Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Annu Rev Immunol ; 31: 635-674, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23330956

RESUMEN

To directly study complex human hemato-lymphoid system physiology and respective system-associated diseases in vivo, human-to-mouse xenotransplantation models for human blood and blood-forming cells and organs have been developed over the past three decades. We here review the fundamental requirements and the remarkable progress made over the past few years in improving these systems, the current major achievements reached by use of these models, and the future challenges to more closely model and study human health and disease and to achieve predictive preclinical testing of both prevention measures and potential new therapies.


Asunto(s)
Hematopoyesis/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/trasplante , Modelos Animales , Animales , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre Hematopoyéticas/tendencias , Humanos , Inmunofenotipificación , Tejido Linfoide/patología , Ratones , Investigación Biomédica Traslacional/métodos , Investigación Biomédica Traslacional/tendencias , Trasplante Heterólogo
2.
Immunity ; 54(2): 259-275.e7, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33382972

RESUMEN

The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Antígenos CD34/metabolismo , Biodiversidad , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Sangre Fetal/citología , Humanos , Receptores de Lipopolisacáridos/metabolismo , Pulmón/irrigación sanguínea , Receptores de IgG/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Nicho de Células Madre
3.
Immunity ; 51(5): 786-788, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31747578

RESUMEN

It is poorly understood how group 3 innate lymphoid cells (ILC3s) recognize metabolites produced by the gut microbiota. In this issue of Immunity, Chun et al. show that short-chain fatty acids sensed through the G protein-coupled receptor Ffar2 promote ILC3 function in the colon.


Asunto(s)
Inmunidad Innata , Linfocitos , Colon , Ácidos Grasos Volátiles , Receptores Acoplados a Proteínas G
4.
Immunity ; 48(1): 120-132.e8, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29343433

RESUMEN

Group 3 innate lymphoid cells (ILC3s) sense environmental signals and are critical for tissue integrity in the intestine. Yet, which signals are sensed and what receptors control ILC3 function remain poorly understood. Here, we show that ILC3s with a lymphoid-tissue-inducer (LTi) phenotype expressed G-protein-coupled receptor 183 (GPR183) and migrated to its oxysterol ligand 7α,25-hydroxycholesterol (7α,25-OHC). In mice lacking Gpr183 or 7α,25-OHC, ILC3s failed to localize to cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Gpr183 deficiency in ILC3s caused a defect in CP and ILF formation in the colon, but not in the small intestine. Localized oxysterol production by fibroblastic stromal cells provided an essential signal for colonic lymphoid tissue development, and inflammation-induced increased oxysterol production caused colitis through GPR183-mediated cell recruitment. Our findings show that GPR183 promotes lymphoid organ development and indicate that oxysterol-GPR183-dependent positioning within tissues controls ILC3 activity and intestinal homeostasis.


Asunto(s)
Colitis/metabolismo , Linfocitos/metabolismo , Tejido Linfoide/metabolismo , Oxiesteroles/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Movimiento Celular/genética , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/patología , Citocinas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ligandos , Linfocitos/patología , Tejido Linfoide/patología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
5.
Immunology ; 160(2): 126-138, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31715003

RESUMEN

Macrophages are tissue-resident myeloid cells with essential roles in host defense, tissue repair, and organ homeostasis. The lung harbors a large number of macrophages that reside in alveoli. As a result of their strategic location, alveolar macrophages are critical sentinels of healthy lung function and barrier immunity. They phagocytose inhaled material and initiate protective immune responses to pathogens, while preventing excessive inflammatory responses and tissue damage. Apart from alveolar macrophages, other macrophage populations are found in the lung and recent single-cell RNA-sequencing studies indicate that lung macrophage heterogeneity is greater than previously appreciated. The cellular origin and development of mouse lung macrophages has been extensively studied, but little is known about the ontogeny of their human counterparts, despite the importance of macrophages for lung health. In this context, humanized mice (mice with a human immune system) can give new insights into the biology of human lung macrophages by allowing in vivo studies that are not possible in humans. In particular, we have created humanized mouse models that support the development of human lung macrophages in vivo. In this review, we will discuss the heterogeneity, development, and homeostasis of lung macrophages. Moreover, we will highlight the impact of age, the microbiota, and pathogen exposure on lung macrophage function. Altered macrophage function has been implicated in respiratory infections as well as in common allergic and inflammatory lung diseases. Therefore, understanding the functional heterogeneity and ontogeny of lung macrophages should help to develop future macrophage-based therapies for important lung diseases in humans.


Asunto(s)
Diferenciación Celular/inmunología , Hipersensibilidad/inmunología , Enfermedades Pulmonares/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Factores de Edad , Alérgenos/inmunología , Animales , Exposición a Riesgos Ambientales/efectos adversos , Interacciones Huésped-Patógeno/inmunología , Humanos , Hipersensibilidad/terapia , Inmunoterapia/métodos , Pulmón/citología , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/terapia , Ratones , Microbiota/inmunología , Modelos Animales , Quimera por Trasplante/inmunología , Trasplante Heterólogo
6.
Proc Natl Acad Sci U S A ; 112(14): 4423-8, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25831514

RESUMEN

Prolonged T-cell receptor (TCR) signaling is required for the proliferation of T lymphocytes. Ligation of the TCR activates signaling, but also causes internalization of the TCR from the cell surface. How TCR signaling is sustained for many hours despite lower surface expression is unknown. Using genetic inhibition of endocytosis, we show here that TCR internalization promotes continued TCR signaling and T-lymphocyte proliferation. T-cell-specific deletion of dynamin 2, an essential component of endocytosis, resulted in reduced TCR signaling strength, impaired homeostatic proliferation, and the inability to undergo clonal expansion in vivo. Blocking endocytosis resulted in a failure to maintain mammalian target of rapamycin (mTOR) activity and to stably induce the transcription factor myelocytomatosis oncogene (c-Myc), which led to metabolic stress and a defect in cell growth. Our results support the concept that the TCR can continue to signal after it is internalized from the cell surface, thereby enabling sustained signaling and cell proliferation.


Asunto(s)
Dinamina II/fisiología , Endocitosis , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Animales , Anticuerpos Monoclonales/química , Autofagia , Membrana Celular/metabolismo , Proliferación Celular , Regulación de la Expresión Génica , Inmunoterapia , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-myc/metabolismo , Linfocitos T/citología , Serina-Treonina Quinasas TOR/metabolismo
7.
Proc Natl Acad Sci U S A ; 109(22): 8670-5, 2012 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-22592798

RESUMEN

The homeostasis of naive T cells is essential for protective immunity against infection, but the cell-intrinsic molecular mechanisms that control naïve T-cell homeostasis are poorly understood. Genetic ablation in lower organisms has revealed a critical role for Vps34, an evolutionary conserved class III phosphoinositide-3 kinase (PI3K), in regulating endocytosis and autophagy; however, the physiological function of Vps34 in the immune system, especially in T cells, is unclear. Here we report that Vps34 is required for the maintenance of naïve T cells, acting in a cell-intrinsic manner. T-cell-specific deletion of the gene encoding Vps34 resulted in reduced stability of Vps15 and Beclin-1, components of the class III PI3K complex, and impaired autophagy in T cells. Vps34 was dispensable for T-cell development but important for the survival of naïve T cells. Vps34-deficient T cells showed increased mitochondrial mass and accumulation of reactive oxygen species, consistent with deficient removal of damaged mitochondria. Thus, Vps34-dependent canonical autophagy plays a critical role in maintaining T-cell homeostasis by promoting T-cell survival through quality control of mitochondria.


Asunto(s)
Autofagia/fisiología , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Homeostasis/fisiología , Linfocitos T/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/genética , Beclina-1 , Western Blotting , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Fosfatidilinositol 3-Quinasas Clase III/genética , Citometría de Flujo , Homeostasis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/citología , Proteína de Clasificación Vacuolar VPS15/metabolismo
8.
Trends Immunol ; 32(7): 321-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21697012

RESUMEN

Human hemato-lymphoid-system mice hold great promise for modeling and studying important human diseases in vivo, and to enable vaccine testing. Until now, several major limitations have restricted the utility of human hemato-lymphoid-system mice in translational research. Recently, however, significant advances have been made in improving these mice, based on the delivery of human cytokines to create a better environment for human cells in the mouse host. In this review, we discuss the various approaches with a particular focus on improving human hemato-lymphoid-system mice by human cytokine knock-in gene replacement.


Asunto(s)
Citocinas/inmunología , Sistema Hematopoyético/inmunología , Tejido Linfoide/inmunología , Animales , Linaje de la Célula , Citocinas/genética , Técnicas de Sustitución del Gen , Sistema Hematopoyético/citología , Humanos , Tejido Linfoide/citología , Ratones , Investigación Biomédica Traslacional
9.
Proc Natl Acad Sci U S A ; 108(6): 2390-5, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262803

RESUMEN

Mice with a functional human immune system have the potential to allow in vivo studies of human infectious diseases and to enable vaccine testing. To this end, mice need to fully support the development of human immune cells, allow infection with human pathogens, and be capable of mounting effective human immune responses. A major limitation of humanized mice is the poor development and function of human myeloid cells and the absence of human immune responses at mucosal surfaces, such as the lung. To overcome this, we generated human IL-3/GM-CSF knock-in (hIL-3/GM-CSF KI) mice. These mice faithfully expressed human GM-CSF and IL-3 and developed pulmonary alveolar proteinosis because of elimination of mouse GM-CSF. We demonstrate that hIL-3/GM-CSF KI mice engrafted with human CD34(+) hematopoietic cells had improved human myeloid cell reconstitution in the lung. In particular, hIL-3/GM-CSF KI mice supported the development of human alveolar macrophages that partially rescued the pulmonary alveolar proteinosis syndrome. Moreover, human alveolar macrophages mounted correlates of a human innate immune response against influenza virus. The hIL-3/GM-CSF KI mice represent a unique mouse model that permits the study of human mucosal immune responses to lung pathogens.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interleucina-3/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Trasplante de Células Madre de Sangre del Cordón Umbilical , Técnicas de Sustitución del Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Inmunidad Mucosa/genética , Interleucina-3/genética , Pulmón/virología , Macrófagos Alveolares/virología , Ratones , Ratones Transgénicos , Modelos Inmunológicos , Infecciones por Orthomyxoviridae/genética , Quimera por Trasplante/genética , Quimera por Trasplante/inmunología , Quimera por Trasplante/virología , Trasplante Heterólogo
10.
Proc Natl Acad Sci U S A ; 108(6): 2378-83, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262827

RESUMEN

Hematopoietic stem cells (HSCs) both self-renew and give rise to all blood cells for the lifetime of an individual. Xenogeneic mouse models are broadly used to study human hematopoietic stem and progenitor cell biology in vivo. However, maintenance, differentiation, and function of human hematopoietic cells are suboptimal in these hosts. Thrombopoietin (TPO) has been demonstrated as a crucial cytokine supporting maintenance and self-renewal of HSCs. We generated RAG2(-/-)γ(c)(-/-) mice in which we replaced the gene encoding mouse TPO by its human homolog. Homozygous humanization of TPO led to increased levels of human engraftment in the bone marrow of the hosts, and multilineage differentiation of hematopoietic cells was improved, with an increased ratio of myelomonocytic verus lymphoid lineages. Moreover, maintenance of human stem and progenitor cells was improved, as demonstrated by serial transplantation. Therefore, RAG2(-/-)γ(c)(-/-) TPO-humanized mice represent a useful model to study human hematopoiesis in vivo.


Asunto(s)
Hematopoyesis , Trombopoyetina/metabolismo , Animales , Técnicas de Sustitución del Gen , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Ratones Transgénicos , Trombopoyetina/genética , Quimera por Trasplante/genética , Quimera por Trasplante/metabolismo , Trasplante Heterólogo
11.
Cells ; 12(2)2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36672175

RESUMEN

Oxysterols, derived from cholesterol oxidation, are formed either by autoxidation, via enzymes, or by both processes [...].


Asunto(s)
Oxiesteroles , Colesterol , Oxidación-Reducción
12.
Discov Immunol ; 2(1): kyad007, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38650756

RESUMEN

Innate lymphoid cells (ILCs) are sentinels of healthy organ function, yet it is unknown how ILCs adapt to distinct anatomical niches within tissues. Here, we used a unique humanized mouse model, MISTRG mice transplanted with human hematopoietic stem and progenitor cells (HSPCs), to define the gene signatures of human ILCs in the vascular versus the tissue (extravascular) compartment of the lung. Single-cell RNA sequencing in combination with intravascular cell labeling demonstrated that heterogeneous populations of human ILCs and natural killer (NK) cells occupied the vascular and tissue niches in the lung of HSPC-engrafted MISTRG mice. Moreover, we discovered that niche-specific cues shape the molecular programs of human ILCs in the distinct sub-anatomical compartments of the lung. Specifically, extravasation of ILCs into the lung tissue was associated with the upregulation of genes involved in the acquisition of tissue residency, cell positioning within the lung, sensing of tissue-derived signals, cellular stress responses, nutrient uptake, and interaction with other tissue-resident immune cells. We also defined a core tissue signature shared between human ILCs and NK cells in the extravascular space of the lung, consistent with imprinting by signals from the local microenvironment. The molecular characterization of human ILCs and NK cells in the vascular and tissue niches of the lung provides new knowledge on the mechanisms of ILC tissue adaptation and represents a resource for further studies.

13.
Proc Natl Acad Sci U S A ; 106(24): 9785-90, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19497879

RESUMEN

Cytolytic CD8(+) T cells (CTLs) kill virally infected cells, tumor cells, or other potentially autoreactive T cells in a calcium-dependent manner. To date, the molecular mechanism that leads to calcium intake during CTL differentiation and function has remained unresolved. We demonstrate that desmoyokin (AHNAK1) is expressed in mature CTLs, but not in naive CD8(+) T cells, and is critical for calcium entry required for their proper function during immune response. We show that mature AHNAK1-deficient CTLs exhibit reduced Ca(v)1.1 alpha1 subunit expression (also referred to as L-type calcium channels or alpha1S pore-forming subunits), which recently were suggested to play a role in calcium entry into CD4(+) T cells. AHNAK1-deficient CTLs show marked reduction in granzyme-B production, cytolytic activity, and IFN-gamma secretion after T cell receptor stimulation. Our results demonstrate an AHNAK1-dependent mechanism controlling calcium entry during CTL effector function.


Asunto(s)
Señalización del Calcio/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Linfocitos T/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática , Interferón gamma/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa , Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología
14.
J Exp Med ; 219(2)2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35019940

RESUMEN

Despite their importance in lung health and disease, it remains unknown how human alveolar macrophages develop early in life. Here we define the ontogeny of human alveolar macrophages from embryonic progenitors in vivo, using a humanized mouse model expressing human cytokines (MISTRG mice). We identified alveolar macrophage progenitors in human fetal liver that expressed the GM-CSF receptor CD116 and the transcription factor MYB. Transplantation experiments in MISTRG mice established a precursor-product relationship between CD34-CD116+ fetal liver cells and human alveolar macrophages in vivo. Moreover, we discovered circulating CD116+CD64-CD115+ macrophage precursors that migrated from the liver to the lung. Similar precursors were present in human fetal lung and expressed the chemokine receptor CX3CR1. Fetal CD116+CD64- macrophage precursors had a proliferative gene signature, outcompeted adult precursors in occupying the perinatal alveolar niche, and developed into functional alveolar macrophages. The discovery of the fetal alveolar macrophage progenitor advances our understanding of human macrophage origin and ontogeny.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Madre/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Feto , Expresión Génica , Genes myb , Humanos , Inmunohistoquímica , Inmunofenotipificación , Hígado/citología , Pulmón/citología , Ratones , Ratones Transgénicos , Células Madre/citología
15.
Front Immunol ; 13: 902881, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35967297

RESUMEN

Innate lymphoid cells (ILCs) play important roles in tissue homeostasis and host defense, but the proliferative properties and migratory behavior of especially human ILCs remain poorly understood. Here we mapped at single-cell resolution the spatial distribution of quiescent and proliferative human ILCs within the vascular versus tissue compartment. For this purpose, we employed MISTRG humanized mice as an in-vivo model to study human ILCs. We uncovered subset-specific differences in the proliferative status between vascular and tissue ILCs within lymphoid and non-lymphoid organs. We also identified CD117-CRTH2-CD45RA+ ILCs in the spleen that were highly proliferative and expressed the transcription factor TCF-1. These proliferative ILCs were present during the neonatal period in human blood and emerged early during population of the human ILC compartment in MISTRG mice transplanted with human hematopoietic stem and progenitor cells (HSPCs). Single-cell RNA-sequencing combined with intravascular cell labeling suggested that proliferative ILCs actively migrated from the local vasculature into the spleen tissue. Collectively, our comprehensive map reveals the proliferative topography of human ILCs, linking cell migration and spatial compartmentalization with cell division.


Asunto(s)
Inmunidad Innata , Linfocitos , Animales , Movimiento Celular , Humanos , Ratones
16.
Front Immunol ; 12: 752104, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867984

RESUMEN

Innate lymphoid cells (ILCs) contribute to immune defense, yet it is poorly understood how ILCs develop and are strategically positioned in the lung. This applies especially to human ILCs due to the difficulty of studying them in vivo. Here we investigated the ontogeny and migration of human ILCs in vivo with a humanized mouse model ("MISTRG") expressing human cytokines. In addition to known tissue-resident ILC subsets, we discovered CD5-expressing ILCs that predominantly resided within the lung vasculature and in the circulation. CD5+ ILCs contained IFNγ-producing mature ILC1s as well as immature ILCs that produced ILC effector cytokines under polarizing conditions in vitro. CD5+ ILCs had a distinct ontogeny compared to conventional CD5- ILCs because they first appeared in the thymus, spleen and liver rather than in the bone marrow after transplantation of MISTRG mice with human CD34+ hematopoietic stem and progenitor cells. Due to their strategic location, human CD5+ ILCs could serve as blood-borne sentinels, ready to be recruited into the lung to respond to environmental challenges. This work emphasizes the uniqueness of human CD5+ ILCs in terms of their anatomical localization and developmental origin compared to well-studied CD5- ILCs.


Asunto(s)
Antígenos CD5/inmunología , Pulmón/inmunología , Linfocitos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Movimiento Celular , Citocinas/inmunología , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunidad Innata , Masculino , Ratones , Persona de Mediana Edad , Bazo/inmunología
17.
Sci Immunol ; 6(56)2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33617461

RESUMEN

Immune cell differentiation is critical for adequate tissue-specific immune responses to occur. Here, we studied differentiation of human uterine natural killer cells (uNK cells). These cells reside in a tissue undergoing constant regeneration and represent the major leukocyte population at the maternal-fetal interface. However, their physiological response during the menstrual cycle and in pregnancy remains elusive. By surface proteome and transcriptome analysis as well as using humanized mice, we identify a differentiation pathway of uNK cells in vitro and in vivo with sequential acquisition of killer cell immunoglobulin-like receptors and CD39. uNK cell differentiation occurred continuously in response to the endometrial regeneration and was driven by interleukin-15. Differentiated uNK cells displayed reduced proliferative capacity and immunomodulatory function including enhanced angiogenic capacity. By studying human uterus transplantation and monozygotic twins, we found that the uNK cell niche could be replenished from circulation and that it was under genetic control. Together, our study uncovers a continuous differentiation pathway of human NK cells in the uterus that is coupled to profound functional changes in response to local tissue regeneration and pregnancy.


Asunto(s)
Diferenciación Celular/inmunología , Endometrio/inmunología , Células Asesinas Naturales/fisiología , Regeneración/inmunología , Animales , Antígenos de Diferenciación/genética , Endometrio/metabolismo , Femenino , Técnicas de Sustitución del Gen , Voluntarios Sanos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-15/metabolismo , Células Asesinas Naturales/trasplante , Estudios Longitudinales , Activación de Linfocitos , Ciclo Menstrual/inmunología , Ratones , Ratones Transgénicos , Embarazo , Progesterona/metabolismo , Receptores Inmunológicos/genética
18.
J Am Soc Nephrol ; 20(1): 86-94, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19005011

RESUMEN

Myosin 1e (Myo1e) is one of two Src homology 3 domain-containing "long-tailed" type I myosins in vertebrates, whose functions in health and disease are incompletely understood. Here, we demonstrate that Myo1e localizes to podocytes in the kidney. We generated Myo1e-knockout mice and found that they exhibit proteinuria, signs of chronic renal injury, and kidney inflammation. At the ultrastructural level, renal tissue from Myo1e-null mice demonstrates changes characteristic of glomerular disease, including a thickened and disorganized glomerular basement membrane and flattened podocyte foot processes. These observations suggest that Myo1e plays an important role in podocyte function and normal glomerular filtration.


Asunto(s)
Miosinas/fisiología , Podocitos/patología , Animales , Nitrógeno de la Urea Sanguínea , Tasa de Filtración Glomerular , Enfermedades Renales/etiología , Glomérulos Renales/patología , Ratones , Ratones Noqueados , Miosina Tipo I , Miosinas/análisis , Podocitos/química
19.
Biochem Pharmacol ; 174: 113672, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31634458

RESUMEN

Macrophages and innate lymphoid cells (ILCs) are tissue-resident cells that play important roles in organ homeostasis and tissue immunity. Their intricate relationship with the organs they reside in allows them to quickly respond to perturbations of organ homeostasis and environmental challenges, such as infection and tissue injury. Macrophages and ILCs have been extensively studied in mice, yet important species-specific differences exist regarding innate immunity between humans and mice. Complementary to ex-vivo studies with human cells, humanized mice (i.e. mice with a human immune system) offer the opportunity to study human macrophages and ILCs in vivo within their surrounding tissue microenvironments. In this review, we will discuss how humanized mice have helped gain new knowledge about the basic biology of these cells, as well as their function in infectious and malignant conditions. Furthermore, we will highlight active areas of investigation related to human macrophages and ILCs, such as their cellular heterogeneity, ontogeny, tissue residency, and plasticity. In the near future, we expect more fundamental discoveries in these areas through the combined use of improved humanized mouse models together with state-of-the-art technologies, such as single-cell RNA-sequencing and CRISPR/Cas9 genome editing.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Tejido Linfoide/inmunología , Macrófagos/inmunología , Modelos Animales , Animales , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , Enfermedades Transmisibles/genética , Enfermedades Transmisibles/inmunología , Modelos Animales de Enfermedad , Edición Génica , Humanos , Inmunidad Innata/genética , Tejido Linfoide/citología , Ratones , Neoplasias/genética , Neoplasias/inmunología , Especificidad de la Especie
20.
Front Immunol ; 10: 2010, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31507605

RESUMEN

Innate lymphoid cells (ILCs) are specialized immune cells that rapidly respond to environmental challenges, such as infection and tissue damage. ILCs play an important role in organ homeostasis, tissue repair, and host defense in the mucosal tissues intestine and lung. ILCs are sentinels of healthy tissue function, yet it is poorly understood how ILCs are recruited, strategically positioned, and maintained within tissues. Accordingly, ILC migration is an area that has recently come into focus and it is important to define the signals that control ILC migration to and within tissues. In this context, signals from the local tissue microenvironment are relevant. For example, ILCs in the intestine are exposed to an environment that is rich in dietary, microbial, and endogenous metabolites. It has been shown that the Vitamin A metabolite retinoic acid promotes ILC1 and ILC3 homing to the intestine. In addition, recent studies have discovered cholesterol metabolites (oxysterols) as a novel class of molecules that regulate ILC migration through the receptor GPR183. ILCs are considered to be largely tissue-resident cells, yet recent data indicate that ILCs actively migrate during inflammation. Furthermore, the discovery of circulating ILC precursors in humans and their presence within tissues has fueled the concept of local ILC-poiesis. However, it is unclear how circulating ILCs enter tissue during embryogenesis and inflammation and how they are directed to local tissue niches. In this review, I will discuss the metabolic signals that regulate ILC homing and their strategic positioning in healthy and inflamed tissues. It is becoming increasingly clear that ILC function is closely linked to their tissue localization. Therefore, understanding the tissue signals that control ILC migration could open new avenues for the treatment of chronic inflammatory diseases and cancer.


Asunto(s)
Movimiento Celular/inmunología , Metabolismo Energético , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Animales , Diferenciación Celular , Susceptibilidad a Enfermedades , Humanos , Especificidad de Órganos , Oxiesteroles/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA