Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Hepatol ; 79(4): 924-932, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37290591

RESUMEN

BACKGROUND & AIMS: Current therapy for chronic hepatitis B virus (cHBV) infection involves lifelong treatment. New treatments that enable HBV functional cure would represent a clinically meaningful advance. ALN-HBV and VIR-2218 are investigational RNA interference therapeutics that target all major HBV transcripts. METHODS: We report on: i) the safety of single doses of VIR-2218 (modified from ALN-HBV by enhanced stabilization chemistry plus technology to reduce off-target, seed-mediated binding while maintaining on-target antiviral activity) and ALN-HBV in humanized mice; ii) a cross-study comparison of the safety of single doses of VIR-2218 and ALN-HBV in healthy human volunteers (n = 24 and n = 49, respectively); and iii) the antiviral activity of two doses of 20, 50, 100, 200 mg of VIR-2218 (total n = 24) vs. placebo (n = 8), given 4 weeks apart, in participants with cHBV infection. RESULTS: In humanized mice, alanine aminotransferase (ALT) levels were markedly lower following administration of VIR-2218 compared with ALN-HBV. In healthy volunteers, post-treatment ALT elevations occurred in 28% of participants receiving ALN-HBV compared with none in those receiving VIR-2218. In participants with cHBV infection, VIR-2218 was associated with dose-dependent reductions in hepatitis B surface antigen (HBsAg). The greatest mean reduction of HBsAg at Week 20 in participants receiving 200 mg was 1.65 log IU/ml. The HBsAg reduction was maintained at 0.87 log IU/ml at Week 48. No participants had serum HBsAg loss or hepatitis B surface antibody seroconversion. CONCLUSIONS: VIR-2218 demonstrated an encouraging hepatic safety profile in preclinical and clinical studies as well as dose-dependent HBsAg reductions in patients with cHBV infection. These data support future studies with VIR-2218 as part of combination regimens with a goal of HBV functional cure. TRIAL REGISTRATION: ClinicalTrials.gov identifiers: NCT02826018 and NCT03672188. IMPACT AND IMPLICATIONS: A significant unmet need exists for therapies for chronic HBV (cHBV) infection that achieve functional cure. We report clinical and non-clinical data on two investigational small-interfering RNAs that target HBx, ALN-HBV and VIR-2218, demonstrating that incorporation of enhanced stabilization chemistry plus technology in VIR-2218 reduces its propensity to cause ALT elevations relative to its parent compound, ALN-HBV. We also show that VIR-2218 reduces hepatitis B surface antigen levels in a dose-dependent manner in participants with cHBV infection. These studies support the continued development of VIR-2218 as part of therapeutic regimens for cHBV infection, with the goal of a functional cure, and are important for HBV researchers and physicians.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Humanos , Animales , Ratones , Hepatitis B Crónica/tratamiento farmacológico , Virus de la Hepatitis B , Antígenos de Superficie de la Hepatitis B , Tratamiento con ARN de Interferencia , Ensayos Clínicos Controlados Aleatorios como Asunto , Antivirales , ADN Viral , Antígenos e de la Hepatitis B , Hepatitis B/tratamiento farmacológico
2.
PLoS Pathog ; 14(5): e1007094, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29847585

RESUMEN

During pregnancy, the placenta protects the fetus against the maternal immune response, as well as bacterial and viral pathogens. Bacterial pathogens that have evolved specific mechanisms of breaching this barrier, such as Listeria monocytogenes, present a unique opportunity for learning how the placenta carries out its protective function. We previously identified the L. monocytogenes protein Internalin P (InlP) as a secreted virulence factor critical for placental infection. Here, we show that InlP, but not the highly similar L. monocytogenes internalin Lmo2027, binds to human afadin (encoded by AF-6), a protein associated with cell-cell junctions. A crystal structure of InlP reveals several unique features, including an extended leucine-rich repeat (LRR) domain with a distinctive Ca2+-binding site. Despite afadin's involvement in the formation of cell-cell junctions, MDCK epithelial cells expressing InlP displayed a decrease in the magnitude of the traction stresses they could exert on deformable substrates, similar to the decrease in traction exhibited by AF-6 knock-out MDCK cells. L. monocytogenes ΔinlP mutants were deficient in their ability to form actin-rich protrusions from the basal face of polarized epithelial monolayers, a necessary step in the crossing of such monolayers (transcytosis). A similar phenotype was observed for bacteria expressing an internal in-frame deletion in inlP (inlP ΔLRR5) that specifically disrupts its interaction with afadin. However, afadin deletion in the host cells did not rescue the transcytosis defect. We conclude that secreted InlP targets cytosolic afadin to specifically promote L. monocytogenes transcytosis across the basal face of epithelial monolayers, which may contribute to the crossing of the basement membrane during placental infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Basal/microbiología , Listeria monocytogenes/patogenicidad , Proteínas de Microfilamentos/metabolismo , Complicaciones Infecciosas del Embarazo/metabolismo , Animales , Femenino , Feto/microbiología , Humanos , Listeriosis/metabolismo , Proteínas de la Membrana/metabolismo , Placenta/metabolismo , Placenta/microbiología , Embarazo , Factores de Virulencia/metabolismo
3.
Infect Immun ; 86(6)2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29483290

RESUMEN

Intrauterine infections lead to serious complications for mother and fetus, including preterm birth, maternal and fetal death, and neurological sequelae in the surviving offspring. Improving maternal and child heath is a global priority. Yet, the development of strategies to prevent and treat pregnancy-related diseases has lagged behind progress made in other medical fields. One of the challenges is finding tractable model systems that replicate the human maternal-fetal interface. Animal models offer the ability to study pathogenesis and host defenses in vivo However, the anatomy of the maternal-fetal interface is highly divergent across species. While many tools are available to study host responses in the pregnant mouse model, other animals have placentas that are more similar to that of humans. Here we describe new developments in animal and human tissue models to investigate the pathogenesis of listeriosis at the maternal-fetal interface. We highlight gaps in existing knowledge and make recommendations on how they can be filled.


Asunto(s)
Listeria monocytogenes/fisiología , Listeriosis/transmisión , Modelos Biológicos , Complicaciones Infecciosas del Embarazo/microbiología , Técnicas de Cultivo de Tejidos , Animales , Femenino , Humanos , Listeriosis/microbiología , Intercambio Materno-Fetal , Placenta/microbiología , Embarazo
4.
Infect Immun ; 86(11)2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30104213

RESUMEN

Listeria monocytogenes is a foodborne pathogen that infects the placenta and can cause pregnancy complications. Listeriosis usually occurs as a sporadic infection, but large outbreaks are also reported. Virulence from clinical isolates is rarely analyzed due to the large number of animals required, but this knowledge could help guide the response to an outbreak. We implemented a DNA barcode system using signature tags that allowed us to efficiently assay variations in virulence across a large number of isolates. We tested 77 signature-tagged clones of clinical L. monocytogenes strains from 72 infected human placentas and 5 immunocompromised patients, all of which were isolated since 2000. These strains were tested for virulence in a modified competition assay in comparison to that of the laboratory strain 10403S. We used two in vivo models of listeriosis: the nonpregnant mouse and the pregnant guinea pig. Strains that were frequently found at a high abundance within infected organs were considered hypervirulent, while strains frequently found at a low abundance were considered hypovirulent. Virulence split relatively evenly among hypovirulent strains, hypervirulent strains, and strains as virulent as 10403S. The laboratory strain was found to have an intermediate virulence phenotype, supporting its suitability for use in pathogenesis studies. Further, we found that splenic virulence and placental virulence are closely linked in both the guinea pig and mouse models. This suggests that outbreak and sporadic pregnancy-associated L. monocytogenes strains are not generally more virulent than lab reference strains. However, some strains did show consistent and reproducible virulence differences, suggesting that their further study may reveal deeper insights into the biological underpinnings of listeriosis.


Asunto(s)
Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Listeriosis/patología , Complicaciones Infecciosas del Embarazo/microbiología , Complicaciones Infecciosas del Embarazo/patología , Factores de Virulencia/análisis , Estructuras Animales/microbiología , Animales , Modelos Animales de Enfermedad , Femenino , Cobayas , Humanos , Listeria monocytogenes/genética , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/aislamiento & purificación , Ratones , Placenta/microbiología , Embarazo , Virulencia , Factores de Virulencia/genética
5.
Infect Immun ; 85(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28507070

RESUMEN

The bacterial pathogen Listeria monocytogenes causes foodborne systemic disease in pregnant women, which can lead to preterm labor, stillbirth, or severe neonatal disease. Colonization of the maternal decidua appears to be an initial step in the maternal component of the disease as well as bacterial transmission to the placenta and fetus. Host-pathogen interactions in the decidua during this early stage of infection remain poorly understood. Here, we assessed the dynamics of L. monocytogenes infection in primary human decidual organ cultures and in the murine decidua in vivo A high inoculum was necessary to infect both human and mouse deciduas, and the data support the existence of a barrier to initial colonization of the murine decidua. If successful, however, colonization in both species was followed by significant bacterial expansion associated with an inability of the decidua to mount appropriate innate cellular immune responses. The innate immune deficits included the failure of bacterial foci to attract macrophages and NK cells, cell types known to be important for early defenses against L. monocytogenes in the spleen, as well as a decrease in the tissue density of inflammatory Ly6Chi monocytes in vivo These results suggest that the infectivity of the decidua is not the result of an enhanced recruitment of L. monocytogenes to the gestational uterus but rather is due to compromised local innate cellular immune responses.


Asunto(s)
Decidua/microbiología , Interacciones Huésped-Patógeno , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Animales , Antígenos Ly/inmunología , Decidua/inmunología , Femenino , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Listeriosis/transmisión , Macrófagos/inmunología , Ratones , Monocitos/microbiología , Técnicas de Cultivo de Órganos , Placenta/inmunología , Placenta/microbiología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/microbiología , Bazo/inmunología , Bazo/microbiología
6.
Infect Immun ; 84(12): 3584-3596, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27736782

RESUMEN

Intrauterine infection is a major detriment for maternal-child health and occurs despite local mechanisms that protect the maternal-fetal interface from a wide variety of pathogens. The bacterial pathogen Listeria monocytogenes causes spontaneous abortion, stillbirth, and preterm labor in humans and serves as a model for placental pathogenesis. Given the unique immunological environment of the maternal-fetal interface, we hypothesized that virulence determinants with placental tropism are required for infection of this tissue. We performed a genomic screen in pregnant guinea pigs that led to the identification of 201 listerial genes important for infection of the placenta but not maternal liver. Among these genes was lmrg1778 (lmo2470), here named inlP, predicted to encode a secreted protein that belongs to the internalin family. InlP is conserved in virulent L. monocytogenes strains but absent in Listeria species that are nonpathogenic for humans. The intracellular life cycle of L. monocytogenes deficient in inlP (ΔinlP) was not impaired. In guinea pigs and mice, InlP increased the placental bacterial burden by a factor of 3 log10 while having only a minor role in other maternal organs. Furthermore, the ΔinlP strain was attenuated in intracellular growth in primary human placental organ cultures and trophoblasts. InlP is a novel virulence factor for listeriosis with a strong tropism for the placenta. This virulence factor represents a tool for the development of new modalities to prevent and treat infection-related pregnancy complications.


Asunto(s)
Proteínas Bacterianas/metabolismo , Listeria monocytogenes/metabolismo , Listeriosis/microbiología , Placenta/microbiología , Complicaciones Infecciosas del Embarazo/microbiología , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Cobayas , Ratones , Movimiento , Embarazo , Factores de Virulencia/genética
7.
PLoS Pathog ; 9(12): e1003821, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24348256

RESUMEN

Fetal syncytiotrophoblasts form a unique fused multinuclear surface that is bathed in maternal blood, and constitutes the main interface between fetus and mother. Syncytiotrophoblasts are exposed to pathogens circulating in maternal blood, and appear to have unique resistance mechanisms against microbial invasion. These are due in part to the lack of intercellular junctions and their receptors, the Achilles heel of polarized mononuclear epithelia. However, the syncytium is immune to receptor-independent invasion as well, suggesting additional general defense mechanisms against infection. The difficulty of maintaining and manipulating primary human syncytiotrophoblasts in culture makes it challenging to investigate the cellular and molecular basis of host defenses in this unique tissue. Here we present a novel system to study placental pathogenesis using murine trophoblast stem cells (mTSC) that can be differentiated into syncytiotrophoblasts and recapitulate human placental syncytium. Consistent with previous results in primary human organ cultures, murine syncytiotrophoblasts were found to be resistant to infection with Listeria monocytogenes via direct invasion and cell-to-cell spread. Atomic force microscopy of murine syncytiotrophoblasts demonstrated that these cells have a greater elastic modulus than mononuclear trophoblasts. Disruption of the unusually dense actin structure--a diffuse meshwork of microfilaments--with Cytochalasin D led to a decrease in its elastic modulus by 25%. This correlated with a small but significant increase in invasion of L. monocytogenes into murine and human syncytium. These results suggest that the syncytial actin cytoskeleton may form a general barrier against pathogen entry in humans and mice. Moreover, murine TSCs are a genetically tractable model system for the investigation of specific pathways in syncytial host defenses.


Asunto(s)
Células Gigantes/microbiología , Listeria monocytogenes/crecimiento & desarrollo , Listeriosis/inmunología , Placenta/citología , Placenta/microbiología , Complicaciones Infecciosas del Embarazo/inmunología , Animales , Fenómenos Biofísicos/inmunología , Células Cultivadas , Femenino , Células Gigantes/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Transmisión Vertical de Enfermedad Infecciosa , Listeria monocytogenes/inmunología , Listeriosis/microbiología , Ratones , Ratones Endogámicos C57BL , Placenta/inmunología , Embarazo , Complicaciones Infecciosas del Embarazo/microbiología , Trofoblastos/citología , Trofoblastos/inmunología , Trofoblastos/microbiología , Células U937
8.
PLoS Pathog ; 7(3): e1002005, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21408203

RESUMEN

Listeria monocytogenes is a facultative intracellular bacterial pathogen that can infect the placenta, a chimeric organ made of maternal and fetal cells. Extravillous trophoblasts (EVT) are specialized fetal cells that invade the uterine implantation site, where they come into direct contact with maternal cells. We have shown previously that EVT are the preferred site of initial placental infection. In this report, we infected primary human EVT with L. monocytogenes. EVT eliminated ∼80% of intracellular bacteria over 24-hours. Bacteria were unable to escape into the cytoplasm and remained confined to vacuolar compartments that became acidified and co-localized with LAMP1, consistent with bacterial degradation in lysosomes. In human placental organ cultures bacterial vacuolar escape rates differed between specific trophoblast subpopulations. The most invasive EVT-those that would be in direct contact with maternal cells in vivo-had lower escape rates than trophoblasts that were surrounded by fetal cells and tissues. Our results suggest that EVT present a bottleneck in the spread of L. monocytogenes from mother to fetus by inhibiting vacuolar escape, and thus intracellular bacterial growth. However, if L. monocytogenes is able to spread beyond EVT it can find a more hospitable environment. Our results elucidate a novel aspect of the maternal-fetal barrier.


Asunto(s)
Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/patogenicidad , Placenta/citología , Placenta/microbiología , Trofoblastos/citología , Trofoblastos/microbiología , Células Cultivadas , Femenino , Feto/microbiología , Técnica del Anticuerpo Fluorescente , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Lisosomas/metabolismo , Microscopía Electrónica de Transmisión , Embarazo , Técnicas de Cultivo de Tejidos , Útero/microbiología , Vacuolas/microbiología
9.
Infect Immun ; 80(1): 418-28, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22083708

RESUMEN

Toxoplasma gondii is a ubiquitous, obligate intracellular parasite capable of crossing the placenta to cause spontaneous abortion, preterm labor, or significant disease in the surviving neonate. Exploration of the cellular and histological components of the placental barrier is in its infancy, and both how and where T. gondii breaches it are unknown. The human placenta presents two anatomical interfaces between maternal cells and fetal cells (trophoblasts): (i) the villous region where maternal blood bathes syncytialized trophoblasts for nutrient exchange and (ii) the maternal decidua, where mononuclear, extravillous trophoblasts anchor the villous region to the uterus. Using first-trimester human placental explants, we demonstrate that the latter site is significantly more vulnerable to infection, despite presenting a vastly smaller surface. This is consistent with past findings concerning two vertically transmitted viruses and one bacterium. We further explore whether three genetically distinct T. gondii types (I, II, and III) are capable of preferential placental infection and survival in this model. We find no difference in these strains' ability to infect placental explants; however, slightly slower growth is evident in type II (Prugniaud [Pru]) parasites relative to other cell types, although this did not quite achieve statistical significance.


Asunto(s)
Placenta/inmunología , Toxoplasma/inmunología , Toxoplasma/patogenicidad , Toxoplasmosis/inmunología , Femenino , Humanos , Modelos Biológicos , Técnicas de Cultivo de Órganos , Placenta/anatomía & histología , Placenta/citología , Embarazo
10.
Infect Immun ; 80(2): 720-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22083714

RESUMEN

Listeria monocytogenes causes a serious food-borne disease due to its ability to spread from the intestine to other organs, a process that is poorly understood. In this study we used 20 signature-tagged wild-type clones of L. monocytogenes in guinea pigs in combination with extensive quantitative data analysis to gain insight into extraintestinal dissemination. We show that L. monocytogenes colonized the liver in all asymptomatic animals. Spread to the liver occurred as early as 4 h after ingestion via a direct pathway from the intestine to the liver. This direct pathway contributed significantly to the bacterial load in the liver and was followed by a second wave of dissemination via the mesenteric lymph nodes (indirect pathway). Furthermore, bacteria were eliminated in the liver, whereas small intestinal villi provided a niche for bacterial replication, indicating organ-specific differences in net bacterial growth. Bacteria were shed back from intestinal villi into the small intestinal lumen and reinfected the Peyer's patches. Together, these results support a novel dissemination model where L. monocytogenes replicates in intestinal villi, is shed into the lumen, and reinfects intestinal immune cells that traffic to liver and mesenteric lymph nodes, a process that occurs even during asymptomatic colonization.


Asunto(s)
Intestinos/microbiología , Listeria monocytogenes , Listeriosis/microbiología , Hígado/microbiología , Ganglios Linfáticos/microbiología , Animales , Carga Bacteriana , Femenino , Cobayas , Listeriosis/patología , Bazo/microbiología , Factores de Tiempo
11.
PLoS Pathog ; 6(1): e1000732, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-20107601

RESUMEN

Listeria monocytogenes is an important cause of maternal-fetal infections and serves as a model organism to study these important but poorly understood events. L. monocytogenes can infect non-phagocytic cells by two means: direct invasion and cell-to-cell spread. The relative contribution of each method to placental infection is controversial, as is the anatomical site of invasion. Here, we report for the first time the use of first trimester placental organ cultures to quantitatively analyze L. monocytogenes infection of the human placenta. Contrary to previous reports, we found that the syncytiotrophoblast, which constitutes most of the placental surface and is bathed in maternal blood, was highly resistant to L. monocytogenes infection by either internalin-mediated invasion or cell-to-cell spread. Instead, extravillous cytotrophoblasts-which anchor the placenta in the decidua (uterine lining) and abundantly express E-cadherin-served as the primary portal of entry for L. monocytogenes from both extracellular and intracellular compartments. Subsequent bacterial dissemination to the villous stroma, where fetal capillaries are found, was hampered by further cellular and histological barriers. Our study suggests the placenta has evolved multiple mechanisms to resist pathogen infection, especially from maternal blood. These findings provide a novel explanation why almost all placental pathogens have intracellular life cycles: they may need maternal cells to reach the decidua and infect the placenta.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa , Listeriosis/transmisión , Enfermedades Placentarias/microbiología , Trofoblastos/microbiología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Listeria monocytogenes , Microscopía Confocal , Técnicas de Cultivo de Órganos , Embarazo , Complicaciones Infecciosas del Embarazo/microbiología
13.
PLoS Pathog ; 2(6): e66, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16846254

RESUMEN

Infection with Listeria monocytogenes is a significant health problem during pregnancy. This study evaluates the role of trafficking between maternal organs and placenta in a pregnant guinea pig model of listeriosis. After intravenous inoculation of guinea pigs, the initial ratio of bacteria in maternal organs to placenta was 10(3)-10(4):1. Rapid increase of bacteria in the placenta changed the ratio to 1:1 after 24 h. Utilizing two wild-type strains, differentially marked by their susceptibility to erythromycin, we found that only a single bacterium was necessary to cause placental infection, and that L. monocytogenes trafficked from maternal organs to the placenta in small numbers. Surprisingly, bacteria trafficked in large numbers from the placenta to maternal organs. Bacterial growth, clearance, and transport between organs were simulated with a mathematical model showing that the rate of bacterial clearance relative to the rate of bacterial replication in the placenta was sufficient to explain the difference in the course of listeriosis in pregnant versus nonpregnant animals. These results provide the basis for a new model where the placenta is relatively protected from infection. Once colonized, the placenta becomes a nidus of infection resulting in massive reseeding of maternal organs, where L. monocytogenes cannot be cleared until trafficking is interrupted by expulsion of the infected placental tissues.


Asunto(s)
Traslocación Bacteriana/fisiología , Listeria monocytogenes/fisiología , Listeriosis/fisiopatología , Hígado/microbiología , Placenta/microbiología , Bazo/microbiología , Animales , Antibacterianos/uso terapéutico , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Eritromicina/uso terapéutico , Femenino , Gentamicinas/uso terapéutico , Cobayas , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/patogenicidad , Listeriosis/tratamiento farmacológico , Listeriosis/patología , Listeriosis/transmisión , Hígado/fisiopatología , Modelos Teóricos , Placenta/fisiopatología , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/fisiopatología , Preñez/fisiología , Bazo/fisiopatología
14.
J Vis Exp ; (113)2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27500727

RESUMEN

The placenta shows a large degree of interspecies anatomic variability. To best understand biology and pathophysiology of the human placenta, it is imperative to design experiments using human cells and tissues. An advantage of organ culture is maintenance of three-dimensional (3D) structural organization and extracellular matrix. The goal of the method described here is successful establishment of ex vivo human gestational tissue organ cultures and their healthy culture maintenance for 72-96 hr. The protocol details the immediate processing of research-consented, placental and decidual specimens fresh from the operating suite. These are abundant specimens that would otherwise be discarded. Detailed instructions on the sterile collection of these samples, including morphologic details on how to select appropriate tissues to establish 3D organ cultures, is provided. Placental villous and decidual tissues are microdissected into 2-3 mm(3) pieces and placed separately on matrix-lined transwell filters and cultured for several days. Villous and decidual organ cultures are well suited for the study of human host-pathogen interaction. As compared to other model organisms, these human cultures are particularly advantageous to examine mechanism of infection for pathogens that demonstrate variable patterns of host specificity. As an example, we demonstrate infection of placental and decidual organ cultures with the clinically relevant, facultative intracellular bacterial pathogen Listeria monocytogenes.


Asunto(s)
Decidua/fisiología , Técnicas de Cultivo de Órganos , Placenta/fisiología , Femenino , Interacciones Huésped-Patógeno , Humanos , Listeria monocytogenes , Embarazo
15.
Fetal Matern Med Rev ; 25(3-4): 332-338, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27695204

RESUMEN

In this article we present a novel model for how the human placenta might get infected via the hematogenous route. We present a list of diverse placental pathogens, like Listeria monocytogenes or Cytomegalovirus, which are familiar to most obstetricians, but others, like Salmonella typhi, have only been reported in case studies or small case series. Remarkably, all of these organisms on this list are either obligate or facultative intracellular organisms. These pathogens are able to enter and survive inside host immune cells for at least a portion of their life cycle. We suggest that many blood-borne pathogens might arrive at the placenta via transportation inside of maternal leukocytes that enter the decidua in early pregnancy. We discuss mechanisms by which extravillous trophoblasts could get infected in the decidua and spread infection to other layers in the placenta. We hope to raise awareness among OB/GYN clinicians that organisms not typically associated with the TORCH list might cause placental infections and pregnancy complications.

16.
Case Rep Med ; 2013: 973297, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24459469

RESUMEN

We report a case in which placental abruption occurred at 16 weeks following first trimester diagnosis and treatment for typhoid fever. Unexpectedly Salmonella enterica serovar Typhi (S. Typhi) was found in fetal tissues at autopsy. Using information from the murine model of typhoid fever in pregnancy, we draw parallels between S. Typhi and L. monocytogenes to develop a plausible hypothesis to explain how this organism was able to cross the placenta in the first trimester to cause abruption, inflammation, and expulsion of the fetus and placenta. We hope that this model for understanding placental infections by the hematogenous route helps to raise awareness that organisms not typically associated with TORCH infection can nevertheless cause placental infection and pregnancy loss.

17.
Curr Opin Microbiol ; 15(1): 36-43, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22169833

RESUMEN

Placental infections are major causes of maternal and fetal disease. This review introduces a new paradigm for placental infections based on current knowledge of placental defenses and how this barrier can be breached. Transmission of pathogens from mother to fetus can occur at two sites of direct contact between maternal cells and specialized fetal cells (trophoblasts) in the human placenta: firstly, maternal immune and endothelial cells juxtaposed to extravillous trophoblasts in the uterine implantation site and secondly, maternal blood surrounding the syncytiotrophoblast (SYN). Recent findings suggest that the primary vulnerability is in the implantation site. We explore evidence that the placental SYN evolved as a defense against pathogens, and that inflammation-mediated spontaneous abortion may benefit mother and pathogen.


Asunto(s)
Infecciones Bacterianas/inmunología , Placenta/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/microbiología , Complicaciones Parasitarias del Embarazo/inmunología , Complicaciones Parasitarias del Embarazo/parasitología , Infecciones por Protozoos/inmunología , Aborto Séptico/inmunología , Aborto Séptico/microbiología , Aborto Séptico/parasitología , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Femenino , Humanos , Placenta/microbiología , Placenta/parasitología , Embarazo , Complicaciones Infecciosas del Embarazo/parasitología , Infecciones por Protozoos/parasitología , Infecciones por Protozoos/patología , Trofoblastos/inmunología
18.
Curr Biol ; 22(13): 1228-34, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22658590

RESUMEN

During human pregnancy, placental trophoblasts differentiate and syncytialize into syncytiotrophoblasts that sustain progesterone production [1]. This process is accompanied by mitochondrial fragmentation and cristae remodeling [2], two facets of mitochondrial apoptosis, whose molecular mechanisms and functional consequences on steroidogenesis are unclear. Here we show that the mitochondria-shaping protein Optic atrophy 1 (Opa1) controls efficiency of steroidogenesis. During syncytialization of trophoblast BeWo cells, levels of the profission mitochondria-shaping protein Drp1 increase, and those of Opa1 and mitofusin (Mfn) decrease, leading to mitochondrial fragmentation and cristae remodeling. Manipulation of the levels of Opa1 reveal an inverse relationship with the efficiency of steroidogenesis in trophoblasts and in mouse embryonic fibroblasts where the mitochondrial steroidogenetic pathway has been engineered. In an in vitro assay, accumulation of cholesterol is facilitated in the inner membrane of isolated mitochondria lacking Opa1. Thus, Opa1-dependent inner membrane remodeling controls efficiency of steroidogenesis.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Mitocondrias/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , Animales , Línea Celular , Colesterol/metabolismo , Dinaminas , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , GTP Fosfohidrolasas/genética , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/metabolismo , Pregnenolona/biosíntesis
19.
J Clin Invest ; 120(4): 1016-25, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20364099

RESUMEN

The placenta provides critical transport functions between the maternal and fetal circulations during intrauterine development. Formation of this interface relies on coordinated interactions among transcriptional, epigenetic, and environmental factors. Here we describe these mechanisms in the context of the differentiation of placental cells (trophoblasts) and synthesize current knowledge about how they interact to generate a functional placenta. Developing an understanding of these pathways contributes to an improvement of our models for studying trophoblast biology and sheds light on the etiology of pregnancy complications and the in utero programming of adult diseases.


Asunto(s)
Desarrollo Embrionario , Epigénesis Genética , Placenta/fisiología , Transcripción Genética , Animales , Comunicación Celular , Linaje de la Célula , Metilación de ADN , Femenino , Histonas/metabolismo , Humanos , Intercambio Materno-Fetal , Embarazo , Útero/fisiología
20.
J Infect Dis ; 191(11): 1889-97, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15871123

RESUMEN

Listeria monocytogenes causes foodborne outbreaks that lead to infection in human and other mammalian fetuses. To elucidate the molecular and cellular mechanisms involved in transplacental transmission, we characterized placental-fetal infection in pregnant guinea pigs inoculated with wild-type (wt) or mutant L. monocytogenes strains. The wt strain increased in number in the placenta by >1000-fold during the first 24 h after inoculation--an increase that was unparalleled in other maternal organs. The ActA- mutant, which is impaired in cell-to-cell spread and attenuated in maternal organs, increased in the placenta by a similar amount, although, in fetal infection, the number of ActA- mutant bacteria was 100-fold lower, compared with that of the wt strain. Furthermore, a mutant impaired in vacuolar escape was rapidly eliminated from maternal organs but persisted in the placenta. We concluded that cell-to-cell spread facilitates maternal-to-fetal transmission. Furthermore, the placenta provides a protective niche for growth of L. monocytogenes.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa , Listeria monocytogenes/patogenicidad , Listeriosis/transmisión , Placenta/microbiología , Complicaciones Infecciosas del Embarazo/microbiología , Animales , Femenino , Cobayas , Riñón/microbiología , Listeria monocytogenes/genética , Listeria monocytogenes/crecimiento & desarrollo , Listeriosis/microbiología , Hígado/microbiología , Pulmón/microbiología , Mutación , Embarazo , Bazo/microbiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA