Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(31): e2404229121, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39052836

RESUMEN

The distinct human leukocyte antigen (HLA) class I expression pattern of human extravillous trophoblasts (EVT) endows them with unique tolerogenic properties that enable successful pregnancy. Nevertheless, how this process is elaborately regulated remains elusive. Previously, E74 like ETS transcription factor 3 (ELF3) was identified to govern high-level HLA-C expression in EVT. In the present study, ELF3 is found to bind to the enhancer region of two adjacent NOD-like receptor (NLR) genes, NLR family pyrin domain-containing 2 and 7 (NLRP2, NLRP7). Notably, our analysis of ELF3-deficient JEG-3 cells, a human choriocarcinoma cell line widely used to study EVT biology, suggests that ELF3 transactivates NLRP7 while suppressing the expression of NLRP2. Moreover, we find that NLRP2 and NLRP7 have opposing effects on HLA-C expression, thus implicating them in immune evasion at the maternal-fetal interface. We confirmed that NLRP2 suppresses HLA-C levels and described a unique role for NLRP7 in promoting HLA-C expression in JEG-3. These results suggest that these two NLR genes, which arose via gene duplication in primates, are fine-tuned by ELF3 yet have acquired divergent functions to enable proper expression levels of HLA-C in EVT, presumably through modulating the degradation kinetics of IkBα. Targeting the ELF3-NLRP2/NLRP7-HLA-C axis may hold therapeutic potential for managing pregnancy-related disorders, such as recurrent hydatidiform moles and fetal growth restriction, and thus improve placental development and pregnancy outcomes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Trofoblastos Extravellosos , Antígenos HLA-C , Trofoblastos , Femenino , Humanos , Embarazo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Regulación de la Expresión Génica , Antígenos HLA-C/metabolismo , Antígenos HLA-C/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Trofoblastos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo
2.
J Reprod Immunol ; 163: 104244, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38555747

RESUMEN

Implantation and maintenance of pregnancy involve intricate immunological processes that enable the developing fetus to coexist with the maternal immune system. Progesterone, a critical hormone during pregnancy, is known to promote immune tolerance and prevent preterm labor. However, the mechanism by which progesterone mediates these effects remains unclear. In this study, we investigated the role of the non-classical progesterone receptor membrane component 1 (PGRMC1) in progesterone signaling at the maternal-fetal interface. Using JEG3 cells, a trophoblast model cell line, we observed that progesterone stimulation increased the expression of human leukocyte antigen-C (HLA-C) and HLA-G, key molecules involved in immune tolerance. We also found that progesterone upregulated the expression of the transcription factor ELF3, which is known to regulate trophoblast-specific HLA-C expression. Interestingly, JEG3 cells lacked expression of classical progesterone receptors (PRs) but exhibited high expression of PGRMC1, a finding we confirmed in primary trophoblasts by mining sc-RNA seq data from human placenta. To investigate the role of PGRMC1 in progesterone signaling, we used CRISPR/Cas9 technology to knockout PGRMC1 in JEG3 cells. PGRMC1-deficient cells showed a diminished response to progesterone stimulation. Furthermore, we found that the progesterone antagonist RU486 inhibited ELF3 expression in a PGRMC1-dependent manner, suggesting that RU486 acts as a progesterone antagonist by competing for receptor binding. Additionally, we found that RU486 inhibited cell invasion, an important process for successful pregnancy, and this inhibitory effect was dependent on PGRMC1. Our findings highlight the crucial role of PGRMC1 in mediating the immunoregulatory effects of progesterone at the maternal-fetal interface.


Asunto(s)
Proteínas de la Membrana , Progesterona , Receptores de Progesterona , Trofoblastos , Humanos , Receptores de Progesterona/metabolismo , Femenino , Embarazo , Progesterona/metabolismo , Progesterona/farmacología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Trofoblastos/metabolismo , Trofoblastos/inmunología , Placenta/inmunología , Placenta/metabolismo , Transducción de Señal/inmunología , Intercambio Materno-Fetal/inmunología , Implantación del Embrión/inmunología
3.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34785597

RESUMEN

Zika virus (ZIKV) during pregnancy infects fetal trophoblasts and causes placental damage and birth defects including microcephaly. Little is known about the anti-ZIKV cellular immune response at the maternal-fetal interface. Decidual natural killer cells (dNK), which directly contact fetal trophoblasts, are the dominant maternal immune cells in the first-trimester placenta, when ZIKV infection is most hazardous. Although dNK express all the cytolytic molecules needed to kill, they usually do not kill infected fetal cells but promote placentation. Here, we show that dNK degranulate and kill ZIKV-infected placental trophoblasts. ZIKV infection of trophoblasts causes endoplasmic reticulum (ER) stress, which makes them dNK targets by down-regulating HLA-C/G, natural killer (NK) inhibitory receptor ligands that help maintain tolerance of the semiallogeneic fetus. ER stress also activates the NK activating receptor NKp46. ZIKV infection of Ifnar1 -/- pregnant mice results in high viral titers and severe intrauterine growth restriction, which are exacerbated by depletion of NK or CD8 T cells, indicating that killer lymphocytes, on balance, protect the fetus from ZIKV by eliminating infected cells and reducing the spread of infection.


Asunto(s)
Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Feto/inmunología , Antígenos HLA-C , Tolerancia Inmunológica , Ratones , Placenta/inmunología , Placentación , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Receptores KIR
4.
Immunology ; 163(3): 338-343, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33565605

RESUMEN

Humanized double transgenic mice express both HLA-DR15 (the MHC gene linked to MS) and TCR.Ob1A12 from a multiple sclerosis patient (that recognizes MBP85-99 presented by HLA-DR15), yet they fail to develop autoimmune encephalomyelitis quickly, although 5-10% develop disease at 12 months. These mice were found to express large numbers of IL-10-secreting splenocytes as early as 4 weeks of age. These regulatory T cells appeared spontaneously without prior immunization with the autoantigen MBP85-99. They were of murine origin and had a cytokine secretion profile and surface phenotype similar to that reported for Tr1 cells. Notably, the frequency of disease appeared to increase at 14 months. The diseased mice had small spleens which averaged 47 mg, while the remaining non-diseased mice in our colony killed at ages 14-15 months had splenocytes that averaged 80 mg (ranging from 47-130 mg). Thus, the appearance of disease was associated with diminution in numbers of IL-10-secreting regulatory T cells with age.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Subtipos Serológicos HLA-DR/metabolismo , Interleucina-10/metabolismo , Esclerosis Múltiple/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Bazo/patología , Linfocitos T Reguladores/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Subtipos Serológicos HLA-DR/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Básica de Mielina/inmunología , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética
5.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33622787

RESUMEN

HLA-C arose during evolution of pregnancy in the great apes 10 to 15 million years ago. It has a dual function on placental extravillous trophoblasts (EVTs) as it contributes to both tolerance and immunity at the maternal-fetal interface. The mode of its regulation is of considerable interest in connection with the biology of pregnancy and pregnancy abnormalities. First-trimester primary EVTs in which HLA-C is highly expressed, as well as JEG3, an EVT model cell line, were employed. Single-cell RNA-seq data and quantitative PCR identified high expression of the transcription factor ELF3 in those cells. Chromatin immunoprecipitation (ChIP)-PCR confirmed that both ELF3 and MED1 bound to the proximal HLA-C promoter region. However, binding of RFX5 to this region was absent or severely reduced, and the adjacent HLA-B locus remained closed. Expression of HLA-C was inhibited by ELF3 small interfering RNAs (siRNAs) and by wrenchnolol treatment. Wrenchnolol is a cell-permeable synthetic organic molecule that mimics ELF3 and is relatively specific for binding to ELF3's coactivator, MED23, as our data also showed in JEG3. Moreover, the ELF3 gene is regulated by a superenhancer that spans more than 5 Mb, identified by assay for transposase-accessible chromatin using sequencing (ATAC-seq), as well as by its sensitivity to (+)-JQ1 (inhibitor of BRD4). ELF3 bound to its own promoter, thus creating an autoregulatory feedback loop that establishes expression of ELF3 and HLA-C in trophoblasts. Wrenchnolol blocked binding of MED23 to ELF3, thus disrupting the positive-feedback loop that drives ELF3 expression, with down-regulation of HLA-C expression as a consequence.


