Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(7): 1270-1282, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38877178

RESUMEN

The relative and synergistic contributions of genetics and environment to interindividual immune response variation remain unclear, despite implications in evolutionary biology and medicine. Here we quantify interactive effects of genotype and environment on immune traits by investigating C57BL/6, 129S1 and PWK/PhJ inbred mice, rewilded in an outdoor enclosure and infected with the parasite Trichuris muris. Whereas cellular composition was shaped by interactions between genotype and environment, cytokine response heterogeneity including IFNγ concentrations was primarily driven by genotype with consequence on worm burden. In addition, we show that other traits, such as expression of CD44, were explained mostly by genetics on T cells, whereas expression of CD44 on B cells was explained more by environment across all strains. Notably, genetic differences under laboratory conditions were decreased following rewilding. These results indicate that nonheritable influences interact with genetic factors to shape immune variation and parasite burden.


Asunto(s)
Interacción Gen-Ambiente , Ratones Endogámicos C57BL , Tricuriasis , Trichuris , Animales , Trichuris/inmunología , Tricuriasis/inmunología , Tricuriasis/parasitología , Ratones , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Linfocitos B/inmunología , Genotipo , Interferón gamma/metabolismo , Linfocitos T/inmunología , Femenino , Masculino
2.
Cell ; 180(1): 79-91.e16, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31866067

RESUMEN

Lymphoid cells that produce interleukin (IL)-17 cytokines protect barrier tissues from pathogenic microbes but are also prominent effectors of inflammation and autoimmune disease. T helper 17 (Th17) cells, defined by RORγt-dependent production of IL-17A and IL-17F, exert homeostatic functions in the gut upon microbiota-directed differentiation from naive CD4+ T cells. In the non-pathogenic setting, their cytokine production is regulated by serum amyloid A proteins (SAA1 and SAA2) secreted by adjacent intestinal epithelial cells. However, Th17 cell behaviors vary markedly according to their environment. Here, we show that SAAs additionally direct a pathogenic pro-inflammatory Th17 cell differentiation program, acting directly on T cells in collaboration with STAT3-activating cytokines. Using loss- and gain-of-function mouse models, we show that SAA1, SAA2, and SAA3 have distinct systemic and local functions in promoting Th17-mediated inflammatory diseases. These studies suggest that T cell signaling pathways modulated by the SAAs may be attractive targets for anti-inflammatory therapies.


Asunto(s)
Síndrome del Colon Irritable/metabolismo , Proteína Amiloide A Sérica/metabolismo , Células Th17/metabolismo , Adulto , Animales , Enfermedades Autoinmunes/metabolismo , Diferenciación Celular/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Interleucina-17/metabolismo , Síndrome del Colon Irritable/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Células TH1 , Células Th17/inmunología
4.
Nat Immunol ; 18(6): 642-653, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28436955

RESUMEN

It remains unclear whether activated inflammatory macrophages can adopt features of tissue-resident macrophages, or what mechanisms might mediate such a phenotypic conversion. Here we show that vitamin A is required for the phenotypic conversion of interleukin 4 (IL-4)-activated monocyte-derived F4/80intCD206+PD-L2+MHCII+ macrophages into macrophages with a tissue-resident F4/80hiCD206-PD-L2-MHCII-UCP1+ phenotype in the peritoneal cavity of mice and during the formation of liver granulomas in mice infected with Schistosoma mansoni. The phenotypic conversion of F4/80intCD206+ macrophages into F4/80hiCD206- macrophages was associated with almost complete remodeling of the chromatin landscape, as well as alteration of the transcriptional profiles. Vitamin A-deficient mice infected with S. mansoni had disrupted liver granuloma architecture and increased mortality, which indicates that failure to convert macrophages from the F4/80intCD206+ phenotype to F4/80hiCD206- may lead to dysregulated inflammation during helminth infection.


Asunto(s)
Granuloma/inmunología , Hígado/inmunología , Macrófagos/inmunología , Esquistosomiasis mansoni/inmunología , Deficiencia de Vitamina A/inmunología , Animales , Antígenos de Diferenciación/metabolismo , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-4/inmunología , Lectinas Tipo C/metabolismo , Hígado/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Cavidad Peritoneal/citología , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/metabolismo , Schistosoma mansoni , Esquistosomiasis mansoni/patología , Tretinoina/farmacología , Proteína Desacopladora 1/metabolismo , Vitaminas/farmacología
5.
Cell ; 158(4): 705-721, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126780

RESUMEN

Acquisition of the intestinal microbiota begins at birth, and a stable microbial community develops from a succession of key organisms. Disruption of the microbiota during maturation by low-dose antibiotic exposure can alter host metabolism and adiposity. We now show that low-dose penicillin (LDP), delivered from birth, induces metabolic alterations and affects ileal expression of genes involved in immunity. LDP that is limited to early life transiently perturbs the microbiota, which is sufficient to induce sustained effects on body composition, indicating that microbiota interactions in infancy may be critical determinants of long-term host metabolic effects. In addition, LDP enhances the effect of high-fat diet induced obesity. The growth promotion phenotype is transferrable to germ-free hosts by LDP-selected microbiota, showing that the altered microbiota, not antibiotics per se, play a causal role. These studies characterize important variables in early-life microbe-host metabolic interaction and identify several taxa consistently linked with metabolic alterations. PAPERCLIP:


Asunto(s)
Antibacterianos/administración & dosificación , Modelos Animales de Enfermedad , Intestinos/microbiología , Microbiota , Obesidad/microbiología , Penicilinas/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/metabolismo , Femenino , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Obesidad/metabolismo
6.
Immunity ; 49(1): 16-18, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30021142

RESUMEN

How type 2 immune responses are initiated is obscure. Nadjsombati et al. (2018), along with two other studies (Lei et al., 2018; Schneider et al., 2018), show that tuft cells can initiate type 2 responses by recognizing the metabolite succinate produced by intestinal parasites.


Asunto(s)
Diabetes Mellitus Tipo 2 , Parásitos , Animales , Intestinos , Ácido Succínico , Gusto
7.
Immunity ; 47(6): 1024-1036, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262347

RESUMEN

Type-2-cell-mediated immune responses play a critical role in mediating both host-resistance and disease-tolerance mechanisms during helminth infections. Recently, type 2 cell responses have emerged as major regulators of tissue repair and metabolic homeostasis even under steady-state conditions. In this review, we consider how studies of helminth infection have contributed toward our expanding cellular and molecular understanding of type-2-cell-mediated immunity, as well as new areas such as the microbiome. By studying how these successful parasites form chronic infections without overt pathology, we are gaining additional insights into allergic and inflammatory diseases, as well as normal physiology.


Asunto(s)
Helmintiasis/inmunología , Inmunidad Celular , Macrófagos/inmunología , Nematodos/inmunología , Células Th2/inmunología , Trematodos/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Células Epiteliales/inmunología , Células Epiteliales/parasitología , Regulación de la Expresión Génica/inmunología , Helmintiasis/genética , Helmintiasis/parasitología , Homeostasis/inmunología , Interacciones Huésped-Parásitos/inmunología , Humanos , Macrófagos/parasitología , Mastocitos/inmunología , Mastocitos/parasitología , Microbiota/inmunología , Células Th2/parasitología
8.
J Immunol ; 212(4): 632-644, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38180236

RESUMEN

Distinct subsets of T lymphocytes express CX3CR1 under inflammatory conditions, but little is known about CX3CR1+CD4+ T cells during type 2 inflammation in helminth infections. In this study, we used a fate-mapping mouse model to characterize CX3CR1+CD4+ T cells during both acute Nippostrongylus brasiliensis and chronic Schistosoma mansoni murine models of helminth infections, revealing CX3CR1+CD4+ T cells to be an activated tissue-homing subset with varying capacity for cytokine production. Tracking these cells over time revealed that maintenance of CX3CR1 itself along with a TH2 phenotype conferred a survival advantage in the inflamed tissue. Single-cell RNA sequencing analysis of fate-mapped CX3CR1+CD4+ T cells from both the peripheral tissue and the spleen revealed a considerable level of diversity and identified a distinct population of BCL6+TCF-1+PD1+CD4+ T cells in the spleen during helminth infections. Conditional deletion of BCL6 in CX3CR1+ cells resulted in fewer CX3CR1+CD4+ T cells during infection, indicating a role in sustaining CD4+ T cell responses to helminth infections. Overall, our studies revealed the behavior and heterogeneity of CX3CR1+CD4+ T cells during type 2 inflammation in helminth infections and identified BCL6 to be important in their maintenance.


Asunto(s)
Linfocitos T CD4-Positivos , Helmintiasis , Schistosoma mansoni , Animales , Ratones , Linfocitos T CD4-Positivos/metabolismo , Helmintiasis/inmunología , Inflamación/metabolismo , Schistosoma mansoni/fisiología
9.
J Immunol ; 211(5): 836-843, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37466391

RESUMEN

Our previous studies identified a population of stem cell-like proliferating myeloid cells within inflamed tissues that could serve as a reservoir for tissue macrophages to adopt different activation states depending on the microenvironment. By lineage-tracing cells derived from CX3CR1+ precursors in mice during infection and profiling by single-cell RNA sequencing, in this study, we identify a cluster of BIRC5+ myeloid cells that expanded in the liver during chronic infection with either the parasite Schistosoma mansoni or the bacterial pathogen Staphylococcus aureus. In the absence of tissue-damaging toxins, S. aureus infection does not elicit these BIRC5+ cells. Moreover, deletion of BIRC5 from CX3CR1-expressing cells results in improved survival during S. aureus infection. Hence the combination of single-cell RNA sequencing and genetic fate-mapping CX3CR1+ cells revealed a toxin-dependent pathogenic role for BIRC5 in myeloid cells during S. aureus infection.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Ratones , Animales , Células Mieloides/patología , Análisis de la Célula Individual , Infecciones Estafilocócicas/microbiología
10.
Proc Natl Acad Sci U S A ; 119(31): e2123017119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35881802

RESUMEN

Staphylococcus aureus is an opportunistic pathogen and chief among bloodstream-infecting bacteria. S. aureus produces an array of human-specific virulence factors that may contribute to immune suppression. Here, we defined the response of primary human phagocytes following infection with S. aureus using RNA-sequencing (RNA-Seq). We found that the overall transcriptional response to S. aureus was weak both in the number of genes and in the magnitude of response. Using an ex vivo bacteremia model with fresh human blood, we uncovered that infection with S. aureus resulted in the down-regulation of genes related to innate immune response and cytokine and chemokine signaling. This muted transcriptional response was conserved across diverse S. aureus clones but absent in blood exposed to heat-killed S. aureus or blood infected with the less virulent staphylococcal species Staphylococcus epidermidis. Notably, this signature was also present in patients with S. aureus bacteremia. We identified the master regulator S. aureus exoprotein expression (SaeRS) and the SaeRS-regulated pore-forming toxins as key mediators of the transcriptional suppression. The S. aureus-mediated suppression of chemokine and cytokine transcription was reflected by circulating protein levels in the plasma. Wild-type S. aureus elicited a soluble milieu that was restrictive in the recruitment of human neutrophils compared with strains lacking saeRS. Thus, S. aureus blunts the inflammatory response resulting in impaired neutrophil recruitment, which could promote the survival of the pathogen during invasive infection.


Asunto(s)
Interacciones Huésped-Patógeno , Neutrófilos , Infecciones Estafilocócicas , Staphylococcus aureus , Bacteriemia/inmunología , Bacteriemia/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citocinas/metabolismo , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Humanos , Neutrófilos/inmunología , Neutrófilos/microbiología , Proteínas Citotóxicas Formadoras de Poros/genética , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Staphylococcus epidermidis/patogenicidad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Immunity ; 41(2): 311-24, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25088769

RESUMEN

Nod2 has been extensively characterized as a bacterial sensor that induces an antimicrobial and inflammatory gene expression program. Therefore, it is unclear why Nod2 mutations that disrupt bacterial recognition are paradoxically among the highest risk factors for Crohn's disease, which involves an exaggerated immune response directed at intestinal bacteria. Here, we identified several abnormalities in the small-intestinal epithelium of Nod2(-/-) mice including inflammatory gene expression and goblet cell dysfunction, which were associated with excess interferon-γ production by intraepithelial lymphocytes and Myd88 activity. Remarkably, these abnormalities were dependent on the expansion of a common member of the intestinal microbiota Bacteroides vulgatus, which also mediated exacerbated inflammation in Nod2(-/-) mice upon small-intestinal injury. These results indicate that Nod2 prevents inflammatory pathologies by controlling the microbiota and support a multihit disease model involving specific gene-microbe interactions.


Asunto(s)
Bacteroides/inmunología , Susceptibilidad a Enfermedades/inmunología , Enteritis/inmunología , Intestino Delgado/inmunología , Proteína Adaptadora de Señalización NOD2/genética , Animales , Técnicas de Tipificación Bacteriana , Enfermedad de Crohn/inmunología , Enteritis/genética , Células Caliciformes/patología , Inflamación/genética , Inflamación/inmunología , Interferón gamma/biosíntesis , Mucosa Intestinal/inmunología , Intestino Delgado/microbiología , Linfocitos/inmunología , Ratones , Ratones Noqueados , Microbiota/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/inmunología
12.
Gastroenterology ; 160(5): 1679-1693, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33359089

RESUMEN

BACKGROUND & AIMS: Restorative proctocolectomy with ileal pouch-anal anastomosis is a surgical procedure in patients with ulcerative colitis refractory to medical therapies. Pouchitis, the most common complication, is inflammation of the pouch of unknown etiology. To define how the intestinal immune system is distinctly organized during pouchitis, we analyzed tissues from patients with and without pouchitis and from patients with ulcerative colitis using single-cell RNA sequencing (scRNA-seq). METHODS: We examined pouch lamina propria CD45+ hematopoietic cells from intestinal tissues of ulcerative colitis patients with (n = 15) and without an ileal pouch-anal anastomosis (n = 11). Further in silico meta-analysis was performed to generate transcriptional interaction networks and identify biomarkers for patients with inflamed pouches. RESULTS: In addition to tissue-specific signatures, we identified a population of IL1B/LYZ+ myeloid cells and FOXP3/BATF+ T cells that distinguish inflamed tissues, which we further validated in other scRNA-seq datasets from patients with inflammatory bowel disease (IBD). Cell-type-specific transcriptional markers obtained from scRNA-seq was used to infer representation from bulk RNA sequencing datasets, which further implicated myeloid cells expressing IL1B and S100A8/A9 calprotectin as interacting with stromal cells, and Bacteroidales and Clostridiales bacterial taxa. We found that nonresponsiveness to anti-integrin biologic therapies in patients with ulcerative colitis was associated with the signature of IL1B+/LYZ+ myeloid cells in a subset of patients. CONCLUSIONS: Features of intestinal inflammation during pouchitis and ulcerative colitis are similar, which may have clinical implications for the management of pouchitis. scRNA-seq enables meta-analysis of multiple studies, which may facilitate the identification of biomarkers to personalize therapy for patients with IBD. The processed single cell count tables are provided in Gene Expression Omnibus; GSE162335. Raw sequence data are not public and are protected by controlled-access for patient privacy.


Asunto(s)
Colitis Ulcerosa/cirugía , Perfilación de la Expresión Génica , Reservoritis/genética , Proctocolectomía Restauradora/efectos adversos , Análisis de la Célula Individual , Transcriptoma , Adolescente , Adulto , Estudios de Casos y Controles , Colitis Ulcerosa/genética , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/inmunología , Colon/patología , Reservorios Cólicos/inmunología , Reservorios Cólicos/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Mieloides/inmunología , Fenotipo , Reservoritis/inmunología , Reservoritis/patología , RNA-Seq , Linfocitos T/inmunología , Resultado del Tratamiento , Adulto Joven
13.
Blood ; 135(26): 2388-2401, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32232483

RESUMEN

A goal in precision medicine is to use patient-derived material to predict disease course and intervention outcomes. Here, we use mechanistic observations in a preclinical animal model to design an ex vivo platform that recreates genetic susceptibility to T-cell-mediated damage. Intestinal graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. We found that intestinal GVHD in mice deficient in Atg16L1, an autophagy gene that is polymorphic in humans, is reversed by inhibiting necroptosis. We further show that cocultured allogeneic T cells kill Atg16L1-mutant intestinal organoids from mice, which was associated with an aberrant epithelial interferon signature. Using this information, we demonstrate that pharmacologically inhibiting necroptosis or interferon signaling protects human organoids derived from individuals harboring a common ATG16L1 variant from allogeneic T-cell attack. Our study provides a roadmap for applying findings in animal models to individualized therapy that targets affected tissues.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Enfermedades Intestinales/prevención & control , Organoides , Linfocitos T/inmunología , Acrilamidas/farmacología , Animales , Autofagia , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Trasplante de Médula Ósea/efectos adversos , Técnicas de Cocultivo , Colon/anomalías , Femenino , Predisposición Genética a la Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Imidazoles/farmacología , Indoles/farmacología , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Intestinales/inmunología , Enfermedades Intestinales/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Necroptosis/efectos de los fármacos , Nitrilos , Células de Paneth/patología , Medicina de Precisión , Pirazoles/farmacología , Pirimidinas , Quimera por Radiación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Sulfonamidas/farmacología , Linfocitos T/trasplante
14.
FASEB J ; 35(2): e21331, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33476078

RESUMEN

Type 2 immunity plays an essential role in the maintenance of metabolic homeostasis and its disruption during obesity promotes meta-inflammation and insulin resistance. Infection with the helminth parasite Schistosoma mansoni and treatment with its soluble egg antigens (SEA) induce a type 2 immune response in metabolic organs and improve insulin sensitivity and glucose tolerance in obese mice, yet, a causal relationship remains unproven. Here, we investigated the effects and underlying mechanisms of the T2 ribonuclease omega-1 (ω1), one of the major S mansoni immunomodulatory glycoproteins, on metabolic homeostasis. We show that treatment of obese mice with plant-produced recombinant ω1, harboring similar glycan motifs as present on the native molecule, decreased body fat mass, and improved systemic insulin sensitivity and glucose tolerance in a time- and dose-dependent manner. This effect was associated with an increase in white adipose tissue (WAT) type 2 T helper cells, eosinophils, and alternatively activated macrophages, without affecting type 2 innate lymphoid cells. In contrast to SEA, the metabolic effects of ω1 were still observed in obese STAT6-deficient mice with impaired type 2 immunity, indicating that its metabolic effects are independent of the type 2 immune response. Instead, we found that ω1 inhibited food intake, without affecting locomotor activity, WAT thermogenic capacity or whole-body energy expenditure, an effect also occurring in leptin receptor-deficient obese and hyperphagic db/db mice. Altogether, we demonstrate that while the helminth glycoprotein ω1 can induce type 2 immunity, it improves whole-body metabolic homeostasis in obese mice by inhibiting food intake via a STAT6-independent mechanism.


Asunto(s)
Ingestión de Alimentos , Endorribonucleasas/uso terapéutico , Glicoproteínas/uso terapéutico , Proteínas del Helminto/uso terapéutico , Obesidad/tratamiento farmacológico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Endorribonucleasas/farmacología , Glicoproteínas/farmacología , Proteínas del Helminto/farmacología , Locomoción , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Schistosoma mansoni/enzimología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Termogénesis , Nicotiana/genética , Nicotiana/metabolismo
15.
16.
Circ Res ; 127(3): 335-353, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32336197

RESUMEN

RATIONALE: Regression of atherosclerosis is an important clinical goal; however, the pathways that mediate the resolution of atherosclerotic inflammation and reversal of plaques are poorly understood. Regulatory T cells (Tregs) have been shown to be atheroprotective, yet the numbers of these immunosuppressive cells decrease with disease progression, and whether they contribute to atherosclerosis regression is not known. OBJECTIVE: We investigated the roles of Tregs in the resolution of atherosclerotic inflammation, tissue remodeling, and plaque contraction during atherosclerosis regression. METHODS AND RESULTS: Using multiple independent mouse models of atherosclerosis regression, we demonstrate that an increase in plaque Tregs is a common signature of regressing plaques. Single-cell RNA-sequencing of plaque immune cells revealed that unlike Tregs from progressing plaques that expressed markers of natural Tregs derived from the thymus, Tregs in regressing plaques lacked Nrp1 expression, suggesting that they are induced in the periphery during lipid-lowering therapy. To test whether Tregs are required for resolution of atherosclerotic inflammation and plaque regression, Tregs were depleted using CD25 monoclonal antibody in atherosclerotic mice during apolipoprotein B antisense oligonucleotide-mediated lipid lowering. Morphometric analyses revealed that Treg depletion blocked plaque remodeling and contraction, and impaired hallmarks of inflammation resolution, including dampening of the T helper 1 response, alternative activation of macrophages, efferocytosis, and upregulation of specialized proresolving lipid mediators. CONCLUSIONS: Our data establish essential roles for Tregs in resolving atherosclerotic cardiovascular disease and provide mechanistic insight into the pathways governing plaque remodeling and regression of disease.


Asunto(s)
Aorta/metabolismo , Aterosclerosis/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Placa Aterosclerótica , Linfocitos T Reguladores/metabolismo , Animales , Anticuerpos/farmacología , Aorta/efectos de los fármacos , Aorta/inmunología , Aorta/patología , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Aterosclerosis/patología , Células Cultivadas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/metabolismo , Subunidad alfa del Receptor de Interleucina-2/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones Noqueados para ApoE , Neuropilina-1/genética , Neuropilina-1/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
17.
J Immunol ; 205(4): 1070-1083, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32661179

RESUMEN

IL-4 activates macrophages to adopt distinct phenotypes associated with clearance of helminth infections and tissue repair, but the phenotype depends on the cellular lineage of these macrophages. The molecular basis of chromatin remodeling in response to IL-4 stimulation in tissue-resident and monocyte-derived macrophages is not understood. In this study, we find that IL-4 activation of different lineages of peritoneal macrophages in mice is accompanied by lineage-specific chromatin remodeling in regions enriched with binding motifs of the pioneer transcription factor PU.1. PU.1 motif is similarly associated with both tissue-resident and monocyte-derived IL-4-induced accessible regions but has different lineage-specific DNA shape features and predicted cofactors. Mutation studies based on natural genetic variation between C57BL/6 and BALB/c mouse strains indicate that accessibility of these IL-4-induced regions can be regulated through differences in DNA shape without direct disruption of PU.1 motifs. We propose a model whereby DNA shape features of stimulation-dependent genomic elements contribute to differences in the accessible chromatin landscape of alternatively activated macrophages on different genetic backgrounds that may contribute to phenotypic variations in immune responses.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/genética , ADN/genética , Macrófagos Peritoneales/fisiología , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Sitios de Unión/genética , Inmunidad/genética , Interleucina-4/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/fisiología , Mutación/genética , Unión Proteica/genética
18.
J Immunol ; 204(12): 3389-3399, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32350082

RESUMEN

Myeloid cells are a vital component of innate immunity and comprise monocytes, macrophages, dendritic cells, and granulocytes. How myeloid cell lineage affects activation states in response to cytokines remains poorly understood. The cytokine environment and cellular infiltrate during an inflammatory response may contain prognostic features that predict disease outcome. In this study, we analyzed the transcriptional responses of human monocytes, macrophages, dendritic cells, and neutrophils in response to stimulation by IFN-γ, IFN-ß, IFN-λ, IL-4, IL-13, and IL-10 cytokines to better understand the heterogeneity of activation states in inflammatory conditions. This generated a myeloid cell-cytokine-specific response matrix that can infer representation of myeloid cells and the cytokine environment they encounter during infection, in tumors and in whole blood. Neutrophils were highly responsive to type 1 and type 2 cytokine stimulation but did not respond to IL-10. We identified transcripts specific to IFN-ß stimulation, whereas other IFN signature genes were upregulated by both IFN-γ and IFN-ß. When we used our matrix to deconvolute blood profiles from tuberculosis patients, the IFN-ß-specific neutrophil signature was reduced in tuberculosis patients with active disease, whereas the shared response to IFN-γ and IFN-ß in neutrophils was increased. When applied to glioma patients, transcripts of neutrophils exposed to IL-4/IL-13 and monocyte responses to IFN-γ or IFN-ß emerged as opposing predictors of patient survival. Hence, by dissecting how different myeloid cells respond to cytokine activation, we can delineate biological roles for myeloid cells in different cytokine environments during disease processes, especially during infection and tumor progression.


Asunto(s)
Citocinas/inmunología , Células Mieloides/inmunología , Neoplasias/inmunología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Tuberculosis/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Humanos , Inmunidad Innata/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Neoplasias/patología , Pronóstico , Tuberculosis/patología
19.
PLoS Pathog ; 15(12): e1008066, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31841569

RESUMEN

Helminth infection and dietary intake can affect the intestinal microbiota, as well as the immune system. Here we analyzed the relationship between fecal microbiota and blood profiles of indigenous Malaysians, referred to locally as Orang Asli, in comparison to urban participants from the capital city of Malaysia, Kuala Lumpur. We found that helminth infections had a larger effect on gut microbial composition than did dietary intake or blood profiles. Trichuris trichiura infection intensity also had the strongest association with blood transcriptional profiles. By characterizing paired longitudinal samples collected before and after deworming treatment, we determined that changes in serum zinc and iron levels among the Orang Asli were driven by changes in helminth infection status, independent of dietary metal intake. Serum zinc and iron levels were associated with changes in the abundance of several microbial taxa. Hence, there is considerable interplay between helminths, micronutrients and the microbiota on the regulation of immune responses in humans.


Asunto(s)
Dieta , Microbioma Gastrointestinal , Helmintiasis/sangre , Helmintiasis/microbiología , Interacciones Huésped-Parásitos/fisiología , Humanos , Hierro/sangre , Malasia , ARN/sangre , Zinc/sangre
20.
PLoS Biol ; 16(3): e2004108, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29518091

RESUMEN

Genetic and environmental factors shape host susceptibility to infection, but how and how rapidly environmental variation might alter the susceptibility of mammalian genotypes remains unknown. Here, we investigate the impacts of seminatural environments upon the nematode susceptibility profiles of inbred C57BL/6 mice. We hypothesized that natural exposure to microbes might directly (e.g., via trophic interactions) or indirectly (e.g., via microbe-induced immune responses) alter the hatching, growth, and survival of nematodes in mice housed outdoors. We found that while C57BL/6 mice are resistant to high doses of nematode (Trichuris muris) eggs under clean laboratory conditions, exposure to outdoor environments significantly increased their susceptibility to infection, as evidenced by increased worm burdens and worm biomass. Indeed, mice kept outdoors harbored as many worms as signal transducer and activator of transcription 6 (STAT6) knockout mice, which are genetically deficient in the type 2 immune response essential for clearing nematodes. Using 16S ribosomal RNA sequencing of fecal samples, we discovered enhanced microbial diversity and specific bacterial taxa predictive of nematode burden in outdoor mice. We also observed decreased type 2 and increased type 1 immune responses in lamina propria and mesenteric lymph node (MLN) cells from infected mice residing outdoors. Importantly, in our experimental design, different groups of mice received nematode eggs either before or after moving outdoors. This contrasting timing of rewilding revealed that enhanced hatching of worms was not sufficient to explain the increased worm burdens; instead, microbial enhancement and type 1 immune facilitation of worm growth and survival, as hypothesized, were also necessary to explain our results. These findings demonstrate that environment can rapidly and significantly shape gut microbial communities and mucosal responses to nematode infections, leading to variation in parasite expulsion rates among genetically similar hosts.


Asunto(s)
Susceptibilidad a Enfermedades , Ambiente , Ratones/parasitología , Tricuriasis/inmunología , Animales , Bacterias/clasificación , Bacterias/genética , Microbioma Gastrointestinal , Inmunidad Innata , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT6/genética , Trichuris
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA