Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nucleic Acids Res ; 52(D1): D590-D596, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37889041

RESUMEN

CRISPR-Cas enzymes enable RNA-guided bacterial immunity and are widely used for biotechnological applications including genome editing. In particular, the Class 2 CRISPR-associated enzymes (Cas9, Cas12 and Cas13 families), have been deployed for numerous research, clinical and agricultural applications. However, the immense genetic and biochemical diversity of these proteins in the public domain poses a barrier for researchers seeking to leverage their activities. We present CasPEDIA (http://caspedia.org), the Cas Protein Effector Database of Information and Assessment, a curated encyclopedia that integrates enzymatic classification for hundreds of different Cas enzymes across 27 phylogenetic groups spanning the Cas9, Cas12 and Cas13 families, as well as evolutionarily related IscB and TnpB proteins. All enzymes in CasPEDIA were annotated with a standard workflow based on their primary nuclease activity, target requirements and guide-RNA design constraints. Our functional classification scheme, CasID, is described alongside current phylogenetic classification, allowing users to search related orthologs by enzymatic function and sequence similarity. CasPEDIA is a comprehensive data portal that summarizes and contextualizes enzymatic properties of widely used Cas enzymes, equipping users with valuable resources to foster biotechnological development. CasPEDIA complements phylogenetic Cas nomenclature and enables researchers to leverage the multi-faceted nucleic-acid targeting rules of diverse Class 2 Cas enzymes.


Asunto(s)
Proteínas Asociadas a CRISPR , Sistemas CRISPR-Cas , Bases de Datos Genéticas , Endodesoxirribonucleasas , Sistemas CRISPR-Cas/genética , Filogenia , Proteínas Asociadas a CRISPR/química , Proteínas Asociadas a CRISPR/clasificación , Proteínas Asociadas a CRISPR/genética , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/clasificación , Endodesoxirribonucleasas/genética , Enciclopedias como Asunto
2.
J Immunol ; 204(6): 1474-1485, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31996456

RESUMEN

An increasing body of evidence suggests that bone marrow-derived myeloid cells play a critical role in the pathophysiology of pulmonary hypertension (PH). However, the true requirement for myeloid cells in PH development has not been demonstrated, and a specific disease-promoting myeloid cell population has not been identified. Using bone marrow chimeras, lineage labeling, and proliferation studies, we determined that, in murine hypoxia-induced PH, Ly6Clo nonclassical monocytes are recruited to small pulmonary arteries and differentiate into pulmonary interstitial macrophages. Accumulation of these nonclassical monocyte-derived pulmonary interstitial macrophages around pulmonary vasculature is associated with increased muscularization of small pulmonary arteries and disease severity. To determine if the sensing of hypoxia by nonclassical monocytes contributes to the development of PH, mice lacking expression of hypoxia-inducible factor-1α in the Ly6Clo monocyte lineage were exposed to hypoxia. In these mice, vascular remodeling and PH severity were significantly reduced. Transcriptome analyses suggest that the Ly6Clo monocyte lineage regulates PH through complement, phagocytosis, Ag presentation, and chemokine/cytokine pathways. Consistent with these murine findings, relative to controls, lungs from pulmonary arterial hypertension patients displayed a significant increase in the frequency of nonclassical monocytes. Taken together, these findings show that, in response to hypoxia, nonclassical monocytes in the lung sense hypoxia, infiltrate small pulmonary arteries, and promote vascular remodeling and development of PH. Our results demonstrate that myeloid cells, specifically cells of the nonclassical monocyte lineage, play a direct role in the pathogenesis of PH.


Asunto(s)
Hipertensión Pulmonar/inmunología , Hipoxia/complicaciones , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Remodelación Vascular/inmunología , Animales , Antígenos Ly/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/cirugía , Hipoxia/inmunología , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/patología , Trasplante de Pulmón , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Quimera por Trasplante/inmunología , Remodelación Vascular/genética
3.
Genome Res ; 28(4): 423-431, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29567674

RESUMEN

Over a decade ago, the Atacama humanoid skeleton (Ata) was discovered in the Atacama region of Chile. The Ata specimen carried a strange phenotype-6-in stature, fewer than expected ribs, elongated cranium, and accelerated bone age-leading to speculation that this was a preserved nonhuman primate, human fetus harboring genetic mutations, or even an extraterrestrial. We previously reported that it was human by DNA analysis with an estimated bone age of about 6-8 yr at the time of demise. To determine the possible genetic drivers of the observed morphology, DNA from the specimen was subjected to whole-genome sequencing using the Illumina HiSeq platform with an average 11.5× coverage of 101-bp, paired-end reads. In total, 3,356,569 single nucleotide variations (SNVs) were found as compared to the human reference genome, 518,365 insertions and deletions (indels), and 1047 structural variations (SVs) were detected. Here, we present the detailed whole-genome analysis showing that Ata is a female of human origin, likely of Chilean descent, and its genome harbors mutations in genes (COL1A1, COL2A1, KMT2D, FLNB, ATR, TRIP11, PCNT) previously linked with diseases of small stature, rib anomalies, cranial malformations, premature joint fusion, and osteochondrodysplasia (also known as skeletal dysplasia). Together, these findings provide a molecular characterization of Ata's peculiar phenotype, which likely results from multiple known and novel putative gene mutations affecting bone development and ossification.


Asunto(s)
ADN Antiguo/análisis , Genoma Humano/genética , Osteocondrodisplasias/genética , Secuenciación Completa del Genoma , Animales , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación INDEL , Anotación de Secuencia Molecular , Mutación/genética , Osteocondrodisplasias/fisiopatología , Fenotipo , Polimorfismo de Nucleótido Simple/genética
4.
Brain Behav Immun ; 76: 104-115, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30447281

RESUMEN

Drugs of abuse promote a potent immune response in central nervous system (CNS) via the activation of microglia and astrocytes. However, the molecular mechanisms underlying microglial activation during addiction are not well known. We developed and functionally characterized a novel transgenic mouse (Cx3cr1-CreBTtg/0:MyD88f/f [Cretg/0]) wherein the immune signaling adaptor gene, MyD88, was specifically deleted in microglia. To test the downstream effects of loss of microglia-specific MyD88 signaling in morphine addiction, Cretg/0 and Cre0/0 mice were tested for reward learning, extinction, and reinstatement using a conditioned place preference (CPP) paradigm. There were no differences in drug acquisition, but Cretg/0 mice had prolonged extinction and enhanced reinstatement compared to Cre0/0 controls. Furthermore, morphine-treated Cretg/0 mice showed increased doublecortin (DCX) signal relative to Cre0/0 control mice in the hippocampus, indicative of increased number of immature neurons. Additionally, there was an increase in colocalization of microglial lysosomal marker CD68 with DCX+cells in morphine-treated Cretg/0 mice but not in Cre0/0 or drug-naїve mice, suggesting a specific role for microglial MyD88 signaling in neuronal phagocytosis in the hippocampus. Our results show that MyD88 deletion in microglia may negatively impact maturing neurons within the adult hippocampus and thus reward memories, suggesting a novel protective role for microglia in opioid addiction.


Asunto(s)
Conducta Adictiva/metabolismo , Giro Dentado/metabolismo , Microglía/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Morfina/farmacología , Factor 88 de Diferenciación Mieloide/deficiencia , Neuropéptidos/metabolismo , Animales , Conducta Adictiva/genética , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Narcóticos/farmacología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Trastornos Relacionados con Opioides/genética , Trastornos Relacionados con Opioides/metabolismo , Recompensa
5.
Am J Pathol ; 186(11): 2846-2856, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27640148

RESUMEN

Inappropriate activation of the renin angiotensin system (RAS) is a key contributor to the pathogenesis of essential hypertension. During RAS activation, infiltration of immune cells into the kidney exacerbates hypertension and renal injury. However, the mechanisms underpinning the accumulation of mononuclear cells in the kidney after RAS stimulation remain unclear. C-C motif chemokine 5 (CCL5) drives recruitment of macrophages and T lymphocytes into injured tissues, and we have found that RAS activation induces CCL5 expression in the kidney during the pathogenesis of hypertension and renal fibrosis. We therefore evaluated the contribution of CCL5 to renal damage and fibrosis in hypertensive and normotensive models of RAS stimulation. Surprisingly, during angiotensin II-induced hypertension, CCL5-deficient (knockout, KO) mice exhibited markedly augmented kidney damage, macrophage infiltration, and expression of proinflammatory macrophage cytokines compared with wild-type controls. When subjected to the normotensive unilateral ureteral obstruction model of endogenous RAS activation, CCL5 KO mice similarly developed more severe renal fibrosis and greater accumulation of macrophages in the kidney, congruent with enhanced renal expression of the macrophage chemokine CCL2. In turn, pharmacologic inhibition of CCL2 abrogated the differences between CCL5 KO and wild-type mice in kidney fibrosis and macrophage infiltration after unilateral ureteral obstruction. These data indicate that CCL5 paradoxically limits macrophage accumulation in the injured kidney during RAS activation by constraining the proinflammatory actions of CCL2.


Asunto(s)
Angiotensina II/inmunología , Quimiocina CCL5/metabolismo , Hipertensión/inmunología , Enfermedades Renales/inmunología , Riñón/patología , Animales , Presión Sanguínea , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL5/genética , Hipertensión Esencial , Femenino , Fibrosis , Hipertensión/etiología , Riñón/inmunología , Riñón/cirugía , Enfermedades Renales/etiología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Nefrectomía , Sistema Renina-Angiotensina/inmunología , Linfocitos T/inmunología , Obstrucción Ureteral
7.
J Neurosci ; 35(15): 5969-82, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878270

RESUMEN

The pathogenesis of Alzheimer's disease (AD) is a critical unsolved question; and although recent studies have demonstrated a strong association between altered brain immune responses and disease progression, the mechanistic cause of neuronal dysfunction and death is unknown. We have previously described the unique CVN-AD mouse model of AD, in which immune-mediated nitric oxide is lowered to mimic human levels, resulting in a mouse model that demonstrates the cardinal features of AD, including amyloid deposition, hyperphosphorylated and aggregated tau, behavioral changes, and age-dependent hippocampal neuronal loss. Using this mouse model, we studied longitudinal changes in brain immunity in relation to neuronal loss and, contrary to the predominant view that AD pathology is driven by proinflammatory factors, we find that the pathology in CVN-AD mice is driven by local immune suppression. Areas of hippocampal neuronal death are associated with the presence of immunosuppressive CD11c(+) microglia and extracellular arginase, resulting in arginine catabolism and reduced levels of total brain arginine. Pharmacologic disruption of the arginine utilization pathway by an inhibitor of arginase and ornithine decarboxylase protected the mice from AD-like pathology and significantly decreased CD11c expression. Our findings strongly implicate local immune-mediated amino acid catabolism as a novel and potentially critical mechanism mediating the age-dependent and regional loss of neurons in humans with AD.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Arginina/metabolismo , Encéfalo/metabolismo , Factores Inmunológicos/metabolismo , Factores de Edad , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Antígenos CD/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Eflornitina/farmacología , Eflornitina/uso terapéutico , Humanos , Factores Inmunológicos/genética , Aprendizaje por Laberinto/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Microglía/metabolismo , Mutación/genética , Óxido Nítrico Sintasa de Tipo II/genética , Inhibidores de la Ornitina Descarboxilasa/farmacología , Inhibidores de la Ornitina Descarboxilasa/uso terapéutico
8.
PLoS Genet ; 9(10): e1003807, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130503

RESUMEN

During ischemic stroke, occlusion of the cerebrovasculature causes neuronal cell death (infarction), but naturally occurring genetic factors modulating infarction have been difficult to identify in human populations. In a surgically induced mouse model of ischemic stroke, we have previously mapped Civq1 to distal chromosome 7 as a quantitative trait locus determining infarct volume. In this study, genome-wide association mapping using 32 inbred mouse strains and an additional linkage scan for infarct volume confirmed that the size of the infarct is determined by ancestral alleles of the causative gene(s). The genetically isolated Civq1 locus in reciprocal recombinant congenic mice refined the critical interval and demonstrated that infarct size is determined by both vascular (collateral vessel anatomy) and non-vascular (neuroprotection) effects. Through the use of interval-specific SNP haplotype analysis, we further refined the Civq1 locus and identified integrin alpha L (Itgal) as one of the causative genes for Civq1. Itgal is the only gene that exhibits both strain-specific amino acid substitutions and expression differences. Coding SNPs, a 5-bp insertion in exon 30b, and increased mRNA and protein expression of a splice variant of the gene (Itgal-003, ENSMUST00000120857), all segregate with infarct volume. Mice lacking Itgal show increased neuronal cell death in both ex vivo brain slice and in vivo focal cerebral ischemia. Our data demonstrate that sequence variation in Itgal modulates ischemic brain injury, and that infarct volume is determined by both vascular and non-vascular mechanisms.


Asunto(s)
Estudio de Asociación del Genoma Completo , Cadenas alfa de Integrinas/genética , Accidente Cerebrovascular/genética , Alelos , Animales , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatología , Modelos Animales de Enfermedad , Ligamiento Genético , Haplotipos , Humanos , Cadenas alfa de Integrinas/metabolismo , Ratones , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Accidente Cerebrovascular/fisiopatología
9.
J Immunol ; 191(9): 4665-75, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24078688

RESUMEN

Macrophages and dendritic cells (DC) are distributed throughout the body and play important roles in pathogen detection and tissue homeostasis. In tissues, resident macrophages exhibit distinct phenotypes and activities, yet the transcriptional pathways that specify tissue-specific macrophages are largely unknown. We investigated the functions and origins of two peritoneal macrophage populations in mice: small and large peritoneal macrophages (SPM and LPM, respectively). SPM and LPM differ in their ability to phagocytose apoptotic cells, as well as in the production of cytokines in response to LPS. In steady-state conditions, SPM are sustained by circulating precursors, whereas LPM are maintained independently of hematopoiesis; however, both populations are replenished by bone marrow precursors following radiation injury. Transcription factor analysis revealed that SPM and LPM express abundant CCAAT/enhancer binding protein (C/EBP)-ß. Cebpb(-/-) mice exhibit elevated numbers of SPM-like cells but lack functional LPM. Alveolar macrophages are also missing in Cebpb(-/-) mice, although macrophage populations in the spleen, kidney, skin, mesenteric lymph nodes, and liver are normal. Adoptive transfer of SPM into Cebpb(-/-) mice results in SPM differentiation into LPM, yet donor SPM do not generate LPM after transfer into C/EBPß-sufficient mice, suggesting that endogenous LPM inhibit differentiation by SPM. We conclude that C/EBPß plays an intrinsic, tissue-restricted role in the generation of resident macrophages.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Peritoneales/metabolismo , Traslado Adoptivo , Animales , Apoptosis , Proteína beta Potenciadora de Unión a CCAAT/genética , Diferenciación Celular , Células Dendríticas/inmunología , Riñón/citología , Hígado/citología , Ganglios Linfáticos/citología , Macrófagos Alveolares/citología , Macrófagos Alveolares/inmunología , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/inmunología , Piel/citología , Bazo/citología
10.
J Pediatric Infect Dis Soc ; 10(1): 49-51, 2021 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32318729

RESUMEN

A 5-week-old infant female admitted for fever without a source subsequently tested positive for severe acute respiratory syndrome coronavirus 2. She had a mild hospital course without respiratory distress. This unexpected presentation changed regional hospital screening for coronavirus disease 2019 and personal protective equipment use by medical providers who evaluate febrile infants.


Asunto(s)
COVID-19/diagnóstico , Fiebre de Origen Desconocido/virología , Prueba de Ácido Nucleico para COVID-19 , California , Diagnóstico Diferencial , Femenino , Humanos , Lactante , Transmisión de Enfermedad Infecciosa de Paciente a Profesional/prevención & control , Equipo de Protección Personal , SARS-CoV-2
11.
Sci Data ; 6(1): 201, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615985

RESUMEN

The identification of novel disease associations using big-data for patient care has had limited success. In this study, we created a longitudinal disease network of traced readmissions (disease trajectories), merging data from over 10.4 million inpatients through the Healthcare Cost and Utilization Project, which allowed the representation of disease progression mapping over 300 diseases. From these disease trajectories, we discovered an interesting association between schizophrenia and rhabdomyolysis, a rare muscle disease (incidence < 1E-04) (relative risk, 2.21 [1.80-2.71, confidence interval = 0.95], P-value 9.54E-15). We validated this association by using independent electronic medical records from over 830,000 patients at the University of California, San Francisco (UCSF) medical center. A case review of 29 rhabdomyolysis incidents in schizophrenia patients at UCSF demonstrated that 62% are idiopathic, without the use of any drug known to lead to this adverse event, suggesting a warning to physicians to watch for this unexpected risk of schizophrenia. Large-scale analysis of disease trajectories can help physicians understand potential sequential events in their patients.


Asunto(s)
Rabdomiólisis/complicaciones , Rabdomiólisis/diagnóstico , Esquizofrenia/complicaciones , Esquizofrenia/diagnóstico , Registros Electrónicos de Salud , Humanos , Riesgo , San Francisco
12.
Cell Rep ; 25(2): 513-522.e3, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30304689

RESUMEN

There is increasing appreciation that the immune system plays critical roles not only in the traditional domains of infection and inflammation but also in many areas of biology, including tumorigenesis, metabolism, and even neurobiology. However, one of the major barriers for understanding human immunological mechanisms is that immune assays have not been reproducibly characterized for a sufficiently large and diverse healthy human cohort. Here, we present the 10,000 Immunomes Project (10KIP), a framework for growing a diverse human immunology reference, from ImmPort, a publicly available resource of subject-level immunology data. Although some measurement types are sparse in the presently deposited ImmPort database, the extant data allow for a diversity of robust comparisons. Using 10KIP, we describe variations in serum cytokines and leukocytes by age, race, and sex; define a baseline cell-cytokine network; and describe immunologic changes in pregnancy. All data in the resource are available for visualization and download at http://10kimmunomes.org/.


Asunto(s)
Biomarcadores/análisis , Biología Computacional/métodos , Citocinas/metabolismo , Bases de Datos Factuales , Redes Reguladoras de Genes/inmunología , Sistema Inmunológico/inmunología , Leucocitos/metabolismo , Adolescente , Adulto , Estudios de Cohortes , Citocinas/inmunología , Femenino , Humanos , Leucocitos/inmunología , Masculino , Embarazo , Adulto Joven
13.
J Control Release ; 269: 364-373, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29146246

RESUMEN

Strategies that enhance the host antitumor immune response promise to revolutionize cancer therapy. Optimally mobilizing the immune system will likely require a multi-pronged approach to overcome the resistance developed by tumors to therapy. Recently, it has become recognized that doxorubicin can contribute to re-establishing host antitumor immunity through the generation of immunogenic cell death. However, the potential for delivery strategies to further enhance the immunological effects of doxorubicin has not been adequately examined. We report herein that Chimeric Polypeptide Doxorubicin (CP-Dox), a nanoparticle formulation of doxorubicin, enhances antitumor immunity. Compared to free doxorubicin, a single intravenous (IV) administration of CP-Dox at the maximum tolerated dose increases the infiltration of leukocytes into the tumor, slowing tumor growth and preventing metastasis in poorly immunogenic 4T1 mammary carcinoma. We demonstrate that the full efficacy of CP-Dox is dependent on CD8+ T cells and IFN-γ. CP-dox treatment also repolarized intratumoral myeloid cells towards an antitumor phenotype. These findings demonstrate that a nanoparticle drug is distinct from the free drug in its ability to productively stimulate antitumor immunity. Our study strongly argues for the use of antitumor immunotherapies combined with nanoparticle-packaged chemotherapy.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Neoplasias Mamarias Experimentales/inmunología , Nanopartículas/administración & dosificación , Péptidos/administración & dosificación , Animales , Antibióticos Antineoplásicos/química , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Doxorrubicina/química , Composición de Medicamentos , Femenino , Interferón gamma/inmunología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones Endogámicos BALB C , Nanopartículas/química , Péptidos/química
14.
PLoS One ; 11(3): e0150606, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26938654

RESUMEN

Flow cytometry is used extensively to examine immune cells in non-lymphoid tissues. However, a method of flow cytometric analysis that is both comprehensive and widely applicable has not been described. We developed a protocol for the flow cytometric analysis of non-lymphoid tissues, including methods of tissue preparation, a 10-fluorochrome panel for cell staining, and a standardized gating strategy, that allows the simultaneous identification and quantification of all major immune cell types in a variety of normal and inflamed non-lymphoid tissues. We demonstrate that our basic protocol minimizes cell loss, reliably distinguishes macrophages from dendritic cells (DC), and identifies all major granulocytic and mononuclear phagocytic cell types. This protocol is able to accurately quantify 11 distinct immune cell types, including T cells, B cells, NK cells, neutrophils, eosinophils, inflammatory monocytes, resident monocytes, alveolar macrophages, resident/interstitial macrophages, CD11b- DC, and CD11b+ DC, in normal lung, heart, liver, kidney, intestine, skin, eyes, and mammary gland. We also characterized the expression patterns of several commonly used myeloid and macrophage markers. This basic protocol can be expanded to identify additional cell types such as mast cells, basophils, and plasmacytoid DC, or perform detailed phenotyping of specific cell types. In examining models of primary and metastatic mammary tumors, this protocol allowed the identification of several distinct tumor associated macrophage phenotypes, the appearance of which was highly specific to individual tumor cell lines. This protocol provides a valuable tool to examine immune cell repertoires and follow immune responses in a wide variety of tissues and experimental conditions.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Inflamación/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Basófilos/inmunología , Basófilos/patología , Células Dendríticas/inmunología , Células Dendríticas/patología , Eosinófilos/inmunología , Eosinófilos/patología , Inflamación/patología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Macrófagos/inmunología , Macrófagos/patología , Mastocitos/inmunología , Mastocitos/patología , Ratones , Neutrófilos/inmunología , Neutrófilos/patología , Linfocitos T/inmunología , Linfocitos T/patología
15.
Cell Rep ; 12(7): 1107-19, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26257171

RESUMEN

The testis produces sperm throughout the male reproductive lifespan by balancing self-renewal and differentiation of spermatogonial stem cells (SSCs). Part of the SSC niche is thought to lie outside the seminiferous tubules of the testis; however, specific interstitial components of the niche that regulate spermatogonial divisions and differentiation remain undefined. We identified distinct populations of testicular macrophages, one of which lies on the surface of seminiferous tubules, in close apposition to areas of tubules enriched for undifferentiated spermatogonia. These macrophages express spermatogonial proliferation- and differentiation-inducing factors, such as colony-stimulating factor 1 (CSF1) and enzymes involved in retinoic acid (RA) biosynthesis. We show that transient depletion of macrophages leads to a disruption in spermatogonial differentiation. These findings reveal an unexpected role for macrophages in the spermatogonial niche in the testis and raise the possibility that macrophages play previously unappreciated roles in stem/progenitor cell regulation in other tissues.


Asunto(s)
Macrófagos/metabolismo , Espermatogénesis , Espermatogonias/citología , Nicho de Células Madre , Testículo/citología , Animales , Factores Estimulantes de Colonias/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Espermatogonias/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Tretinoina/metabolismo
17.
J Immunol ; 179(7): 4661-71, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17878364

RESUMEN

iNKT cells are required for the induction of airway hyperreactivity (AHR), a cardinal feature of asthma, but how iNKT cells traffic to the lungs to induce AHR has not been previously studied. Using several models of asthma, we demonstrated that iNKT cells required the chemokine receptor CCR4 for pulmonary localization and for the induction of AHR. In both allergen-induced and glycolipid-induced models of AHR, wild-type but not CCR4-/- mice developed AHR. Furthermore, adoptive transfer of wild-type but not CCR4-/- iNKT cells reconstituted AHR in iNKT cell-deficient mice. Moreover, we specifically tracked CCR4-/- vs wild-type iNKT cells in CCR4-/-:wild-type mixed BM chimeric mice in the resting state, and when AHR was induced by protein allergen or glycolipid. Using this unique model, we showed that both iNKT cells and conventional T cells required CCR4 for competitive localization into the bronchoalveolar lavage/airways compartment. These results establish for the first time that the pulmonary localization of iNKT cells critical for the induction of AHR requires CCR4 expression by iNKT cells.


Asunto(s)
Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Receptores de Quimiocina/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/patología , Movimiento Celular , Quimiocina CCL17 , Quimiocinas CC/metabolismo , Citocinas/biosíntesis , Glucolípidos/metabolismo , Ratones , Ratones Noqueados , Ovalbúmina/farmacología , Receptores CCR4 , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Linfocitos T Reguladores/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA