Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Nat Immunol ; 25(7): 1270-1282, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38877178

RESUMO

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.


Assuntos
Interação Gene-Ambiente , Camundongos Endogâmicos C57BL , Tricuríase , Trichuris , Animais , Trichuris/imunologia , Tricuríase/imunologia , Tricuríase/parasitologia , Camundongos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Linfócitos B/imunologia , Genótipo , Interferon gama/metabolismo , Linfócitos T/imunologia , Feminino , Masculino
2.
Nat Immunol ; 24(9): 1552-1564, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37524800

RESUMO

The nuclear factor kappa B (NF-κB) family of transcription factors orchestrates signal-induced gene expression in diverse cell types. Cellular responses to NF-κB activation are regulated at the level of cell and signal specificity, as well as differential use of family members (subunit specificity). Here we used time-dependent multi-omics to investigate the selective functions of Rel and RelA, two closely related NF-κB proteins, in primary B lymphocytes activated via the B cell receptor. Despite large numbers of shared binding sites genome wide, Rel and RelA directed kinetically distinct cascades of gene expression in activated B cells. Single-cell RNA sequencing revealed marked heterogeneity of Rel- and RelA-specific responses, and sequential binding of these factors was not a major mechanism of protracted transcription. Moreover, nuclear co-expression of Rel and RelA led to functional antagonism between the factors. By rigorously identifying the target genes of each NF-κB subunit, these studies provide insights into exclusive functions of Rel and RelA in immunity and cancer.


Assuntos
NF-kappa B , Fator de Transcrição RelA , NF-kappa B/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Linfócitos B/metabolismo , Sítios de Ligação , Receptores de Antígenos/metabolismo
3.
Cell ; 180(1): 79-91.e16, 2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31866067

RESUMO

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.


Assuntos
Síndrome do Intestino Irritável/metabolismo , Proteína Amiloide A Sérica/metabolismo , Células Th17/metabolismo , Adulto , Animais , Doenças Autoimunes/metabolismo , Diferenciação Celular/imunologia , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Interleucina-17/metabolismo , Síndrome do Intestino Irritável/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Células Th1 , Células Th17/imunologia
5.
Nat Immunol ; 18(6): 642-653, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28436955

RESUMO

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.


Assuntos
Granuloma/imunologia , Fígado/imunologia , Macrófagos/imunologia , Esquistossomose mansoni/imunologia , Deficiência de Vitamina A/imunologia , Animais , Antígenos de Diferenciação/metabolismo , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/metabolismo , Interleucina-4/imunologia , Lectinas Tipo C/metabolismo , Fígado/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Cavidade Peritoneal/citologia , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/metabolismo , Schistosoma mansoni , Esquistossomose mansoni/patologia , Tretinoína/farmacologia , Proteína Desacopladora 1/metabolismo , Vitaminas/farmacologia
6.
Cell ; 158(4): 705-721, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126780

RESUMO

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:


Assuntos
Antibacterianos/administração & dosagem , Modelos Animais de Doenças , Intestinos/microbiologia , Microbiota , Obesidade/microbiologia , Penicilinas/administração & dosagem , Animais , Bactérias/classificação , Bactérias/metabolismo , Feminino , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Obesidade/metabolismo
7.
Immunity ; 49(1): 16-18, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30021142

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2 , Parasitos , Animais , Intestinos , Ácido Succínico , Paladar
8.
Immunity ; 47(6): 1024-1036, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29262347

RESUMO

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.


Assuntos
Helmintíase/imunologia , Imunidade Celular , Macrófagos/imunologia , Nematoides/imunologia , Células Th2/imunologia , Trematódeos/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Células Epiteliais/imunologia , Células Epiteliais/parasitologia , Regulação da Expressão Gênica/imunologia , Helmintíase/genética , Helmintíase/parasitologia , Homeostase/imunologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Macrófagos/parasitologia , Mastócitos/imunologia , Mastócitos/parasitologia , Microbiota/imunologia , Células Th2/parasitologia
9.
J Immunol ; 212(4): 632-644, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38180236

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos , Helmintíase , Schistosoma mansoni , Animais , Camundongos , Linfócitos T CD4-Positivos/metabolismo , Helmintíase/imunologia , Inflamação/metabolismo , Schistosoma mansoni/fisiologia
10.
J Immunol ; 211(5): 836-843, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37466391

RESUMO

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.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Camundongos , Animais , Células Mieloides/patologia , Análise de Célula Única , Infecções Estafilocócicas/microbiologia
11.
Proc Natl Acad Sci U S A ; 119(31): e2123017119, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35881802

RESUMO

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.


Assuntos
Interações Hospedeiro-Patógeno , Neutrófilos , Infecções Estafilocócicas , Staphylococcus aureus , Bacteriemia/imunologia , Bacteriemia/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Citocinas/metabolismo , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Neutrófilos/imunologia , Neutrófilos/microbiologia , Proteínas Citotóxicas Formadoras de Poros/genética , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Staphylococcus epidermidis/patogenicidade , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
12.
Immunity ; 41(2): 311-24, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25088769

RESUMO

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.


Assuntos
Bacteroides/imunologia , Suscetibilidade a Doenças/imunologia , Enterite/imunologia , Intestino Delgado/imunologia , Proteína Adaptadora de Sinalização NOD2/genética , Animais , Técnicas de Tipagem Bacteriana , Doença de Crohn/imunologia , Enterite/genética , Células Caliciformes/patologia , Inflamação/genética , Inflamação/imunologia , Interferon gama/biossíntese , Mucosa Intestinal/imunologia , Intestino Delgado/microbiologia , Linfócitos/imunologia , Camundongos , Camundongos Knockout , Microbiota/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Proteína Serina-Treonina Quinase 2 de Interação com Receptor , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais/imunologia
13.
Gastroenterology ; 160(5): 1679-1693, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33359089

RESUMO

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.


Assuntos
Colite Ulcerativa/cirurgia , Perfilação da Expressão Gênica , Pouchite/genética , Proctocolectomia Restauradora/efeitos adversos , Análise de Célula Única , Transcriptoma , Adolescente , Adulto , Estudos de Casos e Controles , Colite Ulcerativa/genética , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Colo/imunologia , Colo/patologia , Bolsas Cólicas/imunologia , Bolsas Cólicas/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Células Mieloides/imunologia , Fenótipo , Pouchite/imunologia , Pouchite/patologia , RNA-Seq , Linfócitos T/imunologia , Resultado do Tratamento , Adulto Jovem
14.
Blood ; 135(26): 2388-2401, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32232483

RESUMO

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.


Assuntos
Doença Enxerto-Hospedeiro/prevenção & controle , Enteropatias/prevenção & controle , Organoides , Linfócitos T/imunologia , Acrilamidas/farmacologia , Animais , Autofagia , Proteínas Relacionadas à Autofagia/deficiência , Proteínas Relacionadas à Autofagia/genética , Transplante de Medula Óssea/efeitos adversos , Técnicas de Cocultura , Colo/anormalidades , Feminino , Predisposição Genética para Doença , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Humanos , Imidazóis/farmacologia , Indóis/farmacologia , Doenças Inflamatórias Intestinais/patologia , Enteropatias/imunologia , Enteropatias/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Necroptose/efeitos dos fármacos , Nitrilas , Celulas de Paneth/patologia , Medicina de Precisão , Pirazóis/farmacologia , Pirimidinas , Quimera por Radiação , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Sulfonamidas/farmacologia , Linfócitos T/transplante
15.
FASEB J ; 35(2): e21331, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33476078

RESUMO

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.


Assuntos
Ingestão de Alimentos , Endorribonucleases/uso terapêutico , Glicoproteínas/uso terapêutico , Proteínas de Helminto/uso terapêutico , Obesidade/tratamento farmacológico , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Endorribonucleases/farmacologia , Glicoproteínas/farmacologia , Proteínas de Helminto/farmacologia , Locomoção , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Schistosoma mansoni/enzimologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Termogênese , Nicotiana/genética , Nicotiana/metabolismo
17.
Circ Res ; 127(3): 335-353, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32336197

RESUMO

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.


Assuntos
Aorta/metabolismo , Aterosclerose/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Placa Aterosclerótica , Linfócitos T Reguladores/metabolismo , Animais , Anticorpos/farmacologia , Aorta/efeitos dos fármacos , Aorta/imunologia , Aorta/patologia , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/imunologia , Aterosclerose/patologia , Células Cultivadas , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/metabolismo , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos Knockout para ApoE , Neuropilina-1/genética , Neuropilina-1/metabolismo , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
18.
J Immunol ; 205(4): 1070-1083, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32661179

RESUMO

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.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Cromatina/genética , DNA/genética , Macrófagos Peritoneais/fisiologia , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Animais , Sítios de Ligação/genética , Imunidade/genética , Interleucina-4/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/fisiologia , Mutação/genética , Ligação Proteica/genética
19.
J Immunol ; 204(12): 3389-3399, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32350082

RESUMO

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.


Assuntos
Citocinas/imunologia , Células Mieloides/imunologia , Neoplasias/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Tuberculose/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Humanos , Imunidade Inata/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Neoplasias/patologia , Prognóstico , Tuberculose/patologia
20.
PLoS Pathog ; 15(12): e1008066, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31841569

RESUMO

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.


Assuntos
Dieta , Microbioma Gastrointestinal , Helmintíase/sangue , Helmintíase/microbiologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Ferro/sangue , Malásia , RNA/sangue , Zinco/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA