Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 11(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37487664

RESUMEN

BACKGROUND: Cancer immunotherapies are generally effective in patients whose tumors contain a priori primed T-cells reactive to tumor antigens (TA). One approach to prime TA-reactive T-cells is to administer immunostimulatory molecules, cells, or pathogens directly to the tumor site, that is, in situ vaccination (ISV). We recently described an ISV using Flt3L to expand and recruit dendritic cells (DC), radiotherapy to load DC with TA, and pattern recognition receptor agonists (PRRa) to activate TA-loaded DC. While ISV trials using synthetic PRRa have yielded systemic tumor regressions, the optimal method to activate DCs is unknown. METHODS: To discover optimal DC activators and increase access to clinical grade reagents, we assessed whether viral or bacterial components found in common pathogen vaccines are an effective source of natural PRRa (naPRRa). Using deep profiling (155-metric) of naPRRa immunomodulatory effects and gene editing of specific PRR, we defined specific signatures and molecular mechanisms by which naPRRa potentiate T-cell priming. RESULTS: We observed that vaccine naPRRa can be even more potent in activating Flt3L-expanded murine and human DCs than synthetic PRRa, promoting cross-priming of TA-reactive T-cells. We developed a mechanistically diverse naPRRa combination (BCG, PedvaxHIB, Rabies) and noted more potent T-cell cross-priming than with any single naPRRa. The naPRRa triplet-as part of Flt3L-primed ISV-induced greater intratumoral CD8 T-cell infiltration, T-cells reactive to a newly defined tumorous neoantigen, durable tumor regressions. CONCLUSIONS: This work provides rationale for the translation of pathogen vaccines as FDA-approved clinical-grade DC activators which could be exploited as immune-stimulants for early phase trials.


Asunto(s)
Linfocitos T CD8-positivos , Reactividad Cruzada , Humanos , Animales , Ratones , Vacunación , Edición Génica , Inmunización
2.
Nat Commun ; 13(1): 7149, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36418317

RESUMEN

Immunotherapies directly enhancing anti-tumor CD8+ T cell responses have yielded measurable but limited success, highlighting the need for alternatives. Anti-tumor T cell responses critically depend on antigen presenting dendritic cells (DC), and enhancing mobilization, antigen loading and activation of these cells represent an attractive possibility to potentiate T cell based therapies. Here we show that expansion of DCs by Flt3L administration impacts in situ vaccination with oncolytic Newcastle Disease Virus (NDV). Mechanistically, NDV activates DCs and sensitizes them to dying tumor cells through upregulation of dead-cell receptors and synergizes with Flt3L to promote anti-tumor CD8+ T cell cross-priming. In vivo, Flt3L-NDV in situ vaccination induces parallel amplification of virus- and tumor-specific T cells, including CD8+ T cells reactive to newly-described neoepitopes, promoting long-term tumor control. Cross-presenting conventional Type 1 DCs are indispensable for the anti-tumor, but not anti-viral, T cell response, and type I IFN-dependent CD4+ Th1 effector cells contribute to optimal anti-tumor immunity. These data demonstrate that mobilizing DCs to increase tumor antigen cross-presentation improves oncolytic virotherapy and that neoepitope-specific T cells can be induced without individualized, ex vivo manufactured vaccines.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Vacunas , Animales , Linfocitos T CD8-positivos , Células Dendríticas , Reactividad Cruzada , Antígenos de Neoplasias , Neoplasias/metabolismo , Vacunas/metabolismo
3.
Nat Cancer ; 3(8): 911-926, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35999309

RESUMEN

After several decades, therapeutic cancer vaccines now show signs of efficacy and potential to help patients resistant to other standard-of-care immunotherapies, but they have yet to realize their full potential and expand the oncologic armamentarium. Here, we classify cancer vaccines by what is known of the included antigens, which tumors express those antigens and where the antigens colocalize with antigen-presenting cells, thus delineating predefined vaccines (shared or personalized) and anonymous vaccines (ex vivo or in situ). To expedite clinical development, we highlight the need for accurate immune monitoring of early trials to acknowledge failures and advance the most promising vaccines.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Células Presentadoras de Antígenos , Vacunas contra el Cáncer/uso terapéutico , Humanos , Factores Inmunológicos , Inmunoterapia/efectos adversos , Neoplasias/prevención & control , Neoplasias/terapia
4.
Epigenetics ; 17(9): 1040-1055, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34605719

RESUMEN

Epigenetics may play a central, yet unexplored, role in the profound changes that the maternal immune system undergoes during pregnancy and could be involved in the pregnancy-induced modulation of several autoimmune diseases. We investigated changes in the methylome in isolated circulating CD4+ T-cells in non-pregnant and pregnant women, during the 1st and 2nd trimester, using the Illumina Infinium Human Methylation 450K array, and explored how these changes were related to autoimmune diseases that are known to be affected during pregnancy. Pregnancy was associated with several hundreds of methylation differences, particularly during the 2nd trimester. A network-based modular approach identified several genes, e.g., CD28, FYN, VAV1 and pathways related to T-cell signalling and activation, highlighting T-cell regulation as a central component of the observed methylation alterations. The identified pregnancy module was significantly enriched for disease-associated methylation changes related to multiple sclerosis, rheumatoid arthritis and systemic lupus erythematosus. A negative correlation between pregnancy-associated methylation changes and disease-associated changes was found for multiple sclerosis and rheumatoid arthritis, diseases that are known to improve during pregnancy whereas a positive correlation was found for systemic lupus erythematosus, a disease that instead worsens during pregnancy. Thus, the directionality of the observed changes is in line with the previously observed effect of pregnancy on disease activity. Our systems medicine approach supports the importance of the methylome in immune regulation of T-cells during pregnancy. Our findings highlight the relevance of using pregnancy as a model for understanding and identifying disease-related mechanisms involved in the modulation of autoimmune diseases.Abbreviations: BMIQ: beta-mixture quantile dilation; DMGs: differentially methylated genes; DMPs: differentially methylated probes; FE: fold enrichment; FDR: false discovery rate; GO: gene ontology; GWAS: genome-wide association studies; MDS: multidimensional scaling; MS: multiple sclerosis; PBMC: peripheral blood mononuclear cells; PBS: phosphate buffered saline; PPI; protein-protein interaction; RA: rheumatoid arthritis; SD: standard deviation; SLE: systemic lupus erythematosus; SNP: single nucleotide polymorphism; TH: CD4+ T helper cell; VIStA: diVIsive Shuffling Approach.


Asunto(s)
Artritis Reumatoide , Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Esclerosis Múltiple , Enfermedades Autoinmunes/genética , Antígenos CD28/genética , Linfocitos T CD4-Positivos , Metilación de ADN , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Leucocitos Mononucleares , Lupus Eritematoso Sistémico/genética , Esclerosis Múltiple/genética , Fosfatos , Embarazo , Linfocitos T
5.
Cancer Discov ; 11(3): 599-613, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33334730

RESUMEN

T cell-based therapies have induced cancer remissions, though most tumors ultimately progress, reflecting inherent or acquired resistance including antigen escape. Better understanding of how T cells eliminate tumors will help decipher resistance mechanisms. We used a CRISPR/Cas9 screen and identified a necessary role for Fas-FasL in antigen-specific T-cell killing. We also found that Fas-FasL mediated off-target "bystander" killing of antigen-negative tumor cells. This localized bystander cytotoxicity enhanced clearance of antigen-heterogeneous tumors in vivo, a finding that has not been shown previously. Fas-mediated on-target and bystander killing was reproduced in chimeric antigen receptor (CAR-T) and bispecific antibody T-cell models and was augmented by inhibiting regulators of Fas signaling. Tumoral FAS expression alone predicted survival of CAR-T-treated patients in a large clinical trial (NCT02348216). These data suggest strategies to prevent immune escape by targeting both the antigen expression of most tumor cells and the geography of antigen-loss variants. SIGNIFICANCE: This study demonstrates the first report of in vivo Fas-dependent bystander killing of antigen-negative tumors by T cells, a phenomenon that may be contributing to the high response rates of antigen-directed immunotherapies despite tumoral heterogeneity. Small molecules that target the Fas pathway may potentiate this mechanism to prevent cancer relapse.This article is highlighted in the In This Issue feature, p. 521.


Asunto(s)
Citotoxicidad Inmunológica , Inmunoterapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor fas/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Efecto Espectador/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Edición Génica , Ingeniería Genética , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Ratones , Ratones Noqueados , Neoplasias/etiología , Neoplasias/terapia , Receptores Quiméricos de Antígenos , Especificidad del Receptor de Antígeno de Linfocitos T , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Discov ; 9(11): 1520-1537, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31375522

RESUMEN

T-cell transfer into lymphodepleted recipients induces homeostatic activation and potentiates antitumor efficacy. In contrast to canonical T-cell receptor-induced activation, homeostatic activation yields a distinct phenotype and memory state whose regulatory mechanisms are poorly understood. Here, we show in patients and murine models that, following transfer into lymphodepleted bone marrow transplant (BMT) recipients, CD8+ T cells undergo activation but also simultaneous homeostatic inhibition manifested by upregulation of immune-checkpoint molecules and functional suppression. T cells transferred into BMT recipients were protected from homeostatic inhibition by PD-1/CTLA4 dual checkpoint blockade (dCB). This combination of dCB and BMT-"immunotransplant"-increased T-cell homeostatic activation and antitumor T-cell responses by an order of magnitude. Like homeostatic activation, homeostatic inhibition is IL7/IL15-dependent, revealing mechanistic coupling of these two processes. Marked similarity in ex vivo modulation of post-BMT T cells in mice and patients is promising for the clinical translation of immunotransplant (NCT03305445) and for addressing homeostatic inhibition in T-cell therapies. SIGNIFICANCE: For optimal anticancer effect, T-cell therapies including chimeric antigen receptor T-cell, tumor-infiltrating lymphocyte, and transgenic T-cell therapies require transfer into lymphodepleted recipients and homeostatic activation; however, concomitant homeostatic inhibition mitigates T-cell therapies' efficacy. Checkpoint blockade uncouples homeostatic inhibition from activation, amplifying T-cell responses. Conversely, tumors nonresponsive to checkpoint blockade or BMT are treatable with immunotransplant.See related commentary by Ansell, p. 1487.This article is highlighted in the In This Issue feature, p. 1469.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Trasplante de Médula Ósea/métodos , Antígeno CTLA-4/antagonistas & inhibidores , Neoplasias/terapia , Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Terapia Combinada , Femenino , Homeostasis , Humanos , Huésped Inmunocomprometido/efectos de los fármacos , Inmunoterapia , Masculino , Ratones , Neoplasias/inmunología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Clin Cancer Res ; 25(21): 6283-6294, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31123052

RESUMEN

Immunotherapies such as checkpoint blockade have achieved durable benefits for patients with advanced stage cancer and have changed treatment paradigms. However, these therapies rely on a patient's own a priori primed tumor-specific T cells, limiting their efficacy to a subset of patients. Because checkpoint blockade is most effective in patients with inflamed or "hot" tumors, a priority in the field is learning how to "turn cold tumors hot." Inflammation is generally initiated by innate immune cells, which receive signals through pattern recognition receptors (PRR)-a diverse family of receptors that sense conserved molecular patterns on pathogens, alarming the immune system of an invading microbe. Their immunostimulatory properties can reprogram the immune suppressive tumor microenvironment and activate antigen-presenting cells to present tumors antigens, driving de novo tumor-specific T-cell responses. These features, among others, make PRR-targeting therapies an attractive strategy in immuno-oncology. Here, we discuss mechanisms of PRR activation, highlighting ongoing clinical trials and recent preclinical advances focused on therapeutically targeting PRRs to treat cancer.


Asunto(s)
Inmunoterapia , Inflamación/terapia , Neoplasias/terapia , Receptores de Reconocimiento de Patrones/genética , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/microbiología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Humanos , Inflamación/inmunología , Inflamación/microbiología , Estadificación de Neoplasias , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/microbiología , Receptores de Reconocimiento de Patrones/uso terapéutico , Linfocitos T/inmunología , Linfocitos T/microbiología , Microambiente Tumoral/inmunología
8.
Nat Med ; 25(5): 814-824, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962585

RESUMEN

Indolent non-Hodgkin's lymphomas (iNHLs) are incurable with standard therapy and are poorly responsive to checkpoint blockade. Although lymphoma cells are efficiently killed by primed T cells, in vivo priming of anti-lymphoma T cells has been elusive. Here, we demonstrate that lymphoma cells can directly prime T cells, but in vivo immunity still requires cross-presentation. To address this, we developed an in situ vaccine (ISV), combining Flt3L, radiotherapy, and a TLR3 agonist, which recruited, antigen-loaded and activated intratumoral, cross-presenting dendritic cells (DCs). ISV induced anti-tumor CD8+ T cell responses and systemic (abscopal) cancer remission in patients with advanced stage iNHL in an ongoing trial ( NCT01976585 ). Non-responding patients developed a population of PD1+CD8+ T cells after ISV, and murine tumors became newly responsive to PD1 blockade, prompting a follow-up trial of the combined therapy. Our data substantiate that recruiting and activating intratumoral, cross-priming DCs is achievable and critical to anti-tumor T cell responses and PD1-blockade efficacy.


Asunto(s)
Vacunas contra el Cáncer , Linfoma de Células B/terapia , Adulto , Anciano , Animales , Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Carboximetilcelulosa de Sodio/análogos & derivados , Carboximetilcelulosa de Sodio/uso terapéutico , Línea Celular Tumoral , Terapia Combinada , Células Dendríticas/inmunología , Femenino , Humanos , Inmunoterapia Adoptiva , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Masculino , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Poli I-C/uso terapéutico , Polilisina/análogos & derivados , Polilisina/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Receptor Toll-Like 3/agonistas , Vacunación
9.
Sci Rep ; 8(1): 4166, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29520033

RESUMEN

Low molecular weight heparin (LMWH) is widely used in recurrent miscarriage treatment. The anti-coagulant effects are established, while immunological effects are not fully known. Our aim was to assess LMWH effects on activation and polarization of central regulatory immune cells from healthy women, and on placenta tissues from women undergoing elective abortions. Isolated blood monocytes and T helper (Th) cells under different activation and polarizing conditions were cultured with or without LMWH. Flow cytometry showed that LMWH exposure induced increased expression of HLA-DR and CD206 in macrophages. This phenotype was associated with increased secretion of Th17-associated CCL20, and decreased secretion of CCL2 (M2-associated) and CCL22 (Th2), as measured by multiplex bead array. In accordance, LMWH exposure to Th cells reduced the proportion of CD25highFoxp3+ regulatory T-cells, intensified IFN-γ secretion and showed a tendency to increase the lymphoblast proportions. Collectively, a mainly pro-inflammatory effect was noted on two essential tolerance-promoting cells. Although the biological significancies of these in vitro findings are uncertain and need to be confirmed in vivo, they suggest the possibility that immunological effects of LMWH may be beneficial mainly at an earlier gestational age to provide an appropriate implantation process in women with recurrent miscarriage.


Asunto(s)
Citocinas/inmunología , Heparina de Bajo-Peso-Molecular/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Macrófagos/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Aborto Habitual/inmunología , Aborto Habitual/patología , Adolescente , Adulto , Implantación del Embrión/inmunología , Femenino , Humanos , Macrófagos/patología , Persona de Mediana Edad , Embarazo , Linfocitos T Reguladores/patología , Células Th17/patología
10.
Am J Reprod Immunol ; 74(4): 368-78, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26118401

RESUMEN

PROBLEM: A deficiency in regulatory T (Treg) cells causing reduced immune regulatory capacity has been proposed in preeclampsia. OBJECTIVE: Utilizing recent advances in flow cytometry phenotyping, we aimed to assess whether a deficiency of Treg subpopulations occurs in preeclampsia. METHOD OF STUDY: Six-color flow cytometry was used for Treg phenotyping in 18 preeclamptic women (one early-onset, one severe and 16 both), 20 women with normal pregnancy, and 20 non-pregnant controls. RESULTS: No differences were found in major Treg populations including CD127(low) CD25(+) /CD127(ow) FOXP3(+) , resting (FOXP3(dim) CD45RA(+) ), and activated (FOXP3(bright) CD45RA(-) ) Treg cells, whereas preeclamptic women showed increased CTLA-4(+) and CCR4(+) proportions within resting/activated Treg populations. Corticosteroid treatment prior to blood sampling (n = 10) affected the distribution of Treg populations. CONCLUSIONS: Although we found no major alterations in circulating Treg frequencies, differences in CTLA-4(+) and CCR4(+) frequencies suggest a migratory defect of Treg cells in preeclampsia. Corticosteroid treatment should be taken into account when evaluating Treg cells.


Asunto(s)
Antígeno CTLA-4/metabolismo , Preeclampsia/inmunología , Receptores CCR4/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Corticoesteroides/uso terapéutico , Adulto , Recuento de Linfocito CD4 , Femenino , Citometría de Flujo , Humanos , Embarazo , Adulto Joven
11.
J Immunol ; 194(4): 1534-44, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25560409

RESUMEN

A successful pregnancy requires that the maternal immune system is instructed to a state of tolerance to avoid rejection of the semiallogeneic fetal-placental unit. Although increasing evidence supports that decidual (uterine) macrophages and regulatory T cells (Tregs) are key regulators of fetal tolerance, it is not known how these tolerogenic leukocytes are induced. In this article, we show that the human fetal placenta itself, mainly through trophoblast cells, is able to induce homeostatic M2 macrophages and Tregs. Placental-derived M-CSF and IL-10 induced macrophages that shared the CD14(+)CD163(+)CD206(+)CD209(+) phenotype of decidual macrophages and produced IL-10 and CCL18 but not IL-12 or IL-23. Placental tissue also induced the expansion of CD25(high)CD127(low)Foxp3(+) Tregs in parallel with increased IL-10 production, whereas production of IFN-γ (Th1), IL-13 (Th2), and IL-17 (Th17) was not induced. Tregs expressed the suppressive markers CTLA-4 and CD39, were functionally suppressive, and were induced, in part, by IL-10, TGF-ß, and TRAIL. Placental-derived factors also limited excessive Th cell activation, as shown by decreased HLA-DR expression and reduced secretion of Th1-, Th2-, and Th17-associated cytokines. Thus, our data indicate that the fetal placenta has a central role in promoting the homeostatic environment necessary for successful pregnancy. These findings have implications for immune-mediated pregnancy complications, as well as for our general understanding of tissue-induced tolerance.


Asunto(s)
Tolerancia Inmunológica/inmunología , Macrófagos/inmunología , Placenta/inmunología , Embarazo/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Ensayo de Inmunoadsorción Enzimática , Femenino , Feto/inmunología , Citometría de Flujo , Homeostasis/inmunología , Humanos , Inmunohistoquímica , Técnicas In Vitro , Placenta/citología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
12.
Am J Reprod Immunol ; 74(2): 100-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25582625

RESUMEN

A successful pregnancy requires that the maternal immune system adapts properly to avoid rejection of the semi-allogeneic fetus without compromising the ability to protect the mother and the fetus against infections. In this review, we describe the role of decidual macrophages in creating a homeostatic environment at the fetal-maternal interface. We also discuss their role in pregnancy complications as well as future possibilities to modulate macrophage function therapeutically. Decidual macrophages are enriched at the fetal-maternal interface and play a major role in the regulation of inflammatory responses and the maintenance of a tolerant environment. Their function is, however, not restricted to immune tolerance, but extends to include functions such as the recognition and clearance of infections, the clearance of apoptotic debris, and tissue remodeling. Decidual macrophages seem to largely function as tissue-resident macrophages that are crucial for maintaining homeostasis and reproductive success.


Asunto(s)
Decidua/inmunología , Feto/inmunología , Macrófagos/inmunología , Embarazo/inmunología , Animales , Femenino , Homeostasis , Humanos , Trofoblastos/inmunología
13.
Toxicol Pathol ; 42(2): 339-44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23531795

RESUMEN

This short review is derived from the peer-reviewed literature and the experience and case materials of the authors. Brief illustrated summaries are presented on the gross and histologic normal anatomy of rodent and macaque placentas, including typical organ weights, with comments on differences from the human placenta. Common incidental findings, background lesions, and induced toxic lesions are addressed, and a recommended strategy for pathologic evaluation of placentas is provided.


Asunto(s)
Placenta/patología , Animales , Femenino , Histocitoquímica , Humanos , Patología , Placenta/química , Embarazo , Toxicología
14.
Toxicol Pathol ; 42(2): 327-38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23531796

RESUMEN

During pregnancy, the maternal immune system is challenged by the semiallogeneic fetus, which must be tolerated without compromising fetal or maternal health. This review updates the systemic and local immune changes taking place during human pregnancy, including some examples in rodents. Systemic changes are induced by contact of maternal blood with placental factors and include enhanced innate immunity with increased activation of granulocytes and nonclassical monocytes. Although a bias toward T helper (Th2) and regulatory T cell (Treg) immunity has been associated with healthy pregnancy, the relationship between different circulating Th cell subsets is not straightforward. Instead, these adaptations appear most evidently at the fetal-maternal interface, where for instance Tregs are enriched and promote fetal tolerance. Also innate immune cells, that is, natural killer cells and macrophages, are enriched, constituting the majority of decidual leukocytes. These cells not only contribute to immune regulation but also aid in establishing the placenta by promoting trophoblast recruitment and angiogenesis. Thus, proper interaction between leukocytes and placental trophoblasts is necessary for normal placentation and immune adaptation. Consequently, spontaneous maladaptation or interference of the immune system with toxic substances may be important contributing factors for the development of pregnancy complications such as preeclampsia, preterm labor, and recurrent miscarriages.


Asunto(s)
Placenta/inmunología , Inmunidad Adaptativa/inmunología , Animales , Femenino , Humanos , Inmunidad Innata/inmunología , Intercambio Materno-Fetal/inmunología , Embarazo
15.
Toxicol Pathol ; 42(2): 345-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23548605

RESUMEN

This review summarizes the potential and also some limitations of using human placentas, or placental cells and structures for toxicology testing. The placenta contains a wide spectrum of cell types and tissues, such as trophoblast cells, immune cells, fibroblasts, stem cells, endothelial cells, vessels, glands, membranes, and many others. It may be expected that in many cases the relevance of results obtained from human placenta will be higher than those from animal models due to species specificity of metabolism and placental structure. For practical and economical reasons, we propose to apply a battery of sequential experiments for analysis of potential toxicants. This should start with using cell lines, followed by testing placenta tissue explants and isolated placenta cells, and finally by application of single and dual side ex vivo placenta perfusion. With each of these steps, the relative workload increases while the number of feasible repeats decreases. Simultaneously, the predictive power enhances by increasing similarity with in vivo human conditions. Toxic effects may be detected by performing proliferation, vitality and cell death assays, analysis of protein and hormone expression, immunohistochemistry or testing functionality of signaling pathways, gene expression, transport mechanisms, and so on. When toxic effects appear at any step, the subsequent assays may be cancelled. Such a system may be useful to reduce costs and increase specificity in testing questionable toxicants. Nonetheless, it requires further standardization and end point definitions for better comparability of results from different toxicants and to estimate the respective in vivo translatability and predictive value.


Asunto(s)
Placenta/citología , Placenta/efectos de los fármacos , Pruebas de Toxicidad/métodos , Femenino , Humanos , Embarazo
16.
Toxicol Pathol ; 42(2): 314-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23548606

RESUMEN

The immune system represents a key defense mechanism against potential pathogens and adverse non-self materials. During pregnancy, the placenta is the point of contact between the maternal organism and non-self proteins of the fetal allograft and hence undoubtedly fulfils immune functions. In the placenta bacteria, foreign (non-self) proteins and proteins that might be introduced in toxicological studies or by medication are barred from reaching the progeny, and the maternal immune system is primed for acceptance of non-maternal fetal protein. Both immunologic protection of the fetus and acceptance of the fetus by the mother require effective mechanisms to prevent an immunologic fetomaternal conflict and to keep both organisms in balance. This is why the placenta requires toxicological consideration in view of its immune organ function. The following articles deal with placenta immune-, control-, and tolerance mechanisms in view of both fetal and maternal aspects. Furthermore, models for experimental access to placental immune function are addressed and the pathological evaluation is elucidated. "The Placenta as an Immune Organ and Its Relevance in Toxicological Studies" was subject of a continuing education course at the 2012 Society of Toxicologic Pathology meeting held in Boston, MA.


Asunto(s)
Macaca fascicularis , Modelos Animales , Placenta/inmunología , Placenta/metabolismo , Animales , Femenino , Histocitoquímica , Tolerancia Inmunológica , Placenta/anatomía & histología , Embarazo , Toxicología/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...