Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PLoS Genet ; 17(6): e1009605, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34081701

RESUMEN

Homozygous mutation of the Csf1r locus (Csf1rko) in mice, rats and humans leads to multiple postnatal developmental abnormalities. To enable analysis of the mechanisms underlying the phenotypic impacts of Csf1r mutation, we bred a rat Csf1rko allele to the inbred dark agouti (DA) genetic background and to a Csf1r-mApple reporter transgene. The Csf1rko led to almost complete loss of embryonic macrophages and ablation of most adult tissue macrophage populations. We extended previous analysis of the Csf1rko phenotype to early postnatal development to reveal impacts on musculoskeletal development and proliferation and morphogenesis in multiple organs. Expression profiling of 3-week old wild-type (WT) and Csf1rko livers identified 2760 differentially expressed genes associated with the loss of macrophages, severe hypoplasia, delayed hepatocyte maturation, disrupted lipid metabolism and the IGF1/IGF binding protein system. Older Csf1rko rats developed severe hepatic steatosis. Consistent with the developmental delay in the liver Csf1rko rats had greatly-reduced circulating IGF1. Transfer of WT bone marrow (BM) cells at weaning without conditioning repopulated resident macrophages in all organs, including microglia in the brain, and reversed the mutant phenotypes enabling long term survival and fertility. WT BM transfer restored osteoclasts, eliminated osteopetrosis, restored bone marrow cellularity and architecture and reversed granulocytosis and B cell deficiency. Csf1rko rats had an elevated circulating CSF1 concentration which was rapidly reduced to WT levels following BM transfer. However, CD43hi non-classical monocytes, absent in the Csf1rko, were not rescued and bone marrow progenitors remained unresponsive to CSF1. The results demonstrate that the Csf1rko phenotype is autonomous to BM-derived cells and indicate that BM contains a progenitor of tissue macrophages distinct from hematopoietic stem cells. The model provides a unique system in which to define the pathways of development of resident tissue macrophages and their local and systemic roles in growth and organ maturation.


Asunto(s)
Hígado Graso/genética , Macrófagos/metabolismo , Anomalías Musculoesqueléticas/genética , Desarrollo Musculoesquelético/genética , Osteopetrosis/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Embrión de Mamíferos , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/terapia , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Genes Reporteros , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/deficiencia , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/genética , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología , Macrófagos/patología , Masculino , Anomalías Musculoesqueléticas/metabolismo , Anomalías Musculoesqueléticas/patología , Anomalías Musculoesqueléticas/terapia , Osteopetrosis/metabolismo , Osteopetrosis/patología , Osteopetrosis/terapia , Ratas , Ratas Transgénicas , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia
2.
Biol Reprod ; 104(5): 995-1007, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33524138

RESUMEN

Colony-stimulating factor 2 (CSF2) functions in the reproductive tract to modulate the function of the preimplantation embryo. The ß subunit of the CSF2 receptor (CSF2RB) is not expressed in the embryo, and signal transduction is therefore different than for myeloid cells where the receptor is composed of α (CSF2RA) and ß subunits. Here, we produced embryos in which exons 5 and 6 of CSF2RA were disrupted using the CRISPR/Cas 9 system to test whether CSF2RA signaling was essential for actions of CSF2 in the bovine embryo. Wild-type and CSF2RA knockout embryos were treated with 10 ng/mL CSF2 or vehicle at day 5 of development. Blastocysts were harvested at day 8 to determine transcript abundance of 90 genes by real-time polymerase chain reaction (PCR). Responses in female blastocysts were examined separately from male blastocysts because actions of CSF2 are sex-dependent. For wild-type embryos, CSF2 altered expression of 10 genes in females and 20 in males. Only three genes were affected by CSF2 in a similar manner for both sexes. Disruption of CSF2RA prevented the effect of CSF2 on expression for 9 of 10 CSF2-regulated genes in females and 19 of 20 genes in males. The results confirm the importance of CSF2RA for regulation of gene expression by CSF2 in the blastocyst.


Asunto(s)
Blastocisto/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Animales , Sistemas CRISPR-Cas , Bovinos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
3.
Neurobiol Dis ; 151: 105268, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33450391

RESUMEN

Mutations in the human CSF1R gene have been associated with dominant and recessive forms of neurodegenerative disease. Here we describe the impacts of Csf1r mutation in the rat on development of the brain. Diffusion imaging indicated small reductions in major fiber tracts that may be associated in part with ventricular enlargement. RNA-seq profiling revealed a set of 105 microglial markers depleted in all brain regions of the Csf1rko rats. There was no evidence of region or sex-specific expression of microglia-associated transcripts. Other than the microglial signature, Csf1rko had no effect on any neuronal or region-specific transcript cluster. Expression of markers of oligodendrocytes, astrocytes, dopaminergic neurons and Purkinje cells was minimally affected. However, there were defects in dendritic arborization of doublecortin-positive neurogenic precursors and expression of poly-sialylated neural cell adhesion molecule (PS-NCAM) in the dentate gyrus of the hippocampus. Heterozygous Csf1rko rats had no detectable brain phenotype. We conclude that most brain developmental processes occur normally in the absence of microglia and that CSF1R haploinsufficiency is unlikely to cause leukoencephalopathy.


Asunto(s)
Microglía , Enfermedades Neurodegenerativas/genética , Neurogénesis/fisiología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Mutación , Ratas , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
4.
J Allergy Clin Immunol ; 143(4): 1513-1524.e2, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30244025

RESUMEN

BACKGROUND: Eosinophils are a therapeutic target in asthmatic patients, and GM-CSF has been suggested to control various aspects of eosinophil biology, including development, function, and survival. However, to date, the role of GM-CSF signaling in eosinophils in vivo is largely unclear. OBJECTIVE: We sought to elucidate the role of GM-CSF signaling in asthmatic inflammation. METHODS: Wild-type and GM-CSF receptor α (Csf2ra)-deficient mice reconstituted with Csf2ra-proficient alveolar macrophages were subjected to different models of airway inflammation to evaluate the effect of GM-CSF signaling deficiency on asthmatic inflammation in general and on eosinophils in particular. RESULTS: We demonstrate that GM-CSF signaling, although being largely dispensable for eosinophil development at steady state, intrinsically promotes accumulation of eosinophils in the lung during allergic airway inflammation. In contrast, chitin-induced eosinophil accumulation in the peritoneal cavity occurs independent of GM-CSF, indicating organ specificity. We show that GM-CSF induces chemokinesis and promotes eosinophil survival in vitro, which likely contribute to eosinophil accumulation in the airways in vivo. CONCLUSION: GM-CSF intrinsically promotes eosinophil accumulation in the setting of pulmonary allergic inflammation.


Asunto(s)
Eosinófilos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Eosinofilia Pulmonar/inmunología , Hipersensibilidad Respiratoria/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia
5.
J Immunol ; 201(9): 2683-2699, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30249809

RESUMEN

We have produced Csf1r-deficient rats by homologous recombination in embryonic stem cells. Consistent with the role of Csf1r in macrophage differentiation, there was a loss of peripheral blood monocytes, microglia in the brain, epidermal Langerhans cells, splenic marginal zone macrophages, bone-associated macrophages and osteoclasts, and peritoneal macrophages. Macrophages of splenic red pulp, liver, lung, and gut were less affected. The pleiotropic impacts of the loss of macrophages on development of multiple organ systems in rats were distinct from those reported in mice. Csf1r-/- rats survived well into adulthood with postnatal growth retardation, distinct skeletal and bone marrow abnormalities, infertility, and loss of visceral adipose tissue. Gene expression analysis in spleen revealed selective loss of transcripts associated with the marginal zone and, in brain regions, the loss of known and candidate novel microglia-associated transcripts. Despite the complete absence of microglia, there was little overt phenotype in brain, aside from reduced myelination and increased expression of dopamine receptor-associated transcripts in striatum. The results highlight the redundant and nonredundant functions of CSF1R signaling and of macrophages in development, organogenesis, and homeostasis.


Asunto(s)
Macrófagos , Microglía , Organogénesis/genética , Ratas/crecimiento & desarrollo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Animales , Modelos Animales , Mutación , Ratas/genética
6.
Nat Commun ; 9(1): 2830, 2018 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-30026565

RESUMEN

Circuit refinement during postnatal development is finely regulated by neuron-neuron interactions. Recent studies suggest participation of microglia in this process but it is unclear how microglia cooperatively act with neuronal mechanisms. To examine roles of microglia, we ablate microglia by microglia-selective deletion of colony-stimulating factor 1 receptor (Csf1r) by crossing floxed-Csf1r and Iba1-iCre mice (Csf1r-cKO). In Csf1r-cKO mice, refinement of climbing fiber (CF) to Purkinje cell (PC) innervation after postnatal day 10 (P10)-P12 is severely impaired. However, there is no clear morphological evidence suggesting massive engulfment of CFs by microglia. In Csf1r-cKO mice, inhibitory synaptic transmission is impaired and CF elimination is restored by diazepam, which suggests that impairment of CF elimination is caused by a defect of GABAergic inhibition on PCs, a prerequisite for CF elimination. These results indicate that microglia primarily promote GABAergic inhibition and secondarily facilitate the mechanism for CF elimination inherent in PCs.


Asunto(s)
Envejecimiento/genética , Cerebelo/metabolismo , Neuronas GABAérgicas/metabolismo , Microglía/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Comunicación Celular , Cerebelo/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Cerebelo/patología , Diazepam/farmacología , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/patología , Expresión Génica , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/patología , Fibras Nerviosas Mielínicas/patología , Neurogénesis/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Sinapsis/ultraestructura , Transmisión Sináptica
7.
Methods Mol Biol ; 1784: 243-258, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29761404

RESUMEN

Macrophages are a heterogeneous population of innate immune cells and are distributed in most adult tissues. Certain tissue-resident macrophages with a prenatal origin, together with postnatal monocyte-derived macrophages, serve as the host scavenger system to eliminate invading pathogens, malignant cells, senescent cells, dead cells, cellular debris, and other foreign substances. As a key member of the mononuclear phagocyte system, macrophages play essential roles in regulation of prenatal development, tissue homeostasis, and disease progression. Over the past two decades, considerable efforts have been made to generate genetic models of macrophage ablation in mice. These models support investigations of the precise functions of tissue-specific macrophages under physiological and pathological conditions. Herein, we overview the currently available mouse strains for in vivo genetic ablation of macrophages and discuss their respective advantages and limitations.


Asunto(s)
Macrófagos/citología , Modelos Genéticos , Fagocitos/citología , Animales , Linaje de la Célula/genética , Modelos Animales de Enfermedad , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Especificidad de Órganos/genética , Fagocitos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
9.
Sci Rep ; 7: 43469, 2017 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-28233860

RESUMEN

Pulmonary alveolar proteinosis (PAP) is a rare pulmonary disease in which the abnormalities in alveolar surfactant accumulation are caused by impairments of GM-CSF pathway attributing to defects in a variety of genes. However, hereditary PAP is extremely uncommon and a detailed understanding in the genetic inheritance of PAP in a family may provide timely diagnosis, treatment and proper intervention including genetic consultation. Here, we described a comprehensive analysis of genome and gene expression for a family containing one affected child with a diagnosis of PAP and two other healthy siblings. Family-based whole-genome analysis revealed a homozygous deletion that disrupts CSF2RA, CRLF2, and IL3RA gene in the pseudoautosomal region of the X chromosome in the affected child and one of asymptomatic siblings. Further functional pathway analysis of differentially expressed genes in IL-1ß-treated peripheral blood mononuclear cells highlighted the insufficiency of immune response in the child with PAP, especially the protection against bacterial infection. Collectively, our results reveal a novel allele as the genetic determinant of a family with PAP and provide insights into variable expressivity and incomplete penetrance of this rare disease, which will be helpful for proper genetic consultation and prompt treatment to avoid mortality and morbidity.


Asunto(s)
Cromosomas Humanos X/química , Enfermedades Genéticas Congénitas/genética , Subunidad alfa del Receptor de Interleucina-3/deficiencia , Proteinosis Alveolar Pulmonar/genética , Receptores de Citocinas/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Enfermedades Asintomáticas , Niño , Preescolar , Femenino , Eliminación de Gen , Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/inmunología , Enfermedades Genéticas Congénitas/patología , Homocigoto , Humanos , Interleucina-1beta/farmacología , Subunidad alfa del Receptor de Interleucina-3/genética , Subunidad alfa del Receptor de Interleucina-3/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Masculino , Linaje , Penetrancia , Cultivo Primario de Células , Proteinosis Alveolar Pulmonar/diagnóstico , Proteinosis Alveolar Pulmonar/inmunología , Proteinosis Alveolar Pulmonar/patología , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Índice de Severidad de la Enfermedad , Hermanos , Secuenciación Completa del Genoma
10.
Angiogenesis ; 19(4): 513-24, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27464987

RESUMEN

Lymphatic vessels play important roles in fluid drainage and in immune responses, as well as in pathological processes including cancer progression and inflammation. While the molecular regulation of the earliest lymphatic vessel differentiation and development has been investigated in much detail, less is known about the control and timing of lymphatic vessel maturation in different organs, which often occurs postnatally. We investigated the time course of lymphatic vessel development on the pleural side of the diaphragmatic muscle in mice, the so-called submesothelial initial diaphragmatic lymphatic plexus. We found that this lymphatic network develops largely after birth and that it can serve as a reliable and easily quantifiable model to study physiological lymphangiogenesis in vivo. Lymphangiogenic growth in this tissue was highly dependent on vascular endothelial growth factor receptor (VEGFR)-3 signaling, whereas VEGFR-1 and -2 signaling was dispensable. During diaphragm development, macrophages appeared first in a linearly arranged pattern, followed by ingrowth of lymphatic vessels along these patterned lines. Surprisingly, ablation of macrophages in colony-stimulating factor-1 receptor (Csf1r)-deficient mice and by treatment with a CSF-1R-blocking antibody did not inhibit the general lymphatic vessel development in the diaphragm but specifically promoted branch formation of lymphatic sprouts. In agreement with these findings, incubation of cultured lymphatic endothelial cells with conditioned medium from P7 diaphragmatic macrophages significantly reduced LEC sprouting. These results indicate that the postnatal diaphragm provides a suitable model for studies of physiological lymphangiogenic growth and maturation, and for the identification of modulators of lymphatic vessel growth.


Asunto(s)
Diafragma/crecimiento & desarrollo , Linfangiogénesis/fisiología , Macrófagos/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Medios de Cultivo Condicionados , Diafragma/citología , Diafragma/fisiología , Femenino , Vasos Linfáticos/citología , Vasos Linfáticos/fisiología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Transducción de Señal , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
11.
Arch Bronconeumol ; 51(7): 344-9, 2015 Jul.
Artículo en Inglés, Español | MEDLINE | ID: mdl-25896950

RESUMEN

Pulmonary alveolar proteinosis (PAP) is a rare disease characterized by the accumulation of surfactant-like lipoproteinaceous material in the distal air spaces and terminal bronchi, which may lead to impaired gas exchange. This accumulation of surfactant is due to decreased clearance by the alveolar macrophages. Its primary, most common form, is currently considered an autoimmune disease. Better knowledge of the causes of PAP have led to the emergence of alternatives to whole lung lavage, although this is still considered the treatment of choice. Most studies are case series, often with limited patient numbers, so the level of evidence is low. Since the severity of presentation and clinical course are variable, not all patients will require treatment. Due to the low level of evidence, some objective criteria based on expert opinion have been arbitrarily proposed in an attempt to define in which patients it is best to initiate treatment.


Asunto(s)
Enfermedades Autoinmunes/terapia , Proteinosis Alveolar Pulmonar/terapia , Adulto , Aloinjertos , Animales , Profilaxis Antibiótica , Anticuerpos Neutralizantes/análisis , Anticuerpos Neutralizantes/inmunología , Autoanticuerpos/análisis , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Ensayos Clínicos como Asunto , Terapia Combinada , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoglobulina G/análisis , Inmunoglobulina G/inmunología , Trasplante de Pulmón , Macrófagos/patología , Ratones , Ratones Noqueados , Terapia por Inhalación de Oxígeno , Plasmaféresis , Estudios Prospectivos , Proteinosis Alveolar Pulmonar/epidemiología , Proteinosis Alveolar Pulmonar/genética , Proteinosis Alveolar Pulmonar/inmunología , Intercambio Gaseoso Pulmonar , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Rituximab/uso terapéutico , Irrigación Terapéutica
12.
Neurobiol Dis ; 74: 219-28, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25497733

RESUMEN

Mutations in the colony stimulating factor-1 receptor (CSF1R) that abrogate the expression of the affected allele or lead to the expression of mutant receptor chains devoid of kinase activity have been identified in both familial and sporadic cases of ALSP. To determine the validity of the Csf1r heterozygous mouse as a model of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) we performed behavioral, radiologic, histopathologic, ultrastructural and cytokine expression studies of young and old Csf1r+/- and control Csf1r+/+ mice. Six to 8-month old Csf1r+/- mice exhibit cognitive deficits, and by 9-11 months develop sensorimotor deficits and in male mice, depression and anxiety-like behavior. MRIs of one year-old Csf1r+/- mice reveal lateral ventricle enlargement and thinning of the corpus callosum. Ultrastructural analysis of the corpus callosum uncovers dysmyelinated axons as well as neurodegeneration, evidenced by the presence of axonal spheroids. Histopathological examination of 11-week-old mice reveals increased axonal and myelin staining in the cortex, increase of neuronal cell density in layer V and increase of microglial cell densities throughout the brain, suggesting that early developmental changes contribute to disease. By 10-months of age, the neuronal cell density normalizes, oligodendrocyte precursor cells increase in layers II-III and V and microglial densities remain elevated without an increase in astrocytes. Also, the age-dependent increase in CSF-1R+ neurons in cortical layer V is reduced. Moreover, the expression of Csf2, Csf3, Il27 and Il6 family cytokines is increased, consistent with microglia-mediated inflammation. These results demonstrate that the inactivation of one Csf1r allele is sufficient to cause an ALSP-like disease in mice. The Csf1r+/- mouse is a model of ALSP that will allow the critical events for disease development to be determined and permit rapid evaluation of therapeutic approaches. Furthermore, our results suggest that aberrant activation of microglia in Csf1r+/- mice may play a central role in ALSP pathology.


Asunto(s)
Modelos Animales de Enfermedad , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Animales , Ansiedad/patología , Ansiedad/fisiopatología , Encéfalo/inmunología , Encéfalo/patología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Citocinas/metabolismo , Depresión/patología , Depresión/fisiopatología , Progresión de la Enfermedad , Femenino , Leucoencefalopatías/patología , Leucoencefalopatías/fisiopatología , Leucoencefalopatías/psicología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Células-Madre Neurales/inmunología , Células-Madre Neurales/patología , Neuroglía/inmunología , Neuroglía/patología , Neuronas/inmunología , Neuronas/patología , Percepción Olfatoria/fisiología , Fenotipo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Sustancia Blanca/inmunología , Sustancia Blanca/patología
14.
Am J Respir Crit Care Med ; 189(2): 167-82, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24279725

RESUMEN

RATIONALE: Hereditary pulmonary alveolar proteinosis (hPAP) caused by granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor α-chain (CSF2RA) deficiency is a rare, life-threatening lung disease characterized by accumulation of proteins and phospholipids in the alveolar spaces. The disease is caused by a functional insufficiency of alveolar macrophages, which require GM-CSF signaling for terminal differentiation and effective degradation of alveolar proteins and phospholipids. Therapeutic options are extremely limited, and the pathophysiology underlying the defective protein degradation in hPAP alveolar macrophages remains poorly understood. OBJECTIVES: To further elucidate the cellular mechanisms underlying hPAP and evaluate novel therapeutic strategies, we here investigated the potential of hPAP patient-derived induced pluripotent stem cell (PAP-iPSCs) derived monocytes and macrophages. METHODS: Patient-specific PAP-iPSCs were generated from CD34(+) bone marrow cells of a CSF2RA-deficient patient with PAP. We assessed pluripotency, chromosomal integrity, and genetic correction of established iPSC lines. On hematopoietic differentiation, genetically corrected or noncorrected monocytes and macrophages were investigated in GM-CSF-dependent assays. MEASUREMENTS AND MAIN RESULTS: Although monocytes and macrophages differentiated from noncorrected PAP-iPSCs exhibited distinct defects in GM-CSF-dependent functions, such as perturbed CD11b activation, phagocytic activity, and STAT5 phosphorylation after GM-CSF exposure and lack of GM-CSF uptake, these defects were fully repaired on lentiviral gene transfer of a codon-optimized CSF2RA-cDNA. CONCLUSIONS: These data establish PAP-iPSC-derived monocytes and macrophages as a valid in vitro disease model of CSF2RA-deficient PAP, and introduce gene-corrected iPSC-derived monocytes and macrophages as a potential autologous cell source for innovative therapeutic strategies. Transplantation of such cells to patients with hPAP could serve as a paradigmatic proof for the potential of iPSC-derived cells in clinical gene therapy.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X/terapia , Terapia Genética , Células Madre Pluripotentes Inducidas , Proteinosis Alveolar Pulmonar/terapia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Preescolar , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Humanos , Macrófagos Alveolares/metabolismo , Modelos Biológicos , Monocitos/metabolismo , Proteinosis Alveolar Pulmonar/genética , Proteinosis Alveolar Pulmonar/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
15.
Am J Respir Crit Care Med ; 189(2): 183-93, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24279752

RESUMEN

RATIONALE: In patients with pulmonary alveolar proteinosis (PAP) syndrome, disruption of granulocyte/macrophage colony-stimulating factor (GM-CSF) signaling is associated with pathogenic surfactant accumulation from impaired clearance in alveolar macrophages. OBJECTIVES: The aim of this study was to overcome these barriers by using monocyte-derived induced pluripotent stem (iPS) cells to recapitulate disease-specific and normal macrophages. METHODS: We created iPS cells from two children with hereditary PAP (hPAP) caused by recessive CSF2RA(R217X) mutations and three normal people, differentiated them into macrophages (hPAP-iPS-Mφs and NL-iPS-Mφs, respectively), and evaluated macrophage functions with and without gene-correction to restore GM-CSF signaling in hPAP-iPS-Mφs. MEASUREMENTS AND MAIN RESULTS: Both hPAP and normal iPS cells had human embryonic stem cell-like morphology, expressed pluripotency markers, formed teratomas in vivo, had a normal karyotype, retained and expressed mutant or normal CSF2RA genes, respectively, and could be differentiated into macrophages with the typical morphology and phenotypic markers. Compared with normal, hPAP-iPS-Mφs had impaired GM-CSF receptor signaling and reduced GM-CSF-dependent gene expression, GM-CSF- but not M-CSF-dependent cell proliferation, surfactant clearance, and proinflammatory cytokine secretion. Restoration of GM-CSF receptor signaling corrected the surfactant clearance abnormality in hPAP-iPS-Mφs. CONCLUSIONS: We used patient-specific iPS cells to accurately reproduce the molecular and cellular defects of alveolar macrophages that drive the pathogenesis of PAP in more than 90% of patients. These results demonstrate the critical role of GM-CSF signaling in surfactant homeostasis and PAP pathogenesis in humans and have therapeutic implications for hPAP.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Células Madre Pluripotentes Inducidas/metabolismo , Proteinosis Alveolar Pulmonar/fisiopatología , Surfactantes Pulmonares/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Niño , Técnicas de Transferencia de Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Humanos , Macrófagos Alveolares/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Transducción de Señal
16.
J Immunol ; 184(9): 4625-9, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20357255

RESUMEN

During priming, CD8(+) T lymphocytes can induce robust maturation of dendritic cells (DCs) in a CD40-independent manner by secreting licensing factor(s). In this study, we isolate this so-far elusive licensing factor and identify it, surprisingly, as GM-CSF. This provides a new face for an old factor with a well-known supporting role in DC development and recruitment. Signaling through the GM-CSFR in ex vivo-purified DCs upregulated the expression of costimulatory molecules more efficiently than did any tested TLR agonist and provided a positive feedback loop in the stimulation of CD8(+) T cell proliferation. Combined with a variety of microbial stimuli, GM-CSF supports the formation of potent "effector" DCs capable of secreting a variety of proinflammatory cytokines that guide the differentiation of T cells during the immune response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Animales , Comunicación Celular/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Citocinas/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factores de Transcripción TCF/metabolismo , Factores de Transcripción TCF/fisiología , Regulación hacia Arriba/inmunología
17.
Leuk Res ; 32(2): 235-42, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17686517

RESUMEN

A girl with congenital dysgranulopoietic neutropenia (CDN) and her non-neutropenic mother with aphthae (A) were investigated. Apoptosis in lymphocytes and granulocytes of both patients (mother A+) were documented by high annexin and electron microscopic morphology. CD11b/CD18 of the daughter's granulocytes ranged between low to normal while that of the mother changed between very low to high levels through A(-) to A(+) periods. In both patients, CD11b/CD18 on lymphocytes were high; GM-CSF receptor was negative; CD4-/CD8- lymphocytes were high and the leukocytes which showed abnormal cell cycle were stained by senescence associated beta-galactosidase. We think that increased apoptosis and rapid cell senescence of leukocytes underlies the pathophysiology of CDN.


Asunto(s)
Apoptosis/fisiología , Senescencia Celular/fisiología , Granulocitos/ultraestructura , Linfocitos/ultraestructura , Neutropenia/fisiopatología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Adolescente , Anexina A5/metabolismo , Antígenos CD/metabolismo , Ciclo Celular , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Subgrupos Linfocitarios/ultraestructura , Microscopía Electrónica de Transmisión , Neutropenia/congénito , Neutropenia/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Estomatitis Aftosa/metabolismo , Estomatitis Aftosa/patología , Estomatitis Aftosa/fisiopatología , Receptor fas/metabolismo
18.
J Neuroimmunol ; 140(1-2): 88-96, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12864975

RESUMEN

The hematopoietic factor and inflammatory cytokine GM-CSF is involved in PNS and CNS injury and disease, and in macrophage and microglia function regulation. We presently document that injury to PNS axons induces in vivo production of GM-CSF-inhibitor and GM-CSF-augmenter activities. GM-CSF-inhibitor activity was detected in extract and conditioned medium (CM) of injured PNS but not in extract of intact PNS, and was removed from CM by GM-CSF affinity chromatography, suggesting it is carried by a secreted GM-CSF binding molecule. CM further displayed GM-CSF-augmenter activity along with GM-CSF-inhibitor activity but at contrasting concentrations; augmentation at lowest and inhibition at highest. GM-CSF activity is thus regulated during Wallerian degeneration (WD); augmenter activity characterizes the onset and inhibitor activity the later stages of WD.


Asunto(s)
Axones/patología , Proteínas Portadoras/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Superficie Celular/fisiología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Nervio Ciático/patología , Degeneración Walleriana/metabolismo , Animales , Axones/metabolismo , Axotomía , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/análisis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidades de Proteína/análisis , Subunidades de Proteína/genética , ARN Mensajero/análisis , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/análisis , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Interleucina-3/análisis , Receptores de Interleucina-3/metabolismo , Receptores de Interleucina-3/fisiología , Nervio Ciático/metabolismo , Solubilidad , Factores de Tiempo , Regulación hacia Arriba/fisiología , Degeneración Walleriana/genética , Degeneración Walleriana/patología
19.
J Immunol ; 166(8): 4915-21, 2001 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11290769

RESUMEN

IL-12, like IL-18, was shown to potently inhibit osteoclast formation in cultures of cocultures of murine osteoblast and spleen cells, as well as in adult spleen cells treated with M-CSF and receptor activator of NF-kappaB ligand (RANKL). Neither IL-12 nor IL-18 was able to inhibit RANKL-induced osteoclast formation in cultured RAW264.7 cells, demonstrating that IL-12, like IL-18, was unable to act directly on osteoclastic precursors. IL-12, like IL-18, was found to act by T cells, since depletion of T cells from the adult spleen cell cultures ablated the inhibitory action of IL-12 and addition of either CD4 or CD8 T cells from C57BL/6 mice to RANKL-stimulated RAW264.7 cultures permitted IL-12 or IL-18 to be inhibitory. Additionally, IL-12 was still able to inhibit osteoclast formation in cocultures with osteoblasts and spleen cells from either GM-CSF R(-/-) mice or IFN-gamma R(-/-) mice, indicating that neither GM-CSF nor IFN-gamma was mediating osteoclast inhibition in these cultures. Combined, IL-18 and IL-12 synergistically inhibited osteoclast formation at concentrations 20- to 1000-fold less, respectively, than when added individually. A candidate inhibitor could not be demonstrated using neutralizing Abs to IL-4, IL-10, or IL-13 or from mRNA expression profiles among known cytokine inhibitors of osteoclastogenesis in response to IL-12 and IL-18 treatment, although the unknown inhibitory molecule was determined to be secreted from T cells.


Asunto(s)
Inhibidores de Crecimiento/fisiología , Interleucina-12/fisiología , Interleucina-18/fisiología , Osteoclastos/citología , Osteoclastos/inmunología , Animales , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/inmunología , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Regulación hacia Abajo/inmunología , Combinación de Medicamentos , Sinergismo Farmacológico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Inhibidores de Crecimiento/metabolismo , Interferón gamma/metabolismo , Interferón gamma/fisiología , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...