Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.939
Filtrar
1.
Environ Sci Pollut Res Int ; 29(30): 45338-45348, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35143005

RESUMEN

Monosodium glutamate (MSG) is a flavor enhancer commonly used in modern nutrition. In this study, it was aimed to determine the effect of in ovo administered MSG on the embryonic development of thymus, bursa of Fabricius, and percentages of alpha-naphthyl acetate esterase (ANAE) positive lymphocyte by using histological, histometrical, and enzyme histochemical methods in chickens. For this purpose, 410 fertile eggs were used. The eggs were then divided into five groups: group 1 (control group, n = 40 eggs), group 2 (distilled water-injected group, n = 62 eggs), group 3 (0.12 mg/g egg MSG-injected group, n = 80 eggs), group 4 (0.6 mg/g egg MSG-injected group, n = 90 eggs), and group 5 (1.2 mg/g egg MSG-injected group, n = 138 eggs), and injections were performed via the egg yolk. On the 18th and 21st days of the incubation, the eggs were randomly opened from each group until six live embryos were obtained. The embryos of each group were sacrificed by decapitation, and blood, thymus, and bursa of Fabricius tissue samples were taken from the obtained embryos. The MSG-treated groups were found to be retarded embryonic development of thymus and bursa of Fabricius tissue compared to the control and distilled water groups. MSG treatment also resulted in reduced lymphoid follicles count and follicle diameters in bursa of Fabricius (P < 0.05). The percentage of peripheral blood ANAE positive lymphocytes was significantly lower in the MSG-treated groups than in the control and distilled water groups (P < 0.05). In conclusion, it has been found that in ovo administered MSG can adversely affect the embryonic development of thymus and bursa of Fabricius and decrease percentage of ANAE positive lymphocyte.


Asunto(s)
Bolsa de Fabricio , Pollos , Linfocitos , Timo , Animales , Bolsa de Fabricio/embriología , Embrión de Pollo , Desarrollo Embrionario , Naftol AS D Esterasa , Glutamato de Sodio/farmacología , Timo/embriología , Agua
2.
Pregnancy Hypertens ; 26: 116-120, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34741882

RESUMEN

OBJECTIVE: The immunological factors have role in the development of preeclampsia. The thymus is one of the main organs of the fetal immune system. The aim of this prospective clinical study was to investigate the association between fetal thymus volume and preeclampsia by adding the 3-dimensional measurement of thymus volume to the routine fetal ultrasound scan at 11-14 week of gestation. STUDY DESIGN: Totally 72 pregnant women in their first trimester of pregnancy were included and 3-D fetal thymus volume was measured with sonographic VOCAL programme. All women gestational period was followed. The data of women with preeclampsia (n = 10, study group) and without preeclampsia (n = 62, control group) were compared. MAIN OUTCOME MEASURES: Fetal thymus volume, preeclampsia development. RESULTS: Fetal thymus volume, mean gestational age at birth and newborn birthweight were found to be statistically lower in cases with preeclampsia compared with those without any complications. When the fetal thymus volume measured by the VOCAL programme in the study group was used as a marker for preeclampsia development, the limit value was 0.0375 cm3; sensitivity was 87.1% and specificity was 50% (AUC 85.3%, P < .001, 95% CI 0.751-0.949). As a result of binary logistic regression analysis; the low fetal thymus volume measured at 11-14 gestational weeks can be used as a predictive factor for preeclampsia (P < .001). CONCLUSIONS: According to the results of this study; the development of preeclampsia may be predicted by measuring 3-D fetal thymus volume at the first trimester.


Asunto(s)
Preeclampsia/diagnóstico , Timo/embriología , Adulto , Biomarcadores/análisis , Estudios de Casos y Controles , Femenino , Humanos , Imagenología Tridimensional/métodos , Embarazo , Primer Trimestre del Embarazo , Estudios Prospectivos , Sensibilidad y Especificidad , Timo/diagnóstico por imagen , Ultrasonografía Prenatal/métodos
3.
Aging Cell ; 20(10): e13477, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34596321

RESUMEN

Immunosenescence is a hallmark of aging and manifests as increased susceptibility to infection, autoimmunity, and cancer in the elderly. One component of immunosenescence is thymic involution, age-associated shrinkage of the thymus, observed in all vertebrates studied to date. The naked mole rat (Heterocephalus glaber) has become an attractive animal model in aging research due to its extreme longevity and resistance to disease. Here, we show that naked mole rats display no thymic involution up to 11 years of age. Furthermore, we found large ectopic cervical thymi in addition to the canonical thoracic thymus, both being identical in their cell composition. The developmental landscape in naked mole rat thymi revealed overt differences from the murine T-cell compartment, most notably a decrease of CD4+ /CD8+ double-positive cells and lower abundance of cytotoxic effector T cells. Our observations suggest that naked mole rats display a delayed immunosenescence. Therapeutic interventions aimed at reversing thymic aging remain limited, underscoring the importance of understanding the cellular and molecular mechanisms behind a sustained immune function in the naked mole rat.


Asunto(s)
Longevidad/fisiología , Timo/embriología , Animales , Modelos Animales de Enfermedad , Masculino , Ratas Topo , Ratas
4.
Nucleic Acids Res ; 49(10): 5760-5778, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34037780

RESUMEN

Alternative pre-mRNA splicing is a critical step to generate multiple transcripts, thereby dramatically enlarging the proteomic diversity. Thus, a common feature of most alternative splicing factor knockout models is lethality. However, little is known about lineage-specific alternative splicing regulators in a physiological setting. Here, we report that NSrp70 is selectively expressed in developing thymocytes, highest at the double-positive (DP) stage. Global splicing and transcriptional profiling revealed that NSrp70 regulates the cell cycle and survival of thymocytes by controlling the alternative processing of various RNA splicing factors, including the oncogenic splicing factor SRSF1. A conditional-knockout of Nsrp1 (NSrp70-cKO) using CD4Cre developed severe defects in T cell maturation to single-positive thymocytes, due to insufficient T cell receptor (TCR) signaling and uncontrolled cell growth and death. Mice displayed severe peripheral lymphopenia and could not optimally control tumor growth. This study establishes a model to address the function of lymphoid-lineage-specific alternative splicing factor NSrp70 in a thymic T cell developmental pathway.


Asunto(s)
Empalme Alternativo/genética , Carcinogénesis/metabolismo , Desarrollo Embrionario/genética , Hematopoyesis/genética , Melanoma/metabolismo , Timocitos/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Apoptosis/genética , Carcinogénesis/genética , Proliferación Celular/genética , Genómica , Células HEK293 , Humanos , Lectinas Tipo C/metabolismo , Linfopenia/genética , Linfopenia/metabolismo , Melanoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , RNA-Seq , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Antígenos de Linfocitos T/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Timo/embriología , Timo/metabolismo
5.
Front Immunol ; 12: 634367, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717173

RESUMEN

The thymus supports T-cell development via specialized microenvironments that ensure a diverse, functional and self-tolerant T-cell population. These microenvironments are classically defined as distinct cortex and medulla regions that each contain specialized subsets of stromal cells. Extensive research on thymic epithelial cells (TEC) within the cortex and medulla has defined their essential roles during T-cell development. Significantly, there are additional non-epithelial stromal cells (NES) that exist alongside TEC within thymic microenvironments, including multiple subsets of mesenchymal and endothelial cells. In contrast to our current understanding of TEC biology, the developmental origins, lineage relationships, and functional properties, of NES remain poorly understood. However, experimental evidence suggests these cells are important for thymus function by either directly influencing T-cell development, or by indirectly regulating TEC development and/or function. Here, we focus attention on the contribution of NES to thymic microenvironments, including their phenotypic identification and functional classification, and explore their impact on thymus function.


Asunto(s)
Comunicación Celular , Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Animales , Microambiente Celular , Células Endoteliales/inmunología , Humanos , Células Madre Mesenquimatosas/inmunología , Organogénesis , Fenotipo , Transducción de Señal , Timocitos/inmunología , Timo/embriología , Timo/inmunología
6.
Clin Radiol ; 76(7): 477-487, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33762135

RESUMEN

The appearance of the paediatric thymus changes as the normal process of thymic involution occurs. Thymic tissue may be orthotopic within the anterior mediastinum or ectopically located along the course of its embryological development. The variable appearance of orthotopic and ectopic thymic tissue in children on imaging studies may lead to misinterpretation of the normal thymus as pathology. Recognition of normal thymic tissue can mitigate unnecessary further diagnostic testing and patient anxiety. In this review, we discuss the embryological development and anatomical variants of normal thymus, and demonstrate the multimodality imaging features of the normal thymus in children, including positron-emission tomography, and diffusion-weighted imaging and in- and opposed-phase imaging on magnetic resonance imaging. We demonstrate the normal thymus mimicking pathological processes and discuss features that distinguish normal thymus, including thymic rebound hyperplasia, from pathology.


Asunto(s)
Coristoma/diagnóstico por imagen , Timo , Hiperplasia del Timo/diagnóstico por imagen , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Timo/diagnóstico por imagen , Timo/embriología , Timo/crecimiento & desarrollo
7.
Nat Commun ; 12(1): 1096, 2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33597545

RESUMEN

The thymus' key function in the immune system is to provide the necessary environment for the development of diverse and self-tolerant T lymphocytes. While recent evidence suggests that the thymic stroma is comprised of more functionally distinct subpopulations than previously appreciated, the extent of this cellular heterogeneity in the human thymus is not well understood. Here we use single-cell RNA sequencing to comprehensively profile the human thymic stroma across multiple stages of life. Mesenchyme, pericytes and endothelial cells are identified as potential key regulators of thymic epithelial cell differentiation and thymocyte migration. In-depth analyses of epithelial cells reveal the presence of ionocytes as a medullary population, while the expression of tissue-specific antigens is mapped to different subsets of epithelial cells. This work thus provides important insight on how the diversity of thymic cells is established, and how this heterogeneity contributes to the induction of immune tolerance in humans.


Asunto(s)
Células Epiteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Heterogeneidad Genética , Análisis de la Célula Individual/métodos , Timo/metabolismo , Adulto , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Pericitos/citología , Pericitos/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Timocitos/citología , Timocitos/metabolismo , Timo/citología , Timo/embriología
8.
Nat Immunol ; 22(2): 166-178, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33432227

RESUMEN

Type 2 innate lymphoid cells (ILC2) contribute to immune homeostasis, protective immunity and tissue repair. Here we demonstrate that functional ILC2 cells can arise in the embryonic thymus from shared T cell precursors, preceding the emergence of CD4+CD8+ (double-positive) T cells. Thymic ILC2 cells migrated to mucosal tissues, with colonization of the intestinal lamina propria. Expression of the transcription factor RORα repressed T cell development while promoting ILC2 development in the thymus. From RNA-seq, assay for transposase-accessible chromatin sequencing (ATAC-seq) and chromatin immunoprecipitation followed by sequencing (ChIP-seq) data, we propose a revised transcriptional circuit to explain the co-development of T cells and ILC2 cells from common progenitors in the thymus. When Notch signaling is present, BCL11B dampens Nfil3 and Id2 expression, permitting E protein-directed T cell commitment. However, concomitant expression of RORα overrides the repression of Nfil3 and Id2 repression, allowing ID2 to repress E proteins and promote ILC2 differentiation. Thus, we demonstrate that RORα expression represents a critical checkpoint at the bifurcation of the T cell and ILC2 lineages in the embryonic thymus.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Linaje de la Célula , Inmunidad Innata , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Técnicas de Cultivo de Órganos , Fenotipo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Timocitos/inmunología , Timo/embriología , Timo/inmunología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
9.
Exp Hematol ; 95: 1-12, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33454362

RESUMEN

T-Cell development is a major branch of lymphoid development and a key output of hematopoiesis, especially in early life, but the molecular requirements for T-cell potential have remained obscure. Considerable advances have now been made toward solving this problem through single-cell transcriptome studies, interfaced with in vitro differentiation assays that monitor potential efficiently at the single-cell level. This review focuses on a series of recent reports studying mouse and human early T-cell precursors, both in the developing fetus and in stringently purified postnatal samples of intrathymic and prethymic T-lineage precursors. Cross-comparison of results reveals a robustly conserved core program in mouse and human, but with some informative and provocative variations between species and between ontogenic states. Repeated findings are the multipotent progenitor regulatory signature of thymus-seeding cells and the proximity of the T-cell program to dendritic cell programs, especially to plasmacytoid dendritic cells in humans.


Asunto(s)
Hematopoyesis , Células Precursoras de Linfocitos T/citología , Análisis de la Célula Individual/métodos , Animales , Antígenos de Diferenciación de Linfocitos T/análisis , Linaje de la Célula , Movimiento Celular , Separación Celular , Células Cultivadas , Células Dendríticas/citología , Feto/citología , Feto/inmunología , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Humanos , Ratones , Células Madre Multipotentes/citología , Células Precursoras de Linfocitos T/clasificación , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Proteínas Represoras/fisiología , Especificidad de la Especie , Timo/citología , Timo/embriología , Timo/crecimiento & desarrollo , Transcriptoma , Proteínas Supresoras de Tumor/fisiología
11.
Blood ; 137(8): 1024-1036, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33025012

RESUMEN

During embryonic development, multiple waves of hematopoietic progenitors with distinct lineage potential are differentially regulated in time and space. Two different waves of thymic progenitors colonize the fetal thymus where they contribute to thymic organogenesis and homeostasis. The origin, the lineage differentiation potential of the first wave, and their relative contribution in shaping the thymus architecture, remained, however, unclear. Here, we show that the first wave of thymic progenitors comprises a unique population of bipotent T and innatel lymphoid cells (T/ILC), generating a lymphoid tissue inducer cells (LTi's), in addition to invariant Vγ5+ T cells. Transcriptional analysis revealed that innate lymphoid gene signatures and, more precisely, the LTi-associated transcripts were expressed in the first, but not in the second, wave of thymic progenitors. Depletion of early thymic progenitors in a temporally controlled manner showed that the progeny of the first wave is indispensable for the differentiation of autoimmune regulator-expressing medullary thymic epithelial cells (mTECs). We further show that these progenitors are of strict hematopoietic stem cell origin, despite the overlap between lymphopoiesis initiation and the transient expression of lymphoid-associated transcripts in yolk sac (YS) erythromyeloid-restricted precursors. Our work highlights the relevance of the developmental timing on the emergence of different lymphoid subsets, required for the establishment of a functionally diverse immune system.


Asunto(s)
Células Progenitoras Linfoides/citología , Linfocitos T/citología , Timo/citología , Timo/embriología , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Progenitoras Linfoides/metabolismo , Linfopoyesis , Ratones Endogámicos C57BL , Linfocitos T/metabolismo , Timo/metabolismo , Transcriptoma
12.
Acta Obstet Gynecol Scand ; 100(6): 1040-1050, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32865812

RESUMEN

INTRODUCTION: Infection and inflammation have been implicated in the etiology and subsequent morbidity associated with preterm birth. At present, there are no tests to assess for fetal compartment infection. The thymus, a gland integral in the fetal immune system, has been shown to involute in animal models of antenatal infection, but its response in human fetuses has not been studied. This study aims: (a) to generate magnetic resonance imaging (MRI) -derived fetal thymus volumes standardized for fetal weight; (b) to compare standardized thymus volumes from fetuses that delivered before 32 weeks of gestation with fetuses that subsequently deliver at term; (c) to assess thymus size as a predictor of preterm birth; and (d) to correlate the presence of chorioamnionitis and funisitis at delivery with thymic volumes in utero in fetuses that subsequently deliver preterm. MATERIAL AND METHODS: Women at high-risk of preterm birth at 20-32 weeks of gestation were recruited. A control group was obtained from existing data sets acquired as part of three research studies. A fetal MRI was performed on a 1.5T or 3T MRI scanner: T2 weighted images were obtained of the entire uterine content and specifically the fetal thorax. A slice-to-volume registration method was used for reconstruction of three-dimensional images of the thorax. Thymus segmentations were performed manually. Body volumes were calculated by manual segmentation and thymus:body volume ratios were generated. Comparison of groups was performed using multiple regression analysis. Normal ranges were created for thymus volume and thymus:body volume ratios using the control data. Receiver operating curves (ROC) curves were generated for thymus:body volume ratio and gestation-adjusted thymus volume centiles as predictors of preterm birth. Placental histology was analyzed where available from pregnancies that delivered very preterm and the presence of chorioamnionitis/funisitis was noted. RESULTS: Normative ranges were created for thymus volume, and thymus volume was standardized for fetal size from fetuses that subsequently delivered at term, but were imaged at 20-32 weeks of gestation. Image data sets from 16 women that delivered <32 weeks of gestation (ten with ruptured membranes and six with intact membranes) and 80 control women that delivered >37 weeks were included. Mean gestation at MRI of the study group was 28+4  weeks (SD 3.2) and for the control group was 25+5  weeks (SD 2.4). Both absolute fetal thymus volumes and thymus:body volume ratios were smaller in fetuses that delivered preterm (P < .001). Of the 16 fetuses that delivered preterm, 13 had placental histology, 11 had chorioamnionitis, and 9 had funisitis. The strongest predictors of prematurity were the thymus volume Z-score and thymus:body volume ratio Z-score (ROC areas 0.915 and 0.870, respectively). CONCLUSIONS: We have produced MRI-derived normal ranges for fetal thymus and thymus:body volume ratios between 20 and 32 weeks of gestation. Fetuses that deliver very preterm had reduced thymus volumes when standardized for fetal size. A reduced thymus volume was also a predictor of spontaneous preterm delivery. Thymus volume may be a suitable marker of the fetal inflammatory response, although further work is needed to assess this, increasing the sample size to correlate the extent of chorioamnionitis with thymus size.


Asunto(s)
Nacimiento Prematuro/diagnóstico por imagen , Timo/diagnóstico por imagen , Timo/fisiología , Ultrasonografía Prenatal/métodos , Adulto , Estudios de Casos y Controles , Femenino , Rotura Prematura de Membranas Fetales/diagnóstico por imagen , Edad Gestacional , Humanos , Recién Nacido , Imagen por Resonancia Magnética , Tamaño de los Órganos/fisiología , Proyectos Piloto , Embarazo , Embarazo de Alto Riesgo , Timo/embriología , Timo/patología
13.
PLoS One ; 15(12): e0243842, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33320884

RESUMEN

Activation of the aryl hydrocarbon receptor (AHR) by the environmental toxin dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD) causes diverse toxicities, including thymus atrophy and hepatosteatosis. The mechanisms by which AHR activation by TCDD leads to these toxicities are not fully understood. Here we studied the effects of TCDD on a major energy pathway, glycolysis, using the chick embryo close to hatching, a well-established model for studying dioxin toxicity. We showed that 24 hr of TCDD treatment causes changes in glycolysis in both thymus and liver. In thymus glands, TCDD decreased mRNAs for glycolytic genes and glucose transporters, glycolytic indices and levels of IL7 mRNA, phosphorylated AKT (pAKT) and HIF1A, stimulators of glycolysis and promoters of survival and proliferation of thymic lymphocytes. In contrast, in liver, TCDD increased mRNA levels for glycolytic genes and glucose transporters, glycolytic endpoints and pAKT levels. Similarly, increases by TCDD in mRNA levels for glycolytic genes and glucose transporters in human primary hepatocytes showed that effects in chick embryo liver pertain also to human cells. Treatment with the glycolytic inhibitor 2-deoxy-d-glucose exacerbated the effects on thymus atrophy by TCDD, supporting a role for decreased glycolysis in thymus atrophy by TCDD, but did not prevent hepatosteatosis. NAD+ precursors abolished TCDD effects on glycolytic endpoints in both thymus and liver. In summary, we report here that dioxin disrupts glycolysis mediated energy metabolism in both thymus and liver, and that it does so in opposite ways, decreasing it in the thymus and increasing it in the liver. Further, the findings support NAD+ boosting as a strategy against metabolic effects of environmental pollutants such as dioxins.


Asunto(s)
Dioxinas/toxicidad , Glucólisis/efectos de los fármacos , Especificidad de Órganos/efectos de los fármacos , Receptores de Hidrocarburo de Aril/metabolismo , Adulto , Animales , Células Cultivadas , Embrión de Pollo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Glucólisis/genética , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Compuestos de Piridinio , ARN Mensajero/genética , ARN Mensajero/metabolismo , Timo/efectos de los fármacos , Timo/embriología , Timo/metabolismo
14.
Front Immunol ; 11: 1837, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32922396

RESUMEN

Combined Immunodeficiencies (CID) are rare congenital disorders characterized by defective T-cell development that may be associated with B- and NK-cell deficiency. They are usually due to alterations in genes expressed in hematopoietic precursors but in few cases, they are caused by impaired thymic development. Athymia was classically associated with DiGeorge Syndrome due to TBX1 gene haploinsufficiency. Other genes, implicated in thymic organogenesis include FOXN1, associated with Nude SCID syndrome, PAX1, associated with Otofaciocervical Syndrome type 2, and CHD7, one of the genes implicated in CHARGE syndrome. More recently, chromosome 2p11.2 microdeletion, causing FOXI3 haploinsufficiency, has been identified in 5 families with impaired thymus development. In this review, we will summarize the main genetic, clinical, and immunological features related to the abovementioned gene mutations. We will also focus on different therapeutic approaches to treat SCID in these patients.


Asunto(s)
Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/patología , Linfocitos T/inmunología , Timo/anomalías , Humanos , Timo/embriología , Timo/inmunología
15.
Viruses ; 12(9)2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32911797

RESUMEN

Maternal influenza A viral infections in humans are associated with low birth weight, increased risk of pre-term birth, stillbirth and congenital defects. To examine the effect of maternal influenza virus infection on placental and fetal growth, pregnant C57BL/6 mice were inoculated intranasally with influenza A virus A/CA/07/2009 pandemic H1N1 or phosphate-buffered saline (PBS) at E3.5, E7.5 or E12.5, and the placentae and fetuses collected and weighed at E18.5. Fetal thymuses were pooled from each litter. Placentae were examined histologically, stained by immunohistochemistry (IHC) for CD34 (hematopoietic progenitor cell antigen) and vascular channels quantified. RNA from E7.5 and E12.5 placentae and E7.5 fetal thymuses was subjected to RNA sequencing and pathway analysis. Placental weights were decreased in litters inoculated with influenza at E3.5 and E7.5. Placentae from E7.5 and E12.5 inoculated litters exhibited decreased labyrinth development and the transmembrane protein 150A gene was upregulated in E7.5 placentae. Fetal weights were decreased in litters inoculated at E7.5 and E12.5 compared to controls. RNA sequencing of E7.5 thymuses indicated that 957 genes were downregulated ≥2-fold including Mal, which is associated with Toll-like receptor signaling and T cell differentiation. There were 28 upregulated genes. It is concluded that maternal influenza A virus infection impairs fetal thymic gene expression as well as restricting placental and fetal growth.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/genética , Gripe Humana/fisiopatología , Placenta/metabolismo , Efectos Tardíos de la Exposición Prenatal/genética , Timo/metabolismo , Transcriptoma , Animales , Femenino , Desarrollo Fetal , Regulación del Desarrollo de la Expresión Génica , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/embriología , Gripe Humana/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Placenta/virología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/virología , Timo/embriología
16.
Int J Mol Sci ; 21(16)2020 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-32796710

RESUMEN

The thymus generates central immune tolerance by producing self-restricted and self-tolerant T-cells as a result of interactions between the developing thymocytes and the stromal microenvironment, mainly formed by the thymic epithelial cells. The thymic epithelium derives from the endoderm of the pharyngeal pouches, embryonic structures that rely on environmental cues from the surrounding mesenchyme for its development. Here, we review the most recent advances in our understanding of the molecular mechanisms involved in early thymic organogenesis at stages preceding the expression of the transcription factor Foxn1, the early marker of thymic epithelial cells identity. Foxn1-independent developmental stages, such as the specification of the pharyngeal endoderm, patterning of the pouches, and thymus fate commitment are discussed, with a special focus on epithelial-mesenchymal interactions.


Asunto(s)
Organogénesis , Timo/crecimiento & desarrollo , Animales , Humanos , Transducción de Señal , Timo/embriología , Factores de Transcripción/metabolismo
17.
Front Immunol ; 11: 1656, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849572

RESUMEN

Innate and adaptive immune systems continuously interchange information and orchestrate their immune responses to protect the host. γδT cells play crucial roles, as they incorporate both innate and adaptive immune characteristics. Dendritic epidermal T cells (DETC) are specialized γδT cells, which are uniquely positioned to rapidly respond to skin wounds and infections. Their elongated dendrite morphology allows them to be in continuous contact with multiple neighboring keratinocytes and Langerhans cells. Cellular interactions are fundamental to the formation, activation and maintenance of immune cell functions during steady state and pathology. Recent technological advances, especially in the field of cellular imaging, have contributed greatly to the characterization of complex cellular interactions in a spatiotemporally resolved manner. In this review, we will highlight the often-underappreciated function of DETC and other γδT cells during steady state and an ongoing immune response. More specifically, we discuss how DETC-precursors are shaped in the fetal thymus during embryogenesis as well as how direct cell-cell interactions of DETC with neighboring epidermal cells shape skin homeostasis and effector functions. Furthermore, we will discuss seminal work and recent discoveries made in the γδT cell field, which have highlighted the importance of γδT cells in the skin, both in humans and mice.


Asunto(s)
Células Epidérmicas/inmunología , Epidermis/inmunología , Linfocitos Intraepiteliales/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Inmunidad Adaptativa , Animales , Comunicación Celular , Microambiente Celular , Desarrollo Embrionario , Células Epidérmicas/metabolismo , Epidermis/metabolismo , Humanos , Inmunidad Innata , Linfocitos Intraepiteliales/metabolismo , Fenotipo , Transducción de Señal , Timo/embriología , Timo/inmunología , Timo/metabolismo
18.
J Clin Immunol ; 40(6): 791-806, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32638194

RESUMEN

Down syndrome fits an immunophenotype of combined immunodeficiency with immunodysregulation, manifesting with increased susceptibility to infections, autoimmunity, autoinflammatory diseases, and hematologic malignancies. Qualitative and quantitative alterations in innate and adaptive immunity are found in most individuals with Down syndrome. However, there is substantial heterogeneity and no correlation between immunophenotype and clinical presentation. Previously, it was thought that the immunological changes in Down syndrome were caused by precocious aging. We emphasize in this review that the immune system in Down syndrome is intrinsically different from the very beginning. The overexpression of specific genes located on chromosome 21 contributes to immunodeficiency and immunodysregulation, but gene expression differs between genes located on chromosome 21 and depends on tissue and cell type. In addition, trisomy 21 results in gene dysregulation of the whole genome, reflecting the complex nature of this syndrome in comparison to well-known inborn errors of immunity that result from monogenic germline mutations. In this review, we provide an updated overview focusing on inborn errors of adaptive immunity in Down syndrome.


Asunto(s)
Inmunidad Adaptativa/genética , Síndrome de Down/genética , Síndrome de Down/inmunología , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/inmunología , Animales , Autoinmunidad/genética , Biomarcadores , Relación CD4-CD8 , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad Innata/genética , Mediadores de Inflamación/metabolismo , Interferones/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Modelos Biológicos , Receptores de Antígenos de Linfocitos T , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/embriología , Timo/inmunología , Timo/metabolismo
19.
Front Immunol ; 11: 830, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32431714

RESUMEN

22q11.2 deletion syndrome (DiGeorge), CHARGE syndrome, Nude/SCID and otofaciocervical syndrome type 2 (OTFCS2) are distinct clinical conditions in humans that can result in hypoplasia and occasionally, aplasia of the thymus. Thymic hypoplasia/aplasia is first suggested by absence or significantly reduced numbers of recent thymic emigrants, revealed in standard-of-care newborn screens for T cell receptor excision circles (TRECs). Subsequent clinical assessments will often indicate whether genetic mutations are causal to the low T cell output from the thymus. However, the molecular mechanisms leading to the thymic hypoplasia/aplasia in diverse human syndromes are not fully understood, partly because the problems of the thymus originate during embryogenesis. Rodent and Zebrafish models of these clinical syndromes have been used to better define the underlying basis of the clinical presentations. Results from these animal models are uncovering contributions of different cell types in the specification, differentiation, and expansion of the thymus. Cell populations such as epithelial cells, mesenchymal cells, endothelial cells, and thymocytes are variably affected depending on the human syndrome responsible for the thymic hypoplasia. In the current review, findings from the diverse animal models will be described in relation to the clinical phenotypes. Importantly, these results are suggesting new strategies for regenerating thymic tissue in patients with distinct congenital disorders.


Asunto(s)
Síndrome Branquio Oto Renal/complicaciones , Síndrome CHARGE/complicaciones , Síndrome de DiGeorge/complicaciones , Síndromes de Inmunodeficiencia/etiología , Inmunodeficiencia Combinada Grave/complicaciones , Timo/anomalías , Animales , Síndrome Branquio Oto Renal/genética , Síndrome Branquio Oto Renal/inmunología , Síndrome CHARGE/genética , Síndrome CHARGE/inmunología , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/inmunología , Modelos Animales de Enfermedad , Humanos , Síndromes de Inmunodeficiencia/inmunología , Ratones , Mutación , Ratas , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Timo/embriología , Timo/inmunología , Pez Cebra
20.
Curr Top Dev Biol ; 139: 205-238, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32450961

RESUMEN

Boolean approaches and extensions thereof are becoming increasingly popular to model signaling and regulatory networks, including those controlling cell differentiation, pattern formation and embryonic development. Here, we describe a logical modeling framework relying on three steps: the delineation of a regulatory graph, the specification of multilevel components, and the encoding of Boolean rules specifying the behavior of model components depending on the levels or activities of their regulators. Referring to a non-deterministic, asynchronous updating scheme, we present several complementary methods and tools enabling the computation of stable activity patterns, the verification of the reachability of such patterns, as well as the generation of mean temporal evolution curves and the computation of the probabilities to reach distinct activity patterns. We apply this logical framework to the regulatory network controlling T lymphocyte specification. This process involves cross-regulations between specific T cell regulatory factors and factors driving alternative differentiation pathways, which remain accessible during the early steps of thymocyte development. Many transcription factors needed for T cell specification are required in other hematopoietic differentiation pathways and are combined in a fine-tuned, time-dependent fashion to achieve T cell commitment. Using the software GINsim, we integrated current knowledge into a dynamical model, which recapitulates the main developmental steps from early progenitors entering the thymus up to T cell commitment, as well as the impact of various documented environmental and genetic perturbations. Our model analysis further enabled the identification of several knowledge gaps. The model, software and whole analysis workflow are provided in computer-readable and executable form to ensure reproducibility and ease extensions.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Modelos Genéticos , Linfocitos T/metabolismo , Timo/metabolismo , Animales , Simulación por Computador , Linfocitos T/citología , Timocitos/citología , Timocitos/metabolismo , Timo/citología , Timo/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...