Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.248
Filtrar
2.
Front Immunol ; 15: 1368572, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698852

RESUMEN

Introduction: Interferon-gamma (IFN-γ) is pivotal in orchestrating immune responses during healthy pregnancy. However, its dysregulation, often due to autoimmunity, infections, or chronic inflammatory conditions, is implicated in adverse reproductive outcomes such as pregnancy failure or infertility. Additionally, the underlying immunological mechanisms remain elusive. Methods: Here, we explore the impact of systemic IFN-γ elevation on cytotoxic T cell responses in female reproduction utilizing a systemic lupus-prone mouse model with impaired IFN-γ degradation. Results: Our findings reveal that heightened IFN-γ levels triggered the infiltration of CD8+T cells in the pituitary gland and female reproductive tract (FRT), resulting in prolactin deficiency and subsequent infertility. Furthermore, we demonstrate that chronic IFN-γ elevation increases effector memory CD8+T cells in the murine ovary and uterus. Discussion: These insights broaden our understanding of the role of elevated IFN-γ in female reproductive dysfunction and suggest CD8+T cells as potential immunotherapeutic targets in female reproductive disorders associated with chronic systemic IFN-γ elevation.


Asunto(s)
Linfocitos T CD8-positivos , Interferón gamma , Animales , Femenino , Interferón gamma/metabolismo , Ratones , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/inmunología , Útero/inmunología , Infertilidad Femenina/inmunología , Hipófisis/inmunología , Hipófisis/metabolismo , Ratones Endogámicos C57BL , Embarazo , Prolactina/metabolismo , Ovario/inmunología
3.
J Immunol Res ; 2024: 5582151, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690552

RESUMEN

Unlike T cells in other tissues, uterine T cells must balance strong immune defense against pathogens with tolerance to semiallogeneic fetus. Our previous study fully elucidated the characteristics of γδT cells in nonpregnant uterus and the mechanism modulated by estrogen. However, comprehensive knowledge of the immunological properties of αßT (including CD4+T cells and CD8+T) cells in nonpregnancy uterus has not been acquired. In this study, we fully compared the immunological properties of αßT cells between uterus and blood using mouse and human sample. It showed that most of CD4+T cells and CD8+T cells in murine uterus and human endometrium were tissue resident memory T cells which highly expressed tissue residence markers CD69 and/or CD103. In addition, both CD4+T cells and CD8+T cells in uterus highly expressed inhibitory molecular PD-1 and cytokine IFN-γ. Uterine CD4+T cells highly expressed IL-17 and modulated by transcription factor pSTAT3. Moreover, we compared the similarities and differences between human and murine uterine T cell phenotype. Together, uterine CD4+T cells and CD8+ cells exhibited a unique mixed signature of T cell dysfunction, activation, and effector function which enabled them to balance strong immune defense against pathogens with tolerance to fetus. Our study fully elucidated the unique immunologic properties of uterine CD4+T and CD8+T cells and provided a base for further investigation of functions.


Asunto(s)
Antígenos CD , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Útero , Femenino , Linfocitos T CD8-positivos/inmunología , Animales , Humanos , Ratones , Linfocitos T CD4-Positivos/inmunología , Útero/inmunología , Antígenos CD/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/genética , Cadenas alfa de Integrinas/metabolismo , Células T de Memoria/inmunología , Factor de Transcripción STAT3/metabolismo , Interferón gamma/metabolismo , Lectinas Tipo C/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Interleucina-17/metabolismo , Activación de Linfocitos/inmunología , Memoria Inmunológica
4.
Am J Reprod Immunol ; 91(4): e13842, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38650366

RESUMEN

PROBLEM: Although endometrial receptivity is a key factor in influencing implantation in both naturally conceived and assisted reproductive technology (ART) cycles, very little is known about the endometrium milieu around the time of implantation. Previous studies have demonstrated the presence of several cytokines in the endometrium that affect implantation. However, there is lacking data about the presence of immune cell subtypes within the endometrium and in the uterine cavity at the time of implantation. METHOD OF STUDY: This study was approved by the Institutional Review Board (# 225589). The study was designed as a prospective observational cohort study between May 2021 and December 2022 at a single academic-based fertility center. All patients underwent at least one In Vitro Fertilization (IVF) cycle and have frozen embryos. Twenty-four participants were recruited for this study which was conducted during the frozen embryo transfer (FET) cycle regardless of the outcome of previous cycles. Two samples were acquired from each subject, denoted as lower and upper. A trial transfer catheter was introduced under ultrasound guidance into the lower uterine segment. Upon removal, the tip was rinsed in IMDM medium containing 10% FBS (lower uterus). A transfer catheter was then loaded with the embryo that was placed in the upper uterus under ultrasound guidance. The tip of the transfer catheter was rinsed in separate aliquot of the above media (upper uterus). After centrifugation, pelleted cells were stained for the following surface markers: CD45, CD3, CD19, CD4, CD8, gamma delta TCR, CD25, CD127, CD66b, CD14, CD16, CD56 and acquired on Sony SP6800 Spectral Analyzer. RESULTS: Upon staining the pelleted cells, we were able to identify viable leukocytes from samples obtained from both, upper and lower uterus (0.125 × 106 cells ± SD 0.32), (0.123 × 106 cells ± SD 0.12), respectively. Among total viable cells, there was no significant difference in both percent and number of CD45+ cells between the upper and lower uterus (9.88% ± 6.98 SD, 13.67% ± 9.79 SD, p = .198) respectively. However, there was significantly higher expression of CD3+ (p = .006), CD19+ (p = .032) and CD14+ (p = .019) cells in samples collected from upper compared to lower uterus. Within all CD3+ cells, we found that gamma delta T cells (GDT) were the major population of T cells in both upper and lower uterus. In contrast, CD8+ T cells were significantly higher in the lower uterus when compared to the upper uterus (p = .009). There was no statistically significant difference in the expression of CD4+ T cells, T regulatory cells (CD4+CD25+CD127-), NK cells (CD56+), neutrophils (CD66b+) and FcγRIII+ cells (CD16+) between upper and lower uterus. CONCLUSIONS: We believe the immune milieu at the time of embryo transfer will affect implantation. Understanding the composition of immune cells will guide further research in identifying optimal immune milieus that favor implantation. Comprehensive analysis of endometrium is expected to lead to new diagnostic and therapeutic approaches to improve IVF outcomes.


Asunto(s)
Transferencia de Embrión , Endometrio , Útero , Humanos , Femenino , Adulto , Transferencia de Embrión/métodos , Útero/inmunología , Endometrio/inmunología , Endometrio/citología , Estudios Prospectivos , Implantación del Embrión/inmunología , Fertilización In Vitro , Embarazo , Líquidos Corporales/inmunología
5.
Ginekol Pol ; 95(4): 266-275, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38334340

RESUMEN

Recurrent miscarriage, poses a significant challenge for many couples globally, the causes of which are not fully understood. Recent studies have shown the intricate link between uterine inflammation and recurrent miscarriages. While inflammation is essential during early pregnancy stages, especially in embryo implantation, an imbalance can lead to miscarriage. Key inflammatory mediators and an imbalance in immune cells can significantly alter and contribute to recurrent miscarriages. Lifestyle factors like smoking and obesity exacerbate inflammatory responses, increasing miscarriage risks. Understanding the interaction between the uterine environment, immune cell imbalances, and recurrent miscarriages is essential for devising effective treatments. This paper presents the latest data on inflammation's role in recurrent miscarriage, emphasizing the significance of diagnosing chronic endometritis and immune imbalances, offering practical recommendations for treatment and diagnosis.


Asunto(s)
Aborto Habitual , Humanos , Femenino , Aborto Habitual/inmunología , Aborto Habitual/terapia , Aborto Habitual/prevención & control , Embarazo , Inflamación/inmunología , Útero/inmunología , Endometritis/inmunología , Endometritis/terapia
6.
Nature ; 619(7970): 595-605, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37468587

RESUMEN

Beginning in the first trimester, fetally derived extravillous trophoblasts (EVTs) invade the uterus and remodel its spiral arteries, transforming them into large, dilated blood vessels. Several mechanisms have been proposed to explain how EVTs coordinate with the maternal decidua to promote a tissue microenvironment conducive to spiral artery remodelling (SAR)1-3. However, it remains a matter of debate regarding which immune and stromal cells participate in these interactions and how this evolves with respect to gestational age. Here we used a multiomics approach, combining the strengths of spatial proteomics and transcriptomics, to construct a spatiotemporal atlas of the human maternal-fetal interface in the first half of pregnancy. We used multiplexed ion beam imaging by time-of-flight and a 37-plex antibody panel to analyse around 500,000 cells and 588 arteries within intact decidua from 66 individuals between 6 and 20 weeks of gestation, integrating this dataset with co-registered transcriptomics profiles. Gestational age substantially influenced the frequency of maternal immune and stromal cells, with tolerogenic subsets expressing CD206, CD163, TIM-3, galectin-9 and IDO-1 becoming increasingly enriched and colocalized at later time points. By contrast, SAR progression preferentially correlated with EVT invasion and was transcriptionally defined by 78 gene ontology pathways exhibiting distinct monotonic and biphasic trends. Last, we developed an integrated model of SAR whereby invasion is accompanied by the upregulation of pro-angiogenic, immunoregulatory EVT programmes that promote interactions with the vascular endothelium while avoiding the activation of maternal immune cells.


Asunto(s)
Intercambio Materno-Fetal , Trofoblastos , Útero , Femenino , Humanos , Embarazo , Arterias/fisiología , Decidua/irrigación sanguínea , Decidua/citología , Decidua/inmunología , Decidua/fisiología , Primer Trimestre del Embarazo/genética , Primer Trimestre del Embarazo/metabolismo , Primer Trimestre del Embarazo/fisiología , Trofoblastos/citología , Trofoblastos/inmunología , Trofoblastos/fisiología , Útero/irrigación sanguínea , Útero/citología , Útero/inmunología , Útero/fisiología , Intercambio Materno-Fetal/genética , Intercambio Materno-Fetal/inmunología , Intercambio Materno-Fetal/fisiología , Factores de Tiempo , Proteómica , Perfilación de la Expresión Génica , Conjuntos de Datos como Asunto , Edad Gestacional
7.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994660

RESUMEN

The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.


Asunto(s)
Interleucina-33 , Placenta , Placentación , Útero , Animales , Decidua/irrigación sanguínea , Decidua/citología , Decidua/crecimiento & desarrollo , Decidua/inmunología , Femenino , Feto/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/deficiencia , Interleucina-33/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Placenta/inmunología , Placenta/metabolismo , Embarazo , Útero/irrigación sanguínea , Útero/crecimiento & desarrollo , Útero/inmunología , Útero/metabolismo
8.
Int J Mol Sci ; 23(14)2022 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-35886985

RESUMEN

Recently, it was demonstrated that the expression of BMAL1 was decreased in the endometrium of women suffering from recurrent spontaneous abortion. To investigate the pathological roles of uterine clock genes during pregnancy, we produced conditional deletion of uterine Bmal1 (cKO) mice and found that cKO mice could receive embryo implantation but not sustain pregnancy. Gene ontology analysis of microarray suggested that uterine NK (uNK) cell function was suppressed in cKO mice. Histological examination revealed the poor formation of maternal vascular spaces in the placenta. In contrast to WT mice, uNK cells in the spongiotrophoblast layer, where maternal uNK cells are directly in contact with fetal trophoblast, hardly expressed an immunosuppressive NK marker, CD161, in cKO mice. By progesterone supplementation, pregnancy could be sustained until the end of pregnancy in some cKO mice. Although this treatment did not improve the structural abnormalities of the placenta, it recruited CD161-positive NK cells into the spongiotrophoblast layer in cKO mice. These findings indicate that the uterine clock system may be critical for pregnancy maintenance after embryo implantation.


Asunto(s)
Factores de Transcripción ARNTL , Muerte Fetal , Neovascularización Patológica , Placenta , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/inmunología , Animales , Implantación del Embrión/genética , Femenino , Muerte Fetal/etiología , Células Asesinas Naturales/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Placenta/irrigación sanguínea , Placenta/inmunología , Embarazo/genética , Embarazo/inmunología , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/inmunología , Mortinato/genética , Útero/inmunología
9.
Bull Exp Biol Med ; 172(6): 765-769, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35501657

RESUMEN

We studied the expression of pluripotency factor Oct-4 and the intensity of apoptosis in the uterus during spontaneous and immune abortions in mice. Increased expression of factor Bax and reduced protein Bcl-2 synthesis in cells of the decidual membrane and decreased Oct-4 expression in the myometrium and perimetrium were detected. Thus, both spontaneous and immune-dependent abortions impair the apoptosis processes in the decidua and the formation of a pool of Oct-4+ cells in the uterus. In immune-dependent abortions, the intensity of apoptosis of decidual cells was lower than in spontaneous abortion. Low expression of the transcription factor Oct-4 in the myometrium and perimetrium characterizes pregnancy failure irrespective of its mechanisms.


Asunto(s)
Aborto Espontáneo , Factor 3 de Transcripción de Unión a Octámeros , Útero , Aborto Espontáneo/inmunología , Animales , Apoptosis , Decidua/metabolismo , Femenino , Humanos , Ratones , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/inmunología , Embarazo , Útero/inmunología
10.
Front Immunol ; 12: 765730, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868016

RESUMEN

CD8+ T cells are the most frequent T cell population in the immune cell compartment at the feto-maternal interface. Due to their cytotoxic potential, the presence of CD8+ T cells in the immune privileged pregnant uterus has raised considerable interest. Here, we review our current understanding of CD8+ T cell biology in the uterus of pregnant women and discuss this knowledge in relation to a recently published immune cell Atlas of human decidua. We describe how the expansion of CD8+ T cells with an effector memory phenotype often presenting markers of exhaustion is critical for a successful pregnancy, and host defense towards pathogens. Moreover, we review new evidence on the presence of long-lasting immunological memory to former pregnancies and discuss its impact on prospective pregnancy outcomes. The formation of fetal-specific memory CD8+ T cell subests in the uterus, in particular of tissue resident, and stem cell memory cells requires further investigation, but promises interesting results to come. Advancing the knowledge of CD8+ T cell biology in the pregnant uterus will be pivotal for understanding not only tissue-specific immune tolerance but also the etiology of complications during pregnancy, thus enabling preventive or therapeutic interventions in the future.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Embarazo/inmunología , Útero/inmunología , Decidua/inmunología , Epítopos , Femenino , Humanos , Tolerancia Inmunológica , Memoria Inmunológica/inmunología
11.
Front Immunol ; 12: 785717, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868069

RESUMEN

Intrauterine adhesion (IUA) is an endometrial fibrosis disease caused by repeated operations of the uterus and is a common cause of female infertility. In recent years, treatment using mesenchymal stem cells (MSCs) has been proposed by many researchers and is now widely used in clinics because of the low immunogenicity of MSCs. It is believed that allogeneic MSCs can be used to treat IUA because MSCs express only low levels of MHC class I molecules and no MHC class II or co-stimulatory molecules. However, many scholars still believe that the use of allogeneic MSCs to treat IUA may lead to immune rejection. Compared with allogeneic MSCs, autologous MSCs are safer, more ethical, and can better adapt to the body. Here, we review recently published articles on the immunomodulation of allogeneic and autologous MSCs in IUA therapy, with the aim of proving that the use of autologous MSCs can reduce the possibility of immune rejection in the treatment of IUAs.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Enfermedades Uterinas/terapia , Femenino , Enfermedad Injerto contra Huésped/inmunología , Humanos , Tolerancia Inmunológica , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Adherencias Tisulares/inmunología , Adherencias Tisulares/patología , Adherencias Tisulares/terapia , Trasplante Autólogo/métodos , Trasplante Homólogo/métodos , Resultado del Tratamiento , Enfermedades Uterinas/inmunología , Enfermedades Uterinas/patología , Útero/inmunología , Útero/patología
12.
Medwave ; 21(10): e8484, 2021 Nov 15.
Artículo en Español, Inglés | MEDLINE | ID: mdl-34780395

RESUMEN

Proper communication between natural killer cells and the human leukocyte antigens of the embryonic trophoblast at the maternal-fetal interface during pregnancy is essential for successful reproduction. However, specific combinations of embryonic human leukocyte antigen-C with killer immunoglobulin-like receptors on decidual natural killer cells (the immunological code of pregnancy) can be associated with obstetric morbidity and pregnancy loss. This article presents an updated review of the mechanisms underlying the interaction between embryonic human leukocyte antigen-C and maternal killer immunoglobulin-like receptors and their relevance to the physiology and pathophysiology of human reproduction.


Una adecuada comunicación entre las células asesinas naturales en la interfase materno-fetal con las moléculas de los antígenos de histocompatibilidad del trofoblasto embrionario es clave en el éxito de la reproducción. Sin embargo, combinaciones de determinados antígenos leucocitarios humanos tipo C embrionarios con los receptores tipo inmunoglobulina presentes en las células asesinas naturales deciduales (el código inmunológico del embarazo), pueden asociarse con morbilidad obstétrica y pérdidas gestacionales. En este artículo se presenta una revisión actualizada de los mecanismos subyacentes a la interacción entre el antígeno de histocompatibilidad tipo C embrionario y los receptores tipo inmunoglobulina maternos, y su relevancia tanto en la fisiología como en la fisiopatología de la reproducción humana.


Asunto(s)
Aborto Habitual/inmunología , Antígenos HLA-C/inmunología , Células Asesinas Naturales/inmunología , Placentación/fisiología , Receptores KIR/inmunología , Medicina Reproductiva , Útero/inmunología , Aborto Espontáneo/inmunología , Implantación del Embrión/inmunología , Femenino , Antígenos HLA , Antígenos HLA-C/fisiología , Humanos , Células Asesinas Naturales/fisiología , Embarazo , Receptores KIR/fisiología
13.
Front Immunol ; 12: 723409, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712227

RESUMEN

Seminal plasma (SP), particularly SP exosomes (sExos), alters with age and can affect female mouse uterine immune microenvironment. However, the relationship between fertility decline in reproductively older males, and SP and sExos age-related changes, which may compromise the uterine immune microenvironment, remains unclear. The present study demonstrated that the implantation rate of female mice treated with SP from reproductively older male mice (aged-SP group) was lower than that of those treated with SP from younger male mice (young-SP group). RNA-sequencing analysis revealed altered levels of dendritic cell (DC)-related cytokines and chemokines in the uteri of the former group compared with those of the latter group. In vivo and in vitro experiments demonstrated a weaker inhibitory effect of aged SP on DC maturation than of young SP upon stimulation. After isolating and characterizing sExos from young and advanced-age male mice, we discovered that insemination of a subset of the aged-SP group with sExos from young male mice partially recovered the implantation rate decline. Additional in vivo and in vitro experiments revealed that sExos extracted from age male mice exerted a similar effect on DC maturation as SP of aged mice, indicating an age-related sExos inhibitory effect. In conclusion, our study demonstrated that age-related alterations of sExos may be partially responsible for lower implantation rates in the aged-SP group compared with those in the young-SP group, which were mediated by uterine immunomodulation. These findings provide new insights for clinical seminal adjuvant therapy.


Asunto(s)
Implantación del Embrión/inmunología , Exosomas/fisiología , Inmunomodulación/inmunología , Semen/inmunología , Útero/inmunología , Envejecimiento , Animales , Citocinas/inmunología , Endometrio/citología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Semen/citología , Interacciones Espermatozoide-Óvulo
14.
Front Immunol ; 12: 763067, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712245

RESUMEN

Uterine M1/M2 macrophages activation states undergo dynamic changes throughout pregnancy, and inappropriate macrophages polarization can cause adverse pregnancy outcomes, especially during the peri-implantation period. Our previous studies have confirmed that Cytochrome P450 26A1 (CYP26A1) can affect embryo implantation by regulating uterine NK cells and DCs. The aim of this study was to investigate whether CYP26A1 regulates the polarization of uterine macrophages in early pregnancy. Here, we observed that Cyp26a1 was significantly upregulated in M1 as compared with M2 of uterine macrophages, Raw264.7 and iBMDM. Knockdown of CYP26A1 in mice uterine significantly decreased the number of embryo implantation sites and the proportion of CD45+F4/80+CD206 - M1-like uterine macrophages. Primary uterine macrophages treated with anti-CYP26A1 antibody expressed significantly lower levels of M1 markers Nos2, Il1b, Il6 and Tnf-a. In CYP26A1 knockout Raw264.7 cells, the protein levels of M1 markers TNF-α, IL-6 and CD86 were significantly decreased as compared with the wild type cells. Moreover, CYP26A1 deficiency decreased the ability to produce nitric oxide and increased the phagocytosis capacity of Raw264.7 cells under M1 stimulation state. The re-introduction of CYP26A1 partially reversed the polarization levels of M1 in CYP26A1 knockout Raw264.7 cells. CYP26A1 may regulate the polarization of uterine macrophages to M1 through Stap1 and Slc7a2. In summary, these results indicate that CYP26A1 plays a significant role in macrophage polarization, and knockdown of CYP26A1 can cause insufficient M1 polarization during the peri-implantation period, which has adverse effects on blastocyst implantation.


Asunto(s)
Implantación del Embrión , Macrófagos/fisiología , Ácido Retinoico 4-Hidroxilasa/fisiología , Útero/inmunología , Animales , Polaridad Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Macrófagos/enzimología , Ratones , Ratones Endogámicos BALB C
15.
Front Immunol ; 12: 709229, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691022

RESUMEN

Preterm labor (PTL) is a multifactorial syndrome that results in birth prior to 37 weeks of gestation. However, the specific molecular mechanisms underlying this condition have yet to be elucidated. Previous research demonstrated that the abnormal expression of IL-27, and its receptors, played a role in the pathophysiology of preterm labor. In the present study, we established a Lipopolysaccharide (LPS)-stimulated, infection-induced, preterm mouse model based on wild-type C57BL/6 mice and WSX-1-/-C57BL/6 mice. WSX-1 knockdown led to a significant delay in birth by 11.32 ± 2.157h. In addition, compared with wild-type C57B/6 mice, the expression levels of IFN-γ, IL-1ß, IL-6, TNF-α, and CXCL10, in the fetal membrane and myometrium of WSX-1-/-mice were significantly lower, particularly in the myometrium. We also confirmed similar pro-inflammatory effects arising from IL-27 in human amniotic cell line (WISH) and human myometrial smooth muscle cell line (HMSMC). Once stimulated by LPS, the pro-inflammatory action exhibited a synergistic effect and appeared to be time-dependent. Finally, we demonstrated that LY3214996, an inhibitor of the ERK pathway, significantly inhibited the pro-inflammatory effect mediated by IL-27. Overall, our data confirmed that the inflammatory effect mediated by the IL-27/IFN-r/ERK axis is involved in preterm labor. Our findings, therefore, provide an enhancement in our etiological understanding of the mechanisms underlying PTL.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/fisiología , Inflamación/etiología , Interleucina-27/fisiología , Trabajo de Parto Prematuro/inmunología , Animales , Células Cultivadas , Femenino , Humanos , Interferón gamma/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptores de Interleucina/fisiología , Útero/inmunología
16.
Front Immunol ; 12: 741518, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34675929

RESUMEN

Intrauterine inflammation impacts prenatal neurodevelopment and is linked to adverse neurobehavioral outcomes ranging from cerebral palsy to autism spectrum disorder. However, the mechanism by which a prenatal exposure to intrauterine inflammation contributes to life-long neurobehavioral consequences is unknown. To address this gap in knowledge, this study investigates how inflammation transverses across multiple anatomic compartments from the maternal reproductive tract to the fetal brain and what specific cell types in the fetal brain may cause long-term neuronal injury. Utilizing a well-established mouse model, we found that mid-gestation intrauterine inflammation resulted in a lasting neutrophil influx to the decidua in the absence of maternal systemic inflammation. Fetal immunologic changes were observed at 72-hours post-intrauterine inflammation, including elevated neutrophils and macrophages in the fetal liver, and increased granulocytes and activated microglia in the fetal brain. Through unbiased clustering, a population of Gr-1+ γ/δ T cells was identified as the earliest immune cell shift in the fetal brain of fetuses exposed to intrauterine inflammation and determined to be producing high levels of IFNγ when compared to γ/δ T cells in other compartments. In a case-control study of term infants, IFNγ was found to be elevated in the cord blood of term infants exposed to intrauterine inflammation compared to those without this exposure. Collectively, these data identify a novel cellular immune mechanism for fetal brain injury in the setting of intrauterine inflammation.


Asunto(s)
Lesiones Encefálicas/inmunología , Encéfalo/inmunología , Decidua/inmunología , Inflamación/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Linfocitos T/inmunología , Útero/inmunología , Animales , Trastorno del Espectro Autista/inmunología , Células Cultivadas , Parálisis Cerebral/inmunología , Modelos Animales de Enfermedad , Femenino , Feto , Humanos , Lactante , Interferón gamma/metabolismo , Ratones , Embarazo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo
17.
PLoS Comput Biol ; 17(9): e1009365, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34492008

RESUMEN

Chlamydia trachomatis is a common sexually transmitted infection that is associated with a range of serious reproductive tract sequelae including in women Pelvic Inflammatory Disease (PID), tubal factor infertility, and ectopic pregnancy. Ascension of the pathogen beyond the cervix and into the upper reproductive tract is thought to be necessary for these pathologies. However, Chlamydia trachomatis does not encode a mechanism for movement on its genome, and so the processes that facilitate ascension have not been elucidated. Here, we evaluate the factors that may influence chlamydial ascension in women. We constructed a mathematical model based on a set of stochastic dynamics to elucidate the moderating factors that might influence ascension of infections in the first month of an infection. In the simulations conducted from the stochastic model, 36% of infections ascended, but only 9% had more than 1000 bacteria ascend. The results of the simulations indicated that infectious load and the peristaltic contractions moderate ascension and are inter-related in impact. Smaller initial loads were much more likely to ascend. Ascension was found to be dependent on the neutrophil response. Overall, our results indicate that infectious load, menstrual cycle timing, and the neutrophil response are critical factors in chlamydial ascension in women.


Asunto(s)
Infecciones por Chlamydia/microbiología , Chlamydia trachomatis , Modelos Biológicos , Útero/microbiología , Carga Bacteriana , Cuello del Útero/microbiología , Infecciones por Chlamydia/complicaciones , Infecciones por Chlamydia/fisiopatología , Chlamydia trachomatis/patogenicidad , Biología Computacional , Simulación por Computador , Femenino , Humanos , Infertilidad Femenina/etiología , Ciclo Menstrual/fisiología , Neutrófilos/inmunología , Enfermedad Inflamatoria Pélvica/etiología , Peristaltismo/fisiología , Embarazo , Embarazo Ectópico/etiología , Procesos Estocásticos , Útero/inmunología , Útero/fisiopatología
18.
Front Immunol ; 12: 711231, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34531861

RESUMEN

Women with endometriosis may have a defective immune system. However, evidence of the immune responses of endometriosis patients with a history of endometriosis surgery is lacking, and the association between the location of endometriosis lesions and immune responses is unclear. This retrospective study included 117 females with reproductive failure and a history of endometriosis and 200 females with reproductive failure but without endometriosis to analyze their endometrial and peripheral immune responses. The results show that endometriosis was associated with decreased peripheral natural killer (NK) cytotoxicity and increased uterine macrophages. Peripheral NK cytotoxicity at effector-to-target ratios of 25:1 and 50:1 was significantly reduced in women with a history of endometriosis from that of the control group (26.6% versus 33.3% and 36.1% versus 43.3%, respectively, both P < 0.001). Furthermore, after further division of patients into three subgroups according to the location of endometriosis lesions, we observed that NK cytotoxicity in the endometriosis subgroups, especially the mixed endometriosis group, was strongly decreased from that of the controls (P = 0.001). The endometrial CD68+ macrophage proportion in the mixed endometriosis subgroup was higher than that in the control group (2.8% versus 2.1%, P = 0.043). In addition, the baseline estradiol (E2) level was weakly correlated with the percentage of endometrial macrophages (r = 0.251, P = 0.009), indicating a potential association among the endocrine system, endometrial immune environment, and endometriosis. This study indicated that peripheral NK cytotoxicity and endometrial immune cell profiles could be useful for diagnosing and treating endometriosis and endometriosis-related reproductive diseases.


Asunto(s)
Citotoxicidad Inmunológica , Endometriosis/inmunología , Células Asesinas Naturales/inmunología , Macrófagos/fisiología , Útero/inmunología , Adulto , Antígenos CD/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Femenino , Humanos , Reproducción , Estudios Retrospectivos
19.
Viruses ; 13(9)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34578408

RESUMEN

The host's immune status may affect virus evolution. Little is known about how developing fetal and placental immune milieus affect virus heterogeneity. This knowledge will help us better understand intra-host virus evolution and how new virus variants emerge. The goal of our study was to find out whether the isolated in utero environment-an environment with specialized placental immunity and developing fetal immunity-supports the emergence of RNA and DNA virus variants. We used well-established porcine models for isolated Zika virus (RNA virus) and porcine circovirus 2 (DNA virus) fetal infections. We found that the isolated in utero environment was conducive to the emergence of RNA and DNA virus variants. Next-generation sequencing of nearly whole virus genomes and validated bioinformatics pipelines identified both unique and convergent single nucleotide variations in virus genomes isolated from different fetuses. Zika virus and PCV2 in utero evolution also resulted in single nucleotide variations previously reported in the human and porcine field samples. These findings should encourage further studies on virus evolution in placenta and fetuses, to better understand how virus variants emerge and how in utero viral evolution affects congenital virus transmission and pathogenicity.


Asunto(s)
Circovirus/genética , Evolución Molecular Dirigida , Variación Genética , Placenta/virología , Útero/virología , Virus Zika/genética , Animales , Microambiente Celular , Chlorocebus aethiops , Circovirus/fisiología , Femenino , Feto/virología , Heterogeneidad Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Placenta/inmunología , Embarazo , Porcinos , Útero/inmunología , Células Vero , Carga Viral , Virus Zika/fisiología
20.
FASEB J ; 35(10): e21938, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34547143

RESUMEN

Uterine glands are essential for the establishment of pregnancy and have critical roles in endometrial receptivity to blastocyst implantation, stromal cell decidualization, and placentation. Uterine gland dysfunction is considered a major contributing factor to pregnancy loss, however our understanding of how glands impact embryo survival and stromal cell decidualization is incomplete. Forkhead box A2 (FOXA2) is expressed only in the glandular epithelium and regulates its development and function. Mice with a conditional deletion of FOXA2 in the uterus are infertile due to defective embryo implantation arising from a lack of leukemia inhibitory factor (LIF), a critical factor of uterine gland origin. Here, a glandless FOXA2-deficient mouse model, coupled with LIF repletion to rescue the implantation defect, was used to investigate the roles of uterine glands in embryo survival and decidualization. Studies found that embryo survival and decidualization were compromised in glandless FOXA2-deficient mice on gestational day 6.5, resulting in abrupt pregnancy loss by day 7.5. These findings strongly support the hypothesis that uterine glands secrete factors other than LIF that impact embryo survival and stromal cell decidualization for pregnancy success.


Asunto(s)
Decidua/metabolismo , Pérdida del Embrión , Embrión de Mamíferos/embriología , Desarrollo Embrionario , Células del Estroma/metabolismo , Útero/metabolismo , Animales , Decidua/inmunología , Pérdida del Embrión/inmunología , Embrión de Mamíferos/inmunología , Desarrollo Embrionario/inmunología , Femenino , Factor Nuclear 3-beta del Hepatocito/deficiencia , Factor Inhibidor de Leucemia , Ratones , Embarazo , Resultado del Embarazo , Células del Estroma/inmunología , Transcriptoma , Útero/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...