Asunto(s)
Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Retroalimentación Fisiológica , Antígenos HLA-C/genética , Proteínas Proto-Oncogénicas c-ets/genética , Factores de Transcripción/genética , Trofoblastos/inmunología , Aborto Legal , Adamantano/farmacología , Azepinas/farmacología , Línea Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/inmunología , Femenino , Regulación del Desarrollo de la Expresión Génica/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Antígenos HLA-C/inmunología , Humanos , Inmunidad Materno-Adquirida , Indoles/farmacología , Complejo Mediador/genética , Complejo Mediador/inmunología , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/inmunología , Embarazo , Primer Trimestre del Embarazo , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-ets/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ets/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/inmunología , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/inmunología , Triazoles/farmacología , Trofoblastos/citología , Trofoblastos/efectos de los fármacos
6.
Cell ; 182(5): 1125-1139.e18, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32822574

RESUMEN

Maternal decidual NK (dNK) cells promote placentation, but how they protect against placental infection while maintaining fetal tolerance is unclear. Here we show that human dNK cells highly express the antimicrobial peptide granulysin (GNLY) and selectively transfer it via nanotubes to extravillous trophoblasts to kill intracellular Listeria monocytogenes (Lm) without killing the trophoblast. Transfer of GNLY, but not other cell death-inducing cytotoxic granule proteins, strongly inhibits Lm in human placental cultures and in mouse and human trophoblast cell lines. Placental and fetal Lm loads are lower and pregnancy success is greatly improved in pregnant Lm-infected GNLY-transgenic mice than in wild-type mice that lack GNLY. This immune defense is not restricted to pregnancy; peripheral NK (pNK) cells also transfer GNLY to kill bacteria in macrophages and dendritic cells without killing the host cell. Nanotube transfer of GNLY allows dNK to protect against infection while leaving the maternal-fetal barrier intact.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Bacterias/inmunología , Movimiento Celular/inmunología , Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Células HeLa , Humanos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Placenta/inmunología , Placenta/microbiología , Embarazo , Ratas , Células THP-1 , Trofoblastos/microbiología
7.
Proc Natl Acad Sci U S A ; 117(27): 15772-15777, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32581122

RESUMEN

During pregnancy, invading HLA-G+ extravillous trophoblasts (EVT) play a key role in placental development, uterine spiral artery remodeling, and prevention of detrimental maternal immune responses to placental and fetal antigens. Failures of these processes are suggested to play a role in the development of pregnancy complications, but very little is known about the underlying mechanisms. Here we present validated methods to purify and culture primary HLA-G+ EVT from the placental disk and chorionic membrane from healthy term pregnancy. Characterization of HLA-G+ EVT from term pregnancy compared to first trimester revealed their unique phenotypes, gene expression profiles, and differing capacities to increase regulatory T cells (Treg) during coculture assays, features that cannot be captured by using surrogate cell lines or animal models. Furthermore, clinical variables including gestational age and fetal sex significantly influenced EVT biology and function. These methods and approaches form a solid basis for further investigation of the role of HLA-G+ EVT in the development of detrimental placental inflammatory responses associated with pregnancy complications, including spontaneous preterm delivery and preeclampsia.


Asunto(s)
Antígenos HLA-G/inmunología , Inmunidad Innata/genética , Placentación/inmunología , Preeclampsia/inmunología , Línea Celular , Movimiento Celular/inmunología , Femenino , Regulación del Desarrollo de la Expresión Génica/inmunología , Humanos , Relaciones Materno-Fetales , Placenta/inmunología , Placenta/metabolismo , Preeclampsia/patología , Embarazo , Primer Trimestre del Embarazo , Trofoblastos/inmunología
8.
J Immunol ; 204(12): 3149-3159, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32376646

RESUMEN

Decidual NK cells (dNK) are the main lymphocyte population in early pregnancy decidual mucosa. Although dNK decrease during pregnancy, they remain present in decidual tissues at term. First trimester dNK facilitate trophoblast invasion, provide protection against infections, and were shown to have many differences in their expression of NKRs, cytokines, and cytolytic capacity compared with peripheral blood NK cells (pNK). However, only limited data are available on the phenotype and function of term pregnancy dNK. In this study, dNK from human term pregnancy decidua basalis and decidua parietalis tissues were compared with pNK and first trimester dNK. Profound differences were found, including: 1) term pregnancy dNK have an increased degranulation response to K562 and PMA/ionomycin but lower capacity to respond to human CMV-infected cells; 2) term pregnancy dNK are not skewed toward recognition of HLA-C, as was previously shown for first trimester dNK; and 3) protein and gene expression profiles identified multiple differences between pNK, first trimester, and term pregnancy dNK, suggesting term pregnancy dNK are a distinct type of NK cells. Understanding the role of dNK throughout pregnancy is of high clinical relevance for studies aiming to prevent placental inflammatory disorders as well as maternal-to-fetal transmission of pathogens.


Asunto(s)
Decidua/inmunología , Células Asesinas Naturales/inmunología , Línea Celular Tumoral , Movimiento Celular/inmunología , Células Cultivadas , Femenino , Expresión Génica/inmunología , Antígenos HLA-C/inmunología , Humanos , Células K562 , Embarazo , Trofoblastos/inmunología
9.
Cell Rep ; 27(9): 2537-2547.e5, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31141680

RESUMEN

During pregnancy, maternal regulatory T cells (Tregs) are important in establishing immune tolerance to invading fetal extravillous trophoblasts (EVTs). CD25HIFOXP3+ Tregs are found at high levels in decidual tissues and have been shown to suppress fetus-specific and nonspecific responses. However, limited data are available on additional decidual Treg types and the mechanisms by which they are induced. This study investigated three distinct decidual CD4+ Treg types in healthy pregnancies with a regulatory phenotype and the ability to suppress T cell responses: CD25HIFOXP3+, PD1HIIL-10+, and TIGIT+FOXP3dim. Moreover, co-culture of HLA-G+ EVTs or decidual macrophages with blood CD4+ T cells directly increased the proportions of CD25HIFOXP3+ Tregs compared to T cells cultured alone. EVTs also increased PD1HI Tregs that could be inhibited by HLA-C and CD3 antibodies, suggesting an antigen-specific induction. The presence of distinct Treg types may allow for the modulation of a variety of inflammatory responses in the placenta.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Decidua/inmunología , Feto/inmunología , Antígenos HLA-C/inmunología , Tolerancia Inmunológica/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Decidua/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-10/inmunología , Interleucina-10/metabolismo , Placenta/inmunología , Placenta/metabolismo , Embarazo , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores/metabolismo , Trofoblastos/inmunología , Trofoblastos/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(21): 10441-10446, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31040209

RESUMEN

Polymorphic HLAs form the primary immune barrier to cell therapy. In addition, innate immune surveillance impacts cell engraftment, yet a strategy to control both, adaptive and innate immunity, is lacking. Here we employed multiplex genome editing to specifically ablate the expression of the highly polymorphic HLA-A/-B/-C and HLA class II in human pluripotent stem cells. Furthermore, to prevent innate immune rejection and further suppress adaptive immune responses, we expressed the immunomodulatory factors PD-L1, HLA-G, and the macrophage "don't-eat me" signal CD47 from the AAVS1 safe harbor locus. Utilizing in vitro and in vivo immunoassays, we found that T cell responses were blunted. Moreover, NK cell killing and macrophage engulfment of our engineered cells were minimal. Our results describe an approach that effectively targets adaptive as well as innate immune responses and may therefore enable cell therapy on a broader scale.


Asunto(s)
Ingeniería Genética/métodos , Células Madre Pluripotentes/inmunología , Sistemas CRISPR-Cas , Línea Celular , Técnicas de Inactivación de Genes , Genes MHC Clase I , Genes MHC Clase II , Humanos
11.
Proc Natl Acad Sci U S A ; 116(7): 2634-2639, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30683721

RESUMEN

Random amino acid copolymers used in the treatment of multiple sclerosis in man or experimental autoimmune encephalomyelitis (EAE) in mice [poly(Y,E,A,K)n, known as Copaxone, and poly(Y,F,A,K)n] function at least in part by generation of IL-10-secreting regulatory T cells that mediate bystander immunosuppression. The mechanism through which these copolymers induce Tregs is unknown. To investigate this question, four previously described Vα3.2 Vß14 T cell receptor (TCR) cDNAs, the dominant clonotype generated in splenocytes after immunization of SJL mice, that differed only in their CDR3 sequences were utilized to generate retrogenic mice. The high-level production of IL-10 as well as IL-5 and small amounts of the related cytokines IL-4 and IL-13 by CD4+ T cells isolated from the splenocytes of these mice strongly suggests that the TCR itself encodes information for specific cytokine secretion. The proliferation and production of IL-10 by these Tregs was costimulated by activation of glucocorticoid-induced TNF receptor (GITR) (expressed at high levels by these cells) through its ligand GITRL. A mechanism for generation of cells with this specificity is proposed. Moreover, retrogenic mice expressing these Tregs were protected from induction of EAE by the appropriate autoantigen.


Asunto(s)
Células Madre Hematopoyéticas/citología , Interleucina-10/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Animales , ADN Complementario , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Vectores Genéticos , Tolerancia Inmunológica , Interleucinas/metabolismo , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Factores de Necrosis Tumoral/metabolismo
12.
Front Immunol ; 10: 2730, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921098

RESUMEN

To establish a healthy pregnancy, maternal immune cells must tolerate fetal allo-antigens and remain competent to respond to infections both systemically and in placental tissues. Extravillous trophoblasts (EVT) are the most invasive cells of extra-embryonic origin to invade uterine tissues and express polymorphic Human Leucocyte Antigen-C (HLA-C) of both maternal and paternal origin. Thus, HLA-C is a key molecule that can elicit allogeneic immune responses by maternal T and NK cells and for which maternal-fetal immune tolerance needs to be established. HLA-C is also the only classical MHC molecule expressed by EVT that can present a wide variety of peptides to maternal memory T cells and establish protective immunity. The expression of paternal HLA-C by EVT provides a target for maternal NK and T cells, whereas HLA-C expression levels may influence how this response is shaped. This dual function of HLA-C requires tight transcriptional regulation of its expression to balance induction of tolerance and immunity. Here, we critically review new insights into: (i) the mechanisms controlling expression of HLA-C by EVT, (ii) the mechanisms by which decidual NK cells, effector T cells and regulatory T cells recognize HLA-C allo-antigens, and (iii) immune recognition of pathogen derived antigens in context of HLA-C.


Asunto(s)
Antígenos HLA-C/inmunología , Tolerancia Inmunológica , Inmunidad , Intercambio Materno-Fetal/inmunología , Placenta/inmunología , Placenta/metabolismo , Alelos , Decidua/inmunología , Decidua/metabolismo , Femenino , Regulación de la Expresión Génica , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Antígenos HLA-C/genética , Humanos , Tolerancia Inmunológica/genética , Inmunidad/genética , Inmunomodulación , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Embarazo , Regiones Promotoras Genéticas , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trofoblastos/metabolismo
14.
Proc Natl Acad Sci U S A ; 115(2): 385-390, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29259116

RESUMEN

Understanding how decidual CD8+ T cell (CD8+ dT) cytotoxicity is regulated and how these cells integrate the competing needs for maternal-fetal tolerance and immunity to infection is an important research and clinical goal. Gene-expression analysis of effector-memory CD8+ dT demonstrated a mixed transcriptional signature of T cell dysfunction, activation, and effector function. High protein expression of coinhibitory molecules PD1, CTLA4, and LAG3, accompanied by low expression of cytolytic molecules suggests that the decidual microenvironment reduces CD8+ dT effector responses to maintain tolerance to fetal antigens. However, CD8+ dT degranulated, proliferated, and produced IFN-γ, TNF-α, perforin, and granzymes upon in vitro stimulation, demonstrating that CD8+ dT are not permanently suppressed and retain the capacity to respond to proinflammatory events, such as infections. The balance between transient dysfunction of CD8+ dT that are permissive of placental and fetal development, and reversal of this dysfunctional state, is crucial in understanding the etiology of pregnancy complications and prevention of congenital infections.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Decidua/metabolismo , Perfilación de la Expresión Génica/métodos , Tolerancia Inmunológica/genética , Linfocitos T CD8-positivos/inmunología , Citomegalovirus/inmunología , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Femenino , Granzimas/genética , Granzimas/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Perforina/genética , Perforina/metabolismo , Factores de Tiempo
15.
Sci Rep ; 7(1): 3501, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28615725

RESUMEN

For many years, human peripheral blood natural killer (NK) cells have been divided into functionally distinct CD3- CD56bright CD16- and CD3- CD56dim CD16+ subsets. Recently, several groups of innate lymphoid cells (ILC), distinct from NK cells in development and function, have been defined in mouse. A signature of genes present in mouse ILC except NK cells, defined by Immunological Genome Project studies, is significantly over-represented in human CD56bright cells, by gene set enrichment analysis. Conversely, the signature genes of mouse NK cells are enriched in human CD56dim cells. Correlations are based upon large differences in expression of a few key genes. CD56bright cells show preferential expression of ILC-associated IL7R (CD127), TNFSF10 (TRAIL), KIT (CD117), IL2RA (CD25), CD27, CXCR3, DPP4 (CD26), GPR183, and MHC class II transcripts and proteins. This could indicate an ontological relationship between human CD56bright cells and mouse CD127+ ILC, or conserved networks of transcriptional regulation. In line with the latter hypothesis, among transcription factors known to impact ILC or NK cell development, GATA3, TCF7 (TCF-1), AHR, SOX4, RUNX2, and ZEB1 transcript levels are higher in CD56bright cells, while IKZF3 (AIOLOS), TBX21 (T-bet), NFIL3 (E4BP4), ZEB2, PRDM1 (BLIMP1), and RORA mRNA levels are higher in CD56dim cells.


Asunto(s)
Complejo CD3/genética , Antígeno CD56/genética , Subunidad alfa del Receptor de Interleucina-7/genética , Células Asesinas Naturales/metabolismo , Transcriptoma , Animales , Complejo CD3/sangre , Complejo CD3/inmunología , Antígeno CD56/sangre , Antígeno CD56/inmunología , Perfilación de la Expresión Génica , Humanos , Subunidad alfa del Receptor de Interleucina-7/sangre , Subunidad alfa del Receptor de Interleucina-7/inmunología , Células Asesinas Naturales/inmunología , Ratones , Especificidad de la Especie
16.
Biol Reprod ; 96(4): 831-842, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28340094

RESUMEN

During pregnancy, fetal extravillous trophoblasts (EVT) play a key role in the regulation of maternal T cell and NK cell responses. EVT display a unique combination of human leukocyte antigens (HLA); EVT do not express HLA-A and HLA-B, but do express HLA-C, HLA-E, and HLA-G. The mechanisms establishing this unique HLA expression pattern have not been fully elucidated. The major histocompatibility complex (MHC) class I and class II transcriptional activators NLRC5 and CIITA are expressed neither by EVT nor by the EVT model cell line JEG3, which has an MHC expression pattern identical to that of EVT. Therefore, other MHC regulators must be present to control HLA-C, HLA-E, and HLA-G expression in these cells. CIITA and NLRC5 are both members of the nucleotide-binding domain, leucine-rich repeat (NLR) family of proteins. Another member of this family, NLRP2, is highly expressed by EVT and JEG3, but not in maternal decidual stromal cells. In this study, transcription activator-like effector nuclease technology was used to delete NLRP2 in JEG3. Furthermore, lentiviral delivery of shRNA was used to knockdown NLRP2 in JEG3 and primary EVT. Upon NLRP2 deletion, Tumor Necrosis Factor-α (TNFα)-induced phosphorylation of NF-KB p65 increased in JEG3 and EVT, and more surprisingly a significant increase in constitutive HLA-C expression was observed in JEG3. These data suggest a broader role for NLR family members in the regulation of MHC expression during inflammation, thus forming a bridge between innate and adaptive immune responses. As suppressor of proinflammatory responses, NLRP2 may contribute to preventing unwanted antifetal responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Antígenos HLA-C/metabolismo , FN-kappa B/metabolismo , Trofoblastos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Eliminación de Gen , Genes MHC Clase I/genética , Antígenos HLA-C/genética , Humanos , FN-kappa B/genética , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
17.
Trends Immunol ; 38(4): 272-286, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28279591

RESUMEN

During pregnancy, semiallogeneic fetal extravillous trophoblasts (EVT) invade the uterine mucosa without being rejected by the maternal immune system. Several mechanisms were initially proposed by Peter Medawar half a century ago to explain this apparent violation of the laws of transplantation. Then, three decades ago, an unusual human leukocyte antigen (HLA) molecule was identified: HLA-G. Uniquely expressed in EVT, HLA-G has since become the center of the present understanding of fetus-induced immune tolerance. Despite slow progress in the field, the last few years have seen an explosion in our knowledge of HLA-G biology. Here, we critically review new insights into the mechanisms controlling the expression and function of HLA-G at the maternal-fetal interface, and discuss their relevance for fetal tolerance.


Asunto(s)
Antígenos HLA-G/metabolismo , Relaciones Materno-Fetales , Embarazo/inmunología , Tolerancia al Trasplante , Trofoblastos/inmunología , Animales , Femenino , Antígenos HLA-G/inmunología , Histocompatibilidad , Humanos , Isoantígenos/inmunología
18.
J Reprod Immunol ; 119: 85-90, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27523927

RESUMEN

To establish a healthy pregnancy the maternal immune system must tolerate fetal allo-antigens, yet remain competent to respond to infections. The ability of decidual NK cells (dNK) to promote migration of fetal extravillous trophoblasts (EVT) and placental growth as well as the capacity of EVT to promote immune tolerance are topics of high interest and extensive research. However, the problem of how dNK and decidual CD8+ T cells (CD8+ dT) provide immunity to infections of the placenta and the mechanisms that regulate their cytolytic function has thus far largely been ignored. Fetal EVT are the most invasive cells of the placenta and directly interact with maternal decidual immune cells at this maternal-fetal interface. Besides the expression of non-polymorphic HLA-E and HLA-G molecules that are associated with immune tolerance, EVT also express highly polymorphic HLA-C molecules that can serve as targets for maternal dNK and CD8+ dT responses. HLA-C expression by EVT has a dual role as the main molecule to which immune tolerance needs to be established and as the only molecule that can present pathogen-derived peptides and provide protective immunity when EVT are infected. The focus of this review is to address the regulation of cytotoxicity of dNK and CD8+ dT, which is essential for maternal-fetal immune tolerance as well as recent evidence that both cell types can provide immunity to infections at the maternal-fetal interface. A particular emphasis is given to the role of HLA-C expressed by EVT and its capacity to elicit dNK and CD8+ dT responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Decidua/inmunología , Infecciones/inmunología , Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Inmunidad Adaptativa , Citotoxicidad Inmunológica , Femenino , Antígenos HLA-C/metabolismo , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Inmunidad Materno-Adquirida , Circulación Placentaria , Embarazo
19.
Proc Natl Acad Sci U S A ; 113(52): 15072-15077, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27956621

RESUMEN

The combination of the activating killer cell Ig-like receptor 2DS1 (KIR2DS1) expressed by maternal decidual natural killer cells (dNK) and the presence of its ligand, the HLA-C allotype HLA-C2, expressed by fetal trophoblasts, reduces the risk of developing pregnancy complications. However, no molecular or cellular mechanism explains this genetic correlation. Here we demonstrate that KIR2DS1+ dNK acquired higher cytotoxic function than KIR2DS1- dNK when exposed to human cytomegalovirus (HCMV)-infected decidual stromal cells (DSC), particularly when DSCs express HLA-C2. Furthermore, dNK were unable to degranulate or secrete cytokines in response to HCMV-infected primary fetal extravillous trophoblasts. This emphasizes the immunological challenge to clear placental viral infections within the immune-privileged placenta. Activation of dNK through KIR2DS1/HLA-C2 interaction increases their ability to respond to placental HCMV infection and may limit subsequent virus-induced placental pathology. This mechanism is directly related to how KIR2DS1 expressed by dNK reduces development of severe pregnancy complications such as miscarriages and preterm delivery.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Decidua/inmunología , Células Asesinas Naturales/inmunología , Placenta/inmunología , Receptores KIR/metabolismo , Separación Celular , Citomegalovirus , Decidua/virología , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Antígenos HLA-C/análisis , Humanos , Tolerancia Inmunológica , Inmunoglobulinas/metabolismo , Placenta/virología , Placentación , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , Resultado del Embarazo , Receptores KIR/genética , Trofoblastos/virología
20.
Nat Commun ; 7: 13283, 2016 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-27796305

RESUMEN

The study of amyotrophic lateral sclerosis (ALS) and potential interventions would be facilitated if motor axon degeneration could be more readily visualized. Here we demonstrate that stimulated Raman scattering (SRS) microscopy could be used to sensitively monitor peripheral nerve degeneration in ALS mouse models and ALS autopsy materials. Three-dimensional imaging of pre-symptomatic SOD1 mouse models and data processing by a correlation-based algorithm revealed that significant degeneration of peripheral nerves could be detected coincidentally with the earliest detectable signs of muscle denervation and preceded physiologically measurable motor function decline. We also found that peripheral degeneration was an early event in FUS as well as C9ORF72 repeat expansion models of ALS, and that serial imaging allowed long-term observation of disease progression and drug effects in living animals. Our study demonstrates that SRS imaging is a sensitive and quantitative means of measuring disease progression, greatly facilitating future studies of disease mechanisms and candidate therapeutics.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Degeneración Nerviosa/patología , Nervios Periféricos/patología , Espectrometría Raman , Algoritmos , Animales , Antibacterianos , Artefactos , Simulación por Computador , Progresión de la Enfermedad , Electromiografía , Femenino , Humanos , Imagenología Tridimensional , Lípidos/química , Masculino , Ratones , Ratones Transgénicos , Minociclina/química , Neuronas Motoras/patología , Vaina de Mielina/química , Nervio Ciático/patología , Superóxido Dismutasa-1/genética , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA