma spp. in neonatal blood, cerebrospinal fluid, or tissue has been variably associated with increased risk of neonatal comorbidities, especially bronchopulmonary dysplasia (BPD). Although the treatment of the mother or neonate with antibiotics is effective in eradicating ureaplasma, it is not clear that the treatment is effective in reducing the incidence of major morbidities of the preterm neonate (eg, BPD). In this article, we review the animal and clinical data for ureaplasma-related complications and treatment strategies. [Pediatr Ann. 2020;49(7):e305-e312.].


Assuntos
Antibacterianos/uso terapêutico , Infecções por Mycoplasma/tratamento farmacológico , Infecções por Mycoplasma/patologia , Complicações Infecciosas na Gravidez/tratamento farmacológico , Complicações Infecciosas na Gravidez/patologia , Infecções por Ureaplasma/tratamento farmacológico , Infecções por Ureaplasma/patologia , Animais , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Transmissão Vertical de Doenças Infecciosas , Infecções por Mycoplasma/diagnóstico , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico , Nascimento Prematuro , Infecções por Ureaplasma/diagnóstico
17.
Syst Biol Reprod Med ; 66(3): 151-169, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32482148

RESUMO

The placenta works as a selective barrier, protecting the fetus from potential infections that may affect the maternal organism during pregnancy. In this review, we will discuss several challenging infections that are common within Latin American countries and that may affect the maternal-fetal interface and pose risks to fetal development. Specifically, we will focus on emerging infectious diseases including the arboviruses, malaria, leishmaniasis, and the bacterial foodborne disease caused by Shiga toxin-producing Escherichia coli. We will also highlight some topics of interest currently being studied by research groups that comprise an international effort aimed at filling the knowledge gaps in this field. These topics address the relationship between exposure to microorganisms and placental abnormalities, congenital anomalies, and complications of pregnancy. ABBREVIATIONS: ADE: antibody-dependent enhancement; CCL2: monocyte chemoattractant protein-1; CCL3: macrophage inflammatory protein-1 α; CCL5: chemokine (C-C motif) ligand 5; CHIKV: chikungunya virus; DCL: diffuse cutaneous leishmaniasis; DENV: dengue virus; Gb3: glycolipid globotriaosylceramyde; HIF: hypoxia-inducible factor; HUS: hemolytic uremic syndrome; IFN: interferon; Ig: immunoglobulins; IL: interleukin; IUGR: intrauterine growth restriction; LCL: localized cutaneous leishmaniasis; LPS: lipopolysaccharid; MCL: mucocutaneous leishmaniasis; NO: nitric oxide; PCR: polymerase chain reaction; PGF: placental growth factor; PM: placental malaria; RIVATREM: Red Iberoamericana de Alteraciones Vasculares em transtornos del Embarazo; sVEGFR: soluble vascular endothelial growth factor receptor; STEC: shiga toxin-producing Escherichia coli; stx: shiga toxin protein; TNF: tumor necrosis factor; TOAS: T cell original antigenic sin; Var2CSA: variant surface antigen 2-CSA; VEGF: vascular endothelial growth factor; VL: visceral leishmaniasis; WHO: world health organization; YFV: yellow fever virus; ZIKV: Zika virus.


Assuntos
Doenças Placentárias/etiologia , Placenta/patologia , Complicações Infecciosas na Gravidez/patologia , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/microbiologia , Feminino , Humanos , América Latina , Leishmaniose/complicações , Malária/complicações , Doenças Placentárias/patologia , Gravidez , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/virologia , Saúde Pública , Escherichia coli Shiga Toxigênica , Doenças Vasculares/complicações , Viroses/complicações
18.
Methods Mol Biol ; 2142: 181-195, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32367368

RESUMO

Pigs are highly relevant to model human in utero Zika virus (ZIKV) infection because both species have similar physiology, genetics, immunity, fetal brain development, and postnatal brain growth. The virus causes persistent in utero infection and replicates in the fetal brain, fetal membranes, and placenta. Subclinical persistent in utero infection in mid-gestation also increases interferon alpha (IFN-α) levels in fetal blood plasma and amniotic fluid. Moreover, we demonstrated altered IFN-α responses in porcine offspring affected with subclinical in utero ZIKV infection. Elevated levels of in utero type I interferons were suggested to play a role in fetal pathology. Thus, the porcine model may provide an understanding of ZIKV-induced immunopathology in fetuses and sequelae in offspring, which is important for the development of targeted interventions. Here, we describe surgery, ultrasound-guided in utero injection, postoperative monitoring, sampling, and cytokine testing protocols.


Assuntos
Modelos Animais de Doenças , Doenças Fetais , Interferon-alfa/metabolismo , Complicações Infecciosas na Gravidez , Suínos , Infecção por Zika virus , Líquido Amniótico/metabolismo , Animais , Análise Química do Sangue/métodos , Análise Química do Sangue/veterinária , Feminino , Sangue Fetal/metabolismo , Doenças Fetais/diagnóstico por imagem , Doenças Fetais/metabolismo , Doenças Fetais/patologia , Doenças Fetais/virologia , Fetoscopia/métodos , Fetoscopia/veterinária , Injeções , Interferon-alfa/análise , Interferon-alfa/sangue , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico por imagem , Complicações Infecciosas na Gravidez/metabolismo , Complicações Infecciosas na Gravidez/patologia , Ultrassonografia de Intervenção/métodos , Ultrassonografia de Intervenção/veterinária , Ultrassonografia Pré-Natal/métodos , Ultrassonografia Pré-Natal/veterinária , Zika virus/fisiologia , Infecção por Zika virus/diagnóstico por imagem , Infecção por Zika virus/metabolismo , Infecção por Zika virus/patologia
19.
Methods Mol Biol ; 2142: 235-249, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32367371

RESUMO

The 2016 Zika virus (ZIKV) outbreak in the Americas has been characterized by an increased association frequency of fetal neuropathological abnormalities. To have a comprehensive and accurate knowledge of key elements of the clinically observed neurologic dysfunctions in Zika-infected babies, ZIKV transmission from mother to fetus needs to be deeply studied. Thus, it is important to determine the role of both virus-targeted cells and tissues within the mother-fetus interface. Cellular tropism and mechanisms of ZIKV transmission from mother to the fetus during early pregnancy still remain unknown on many aspects. To improve the characterization of the maternal-fetal ZIKV transmission, we have set up an ex vivo model using an organ culture approach with a light-invasive sampling from the first trimester of pregnancy samples. Thus, here we provide evidence that circulating epidemic ZIKV strains from Latin America widely target and destroy reproductive tissues, including the decidua basalis, fetal placenta, and umbilical cord. In addition, we show that ZIKV is able to differentially replicate in a large range of both maternal and fetal cells, including decidual fibroblasts and macrophages, fetal trophoblast and Hofbauer cells, as well as umbilical cord mesenchymal stem cells. This primary and broad ZIKV cellular tropism and the resulting abundant cytopathic-induced tissue effects during the first trimester of pregnancy show the upstream path of clinically observed congenital damages.


Assuntos
Complicações Infecciosas na Gravidez/patologia , Primeiro Trimestre da Gravidez , Infecção por Zika virus/patologia , Zika virus/patogenicidade , Animais , Chlorocebus aethiops , Feminino , Feto/patologia , Feto/virologia , Humanos , Transmissão Vertical de Doenças Infecciosas , Macrófagos/patologia , Macrófagos/virologia , Especificidade de Órgãos , Placenta/patologia , Placenta/virologia , Gravidez , Complicações Infecciosas na Gravidez/metabolismo , Complicações Infecciosas na Gravidez/virologia , Cultura Primária de Células/métodos , Manejo de Espécimes/métodos , Distribuição Tecidual , Trofoblastos/patologia , Trofoblastos/virologia , Cordão Umbilical/patologia , Cordão Umbilical/virologia , Células Vero , Carga Viral/métodos , Cultura de Vírus/métodos , Zika virus/metabolismo , Infecção por Zika virus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA