Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Int Immunol ; 33(12): 653-657, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34049386

RESUMO

Pulmonary fibrosis is caused by the interplay between genetic and environmental factors. Recent studies have revealed various genes associated with idiopathic pulmonary fibrosis, as well as the causative genes for familial pulmonary fibrosis. Although increased death or dysfunction of type 2 alveolar epithelial (AT2) cells has been detected in lung specimens from pulmonary fibrosis patients, it remains unclear whether and how AT2 cell death or dysfunction is responsible for the progression of pulmonary fibrosis. A recent study showed that increased AT2 cell necroptosis is the initial event in pulmonary fibrosis by analyzing patients with familial pulmonary fibrosis and an animal model that harbors the same mutation as patients. The contribution of AT2 cell necroptosis to the pathogenesis of pulmonary fibrosis has not been identified in animal model studies, which validates the effectiveness of genetic analysis of familial diseases to uncover unknown pathogeneses. Thus, further extensive genetic studies of pulmonary fibrosis along with functional studies based on genetic analysis will be crucial not only in elucidating the precise disease process but also, ultimately, in identifying novel treatment strategies for both familial and non-familial pulmonary fibrosis.


Assuntos
Modelos Animais de Doenças , Fibrose Pulmonar Idiopática/genética , Animais , Fibrose Pulmonar Idiopática/imunologia , Fibrose Pulmonar Idiopática/patologia
2.
PLoS Biol ; 17(5): e3000262, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31071093

RESUMO

Intestinal intraepithelial lymphocytes (IELs) expressing CD8αα on αß T cells (TCRαß+CD8αα+ IELs) have suppressive capabilities in enterocolitis, but the mechanism that maintains homeostasis and cell number is not fully understood. Here, we demonstrated that the number of TCRαß+CD8αα+ IELs was severely reduced in mice lacking recombination signal binding protein for immunoglobulin kappa J region (Rbpj) or Notch1 and Notch2 in T cells. Rbpj-deficient TCRαß+CD8αα+ IELs expressed low levels of Atp8a2, which encodes a protein with flippase activity that regulates phospholipid asymmetry of plasma membrane such as flipping phosphatidylserine in the inner leaflet of plasma membrane. Rbpj-deficient TCRαß+CD8αα+ IELs cannot maintain phosphatidylserine in the inner leaflet of the plasma membrane. Furthermore, depletion of intestinal macrophages restored TCRαß+CD8αα+ IELs in Rbpj-deficient mice, suggesting that exposure of phosphatidylserine on the plasma membrane in Rbpj-deficient TCRαß+CD8αα+ IELs acts as an "eat-me" signal. Together, these results revealed that Notch-Atp8a2 is a fundamental regulator for IELs and highlighted that membrane phospholipid asymmetry controlled by Notch-mediated flippase expression is a critical determinant in setting or balancing the number of TCRαß+CD8αα+ IELs.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Linfócitos Intraepiteliais/metabolismo , Fosfolipídeos/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores Notch/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Proteínas de Transferência de Fosfolipídeos/metabolismo
3.
Nat Immunol ; 9(10): 1140-7, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18724371

RESUMO

The acquisition of cytotoxic effector function by CD8(+) T cells is crucial for the control of intracellular infection and tumor invasion. However, it remains unclear which signaling pathways are required for the differentiation of CD8(+) cytotoxic T lymphocytes. We show here that Notch2-deficient T cells had impaired differentiation into cytotoxic T lymphocytes. In addition, dendritic cells with lower expression of the Notch ligand Delta-like 1 induced the differentiation of cytotoxic T lymphocytes less efficiently. We found that the intracellular domain of Notch2 interacted with a phosphorylated form of the transcription factor CREB1, and together these proteins bound the transcriptional coactivator p300 to form a complex on the promoter of the gene encoding granzyme B. Our results suggest that the highly regulated, dynamic control of T cell cytotoxicity depends on the integration of Notch2 and CREB1 signals.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular/imunologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/citologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/imunologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/imunologia , Células Dendríticas/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Granzimas/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Receptor Notch2/imunologia , Linfócitos T Citotóxicos/imunologia , Transcrição Gênica/imunologia , Fatores de Transcrição de p300-CBP/imunologia , Fatores de Transcrição de p300-CBP/metabolismo
4.
Int Immunol ; 31(10): 631-637, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30169676

RESUMO

Immunoproteasomes degrade ubiquitin-coupled proteins and play a role in creating peptides for presentation by MHC class I proteins. Studies of gene-deficient mice, in which each immunoproteasomal subunit was affected, have demonstrated that dysfunction of immunoproteasomes leads to immunodeficiency, i.e. reduced expression of MHC class I and attenuation of CD8 T-cell responses. Recent studies, however, have uncovered a new type of autoinflammatory syndrome characterized by fever, nodular erythema and progressive partial lipodystrophy that is caused by genetic mutations in immunoproteasome subunits. These mutations disturbed the assembly of immunoproteasomes, which led to reduced proteasomal activity and thus accumulation of ubiquitin-coupled proteins. Those findings suggest that immunoproteasomes function as anti-inflammatory machinery in humans. The discovery of a new type of autoinflammatory syndrome caused by dysregulated immunoproteasomes provides novel insights into the important roles of immunoproteasomes in inflammation as well as the spectrum of autoinflammatory diseases.


Assuntos
Doenças Autoimunes/metabolismo , Inflamação/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Humanos
5.
Proc Natl Acad Sci U S A ; 114(47): E10151-E10160, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29109283

RESUMO

In secondary lymphoid organs, development and homeostasis of stromal cells such as podoplanin (Pdpn)-positive fibroblastic reticular cells (FRCs) are regulated by hematopoietic cells, but the cellular and molecular mechanisms of such regulation have remained unclear. Here we show that ablation of either signal regulatory protein α (SIRPα), an Ig superfamily protein, or its ligand CD47 in conventional dendritic cells (cDCs) markedly reduced the number of CD4+ cDCs as well as that of Pdpn+ FRCs and T cells in the adult mouse spleen. Such ablation also impaired the survival of FRCs as well as the production by CD4+ cDCs of tumor necrosis factor receptor (TNFR) ligands, including TNF-α, which was shown to promote the proliferation and survival of Pdpn+ FRCs. CD4+ cDCs thus regulate the steady-state homeostasis of FRCs in the adult spleen via the production of TNFR ligands, with the CD47-SIRPα interaction in cDCs likely being indispensable for such regulation.


Assuntos
Células Dendríticas/imunologia , Fibroblastos/imunologia , Homeostase/imunologia , Receptores Imunológicos/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Baço/imunologia , Animais , Antígenos CD4/genética , Antígenos CD4/imunologia , Antígeno CD47/genética , Antígeno CD47/imunologia , Sobrevivência Celular , Células Dendríticas/citologia , Fibroblastos/citologia , Regulação da Expressão Gênica , Homeostase/genética , Linfonodos/citologia , Linfonodos/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Imunológicos/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais , Baço/citologia , Linfócitos T/citologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
6.
J Immunol ; 199(6): 1989-1997, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28779023

RESUMO

Dendritic cells (DCs) are important for adaptive immune responses through the activation of T cells. The molecular interplay between DCs and T cells determines the magnitude of T cell responses or outcomes of functional differentiation of T cells. In this study, we demonstrated that DCs in mice that are Rbpj deficient in CD11c+ cells (Rbpj-/- mice) promoted the differentiation of IL-17A-producing Th17 cells. Rbpj-deficient DCs expressed little Aldh1a2 protein that is required for generating retinoic acid. Those DCs exhibited a reduced ability for differentiating regulatory T cells induced by TGF-ß. Rbpj protein directly regulated Aldh1a2 transcription by binding to its promoter region. The overexpression of Aldh1a2 in Rbpj-deficient DCs negated their Th17-promoting ability. Transfer of naive CD4+ T cells into Rag1-deficient Rbpj-/- mice enhanced colitis with increased Th17 and reduced induced regulatory T cells (iTreg) compared with control Rag1-deficient mice. The cotransfer of iTreg and naive CD4+ T cells into Rag1-deficient Rbpj-/- mice improved colitis compared with transfer of naive CD4+ T cell alone. Furthermore, cotransfer of DCs from Rbpj-/- mice that overexpressed Aldh1a2 or Notch-stimulated DCs together with naive CD4+ T cells into Rbpj-/-Rag1-deficient mice led to reduced colitis with increased iTreg numbers. Therefore, our studies identify Notch signaling in DCs as a crucial balancer of Th17/iTreg, which depends on the direct regulation of Aldh1a2 transcription in DCs.


Assuntos
Colite/imunologia , Células Dendríticas/imunologia , Retinal Desidrogenase/metabolismo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Família Aldeído Desidrogenase 1 , Animais , Antígeno CD11c/metabolismo , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Genes RAG-1 , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Interleucina-17/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Notch/metabolismo , Retinal Desidrogenase/genética , Linfócitos T Reguladores/transplante , Tretinoína/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(16): 5986-91, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24711412

RESUMO

The gastrointestinal tract comes into direct contact with environmental agents, including bacteria, viruses, and foods. Intestine-specific subsets of immune cells maintain gut homeostasis by continuously sampling luminal antigens and maintaining immune tolerance. CD11c(+)CX3CR1(+) cells sample luminal antigens in the small intestine and contribute to the trafficking of bacteria to lymph nodes under dysbiotic conditions. The molecular mechanisms crucial for the differentiation of CD11c(+)CX3CR1(+) cells remain unclear. Here we demonstrate that the Notch1- or Notch2-Rbpj axis is essential for the development of CD11c(+)CX3CR1(+) cells. In mice in which Rbpj or Notch1 and Notch2 were deleted from CD11c(+) cells, there was a deficit of CD11c(+)CX3CR1(+) cells and an accumulation of CD11c(low)CX3CR1(+) cells. The CD11c(low)CX3CR1(+) cells could not differentiate to CD11c(+)CX3CR1(+) cells, suggesting that CD11c(low)CX3CR1(+) cells represent a lineage distinct from CD11c(+)CX3CR1(+) cells. These data indicate that Notch signaling is essential for lineage fixation of intestinal CD11c(+)CX3CR1(+) cells.


Assuntos
Antígeno CD11c/metabolismo , Diferenciação Celular , Intestino Delgado/citologia , Receptores de Quimiocinas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Receptor 1 de Quimiocina CX3C , Contagem de Células , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
8.
Biochem Biophys Res Commun ; 444(4): 628-33, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24491544

RESUMO

CD4(+) T cell activation is controlled by signaling through the T cell receptor in addition to various co-receptors, and is also affected by their interactions with effector and regulatory T cells in the microenvironment. Inflammatory bowel diseases (IBD) are caused by the persistent activation and expansion of auto-aggressive CD4(+) T cells that attack intestinal epithelial cells. However, the molecular basis for the persistent activation of CD4(+) T cells in IBD remains unclear. In this study, we investigated how the CD98 heavy chain (CD98hc, Slc3a2) affected the development of colitis in an experimental animal model. Transferring CD98hc-deficient CD4(+)CD25(-) T cells into Rag2(-/-) mice did not cause colitis accompanied by increasing Foxp3(+) inducible regulatory T cells. By comparison, CD98hc-deficient naturally occurring regulatory T cells (nTregs) had a decreased capability to suppress colitis induced by CD4(+)CD25(-) T cells, although CD98hc-deficient mice did not have a defect in the development of nTregs. Blocking CD98hc with an anti-CD98 blocking antibody prevented the development of colitis. Our results indicate that CD98hc regulates the expansion of autoimmune CD4(+) T cells in addition to controlling nTregs functions, which suggests the CD98hc as an important target molecule for establishing strategies for treating colitis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Colite/imunologia , Colite/patologia , Colo/patologia , Cadeia Pesada da Proteína-1 Reguladora de Fusão/imunologia , Animais , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Colite/genética , Colo/imunologia , Colo/metabolismo , Proteínas de Ligação a DNA/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Deleção de Genes , Subunidade alfa de Receptor de Interleucina-2/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
9.
J Immunol ; 188(5): 2227-34, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22291182

RESUMO

The interplay of CD4(+) and CD8(+) T cells targeting autoantigens is responsible for the progression of a number of autoimmune diseases, including type 1 diabetes mellitus (T1D). Understanding the molecular mechanisms that regulate T cell activation is crucial for designing effective therapies for autoimmune diseases. We probed a panel of Abs with T cell-modulating activity and identified a mAb specific for the H chain of CD98 (CD98hc) that was able to suppress T cell proliferation. The anti-CD98hc mAb also inhibited Ag-specific proliferation and the acquisition of effector function by CD4(+) and CD8(+) T cells in vitro and in vivo. Injection of the anti-CD98hc mAb completely prevented the onset of cyclophosphamide-induced diabetes in NOD mice. Treatment of diabetic NOD mice with anti-CD98hc reversed the diabetic state to normal levels, coincident with decreased proliferation of CD4(+) T cells. Furthermore, treatment of diabetic NOD mice with CD98hc small interfering RNA resolved T1D. These data indicate that strategies targeting CD98hc might have clinical application for treating T1D and other T cell-mediated autoimmune diseases.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Cadeia Pesada da Proteína-1 Reguladora de Fusão/fisiologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Ciclofosfamida/administração & dosagem , Diabetes Mellitus Tipo 1/induzido quimicamente , Feminino , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/imunologia , Inibidores do Crescimento/genética , Inibidores do Crescimento/metabolismo , Inibidores do Crescimento/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Ratos , Ratos Wistar
10.
J Basic Microbiol ; 54(6): 558-67, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23686946

RESUMO

Bacteroides is one of the most predominant groups of human gut microbiota. Recent metagenomic analyses and studies on gnotobiotic mice demonstrated the tight association of Bacteroides with epithelial function, the gut immune system and systemic metabolism in the host. The mariner family transposon shows relatively low target site specificity and has hosts ranging from prokaryotes to eukaryotes. Thereby, random mutagenesis using the mariner family transposon is expected to identify key molecules for human-Bacteroides symbiosis. In this study, we constructed the plasmid pMI07 to deliver the gene cassette (ermF/ITR), which harbors the erythromycin resistant marker (ermF) and the inverted repeat sequences (ITRs) recognized by Himar1 transposase, to Bacteroides via electrotransformation. pMI07 successfully delivered ermF/ITR to the Bacteroides genomes and generated thousands of insertion mutants/µg of pMI07 in B. thetaiotaomicron, B. fragilis, B. ovatus, and also, although to a lesser extent, B. vulgatus. Analyses of the ermF/ITR insertion sites in B. thetaiotaomicron and B. vulgatus revealed that the cassette targeted the dinucleotide TA and integrated into the genomes in an unbiased manner. The data reported here will provide useful information for transposon mutagenesis in Bacteroides species, which will enable identification of the genes responsible for their unique phenotypes.


Assuntos
Bacteroides/genética , Elementos de DNA Transponíveis , Genética Microbiana/métodos , Mutagênese Insercional/métodos , Plasmídeos , Transformação Bacteriana
11.
Blood ; 117(1): 128-34, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20971948

RESUMO

Notch receptor-mediated signaling is involved in the developmental process and functional modulation of lymphocytes, as well as in mast cell differentiation. Here, we investigated whether Notch signaling is required for antipathogen host defense regulated by mast cells. Mast cells were rarely found in the small intestine of wild-type C57BL/6 mice but accumulated abnormally in the lamina propria of the small-intestinal mucosa of the Notch2-conditional knockout mice in naive status. When transplanted into mast cell-deficient W(sh)/W(sh) mice, Notch2-null bone marrow-derived mast cells were rarely found within the epithelial layer but abnormally localized to the lamina propria, whereas control bone marrow-derived mast cells were mainly found within the epithelial layer. After the infection of Notch2 knockout and control mice with L3 larvae of Strongyloides venezuelensis, the abundant number of mast cells was rapidly mobilized to the epithelial layer in the control mice. In contrast, mast cells were massively accumulated in the lamina propria of the small intestinal mucosa in Notch2-conditional knockout mice, accompanied by impaired eradication of Strongyloides venezuelensis. These findings indicate that cell-autonomous Notch2 signaling in mast cells is required for proper localization of intestinal mast cells and further imply a critical role of Notch signaling in the host-pathogen interface in the small intestine.


Assuntos
Mucosa Intestinal/imunologia , Intestino Delgado/imunologia , Mastócitos/imunologia , Receptor Notch2/fisiologia , Estrongiloidíase/imunologia , Animais , Membrana Basal/metabolismo , Medula Óssea/imunologia , Transplante de Medula Óssea , Movimento Celular , Células Cultivadas , Feminino , Integrases/metabolismo , Mucosa Intestinal/parasitologia , Intestino Delgado/parasitologia , Masculino , Mastócitos/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Strongyloides/patogenicidade , Estrongiloidíase/parasitologia
12.
Proc Natl Acad Sci U S A ; 107(13): 5943-8, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20231432

RESUMO

CD4(+) helper T (Th) cells differentiate toward distinct effector cell lineages characterized by their distinct cytokine expression patterns and functions. Multiple Th cell populations secrete IL-22 that contributes to both protective and pathological inflammatory responses. Although the differentiation of IL-22-producing Th cells is controlled by the aryl hydrocarbon receptor (AhR), little is known about the regulatory mechanisms inducing physiological stimulators for AhR. Here, we show that Notch signaling enhances IL-22 production by CD4(+) T cells by a mechanism involving AhR stimulation. Notch-mediated stimulation of CD4(+) T cells increased the production of IL-22 even in the absence of STAT3. CD4(+) T cells from RBP-J-deficient mice had little ability to produce IL-22 through T cell receptor-mediated stimulation. RBP-J-deficient mice were highly susceptible to the detrimental immunopathology associated with ConA-induced hepatitis with little IL-22 production by CD4(+) T cells. Exogenous IL-22 protected RBP-J-deficient mice from ConA-induced hepatitis. Notch signaling promoted production of endogenous stimulators for AhR, which further augmented IL-22 secretion. Our studies identify a Notch-AhR axis that regulates IL-22 expression and fine-tunes immune system control of inflammatory responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Interleucinas/biossíntese , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores Notch/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular , Feminino , Hepatite Animal/etiologia , Hepatite Animal/imunologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estrutura Terciária de Proteína , Receptores Notch/química , Receptores Notch/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT3/deficiência , Fator de Transcrição STAT3/genética , Transdução de Sinais , Interleucina 22
13.
Front Immunol ; 14: 1065790, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36776885

RESUMO

FXR is a key molecule that modulates anti-inflammatory activity in the intestinal-liver axis. Although FXR has pleiotropic functions including regulation of liver inflammation and activation of macrophages, it remains unclear whether it is involved in macrophage polarization. In this paper we demonstrated that stimulation of macrophages derived from the bone marrow using an FXR agonist activated polarization toward M2 but not M1 macrophages. The treatment of mice with chitin skewed macrophage polarization towards M2 macrophages, while co-treatment with an FXR agonist further promoted the polarization toward M2 macrophages in vivo. This skewed polarization towards M2 macrophages by an FXR agonist was accompanied by increased expression of signaling molecules related to the retinoic acid receptor. Inhibition of the retinoic acid receptor suppressed FXR agonist-mediated M2 macrophage polarization, indicating that this polarization was, at least, partly dependent on the retinoic acid receptor pathway. These data demonstrate that FXR has a role in polarization toward M2 macrophages and suggest a possible therapeutic potential of FXR agonists in M2 macrophage-related conditions.


Assuntos
Macrófagos , Receptores Citoplasmáticos e Nucleares , Transdução de Sinais , Animais , Camundongos , Anti-Inflamatórios/metabolismo , Macrófagos/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptores Citoplasmáticos e Nucleares/agonistas
14.
Sci Rep ; 13(1): 14541, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37752134

RESUMO

Rapid, sensitive detection of biomolecules is important for biosensing of infectious pathogens as well as biomarkers and pollutants. For example, biosensing of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still strongly required for the fight against coronavirus disease 2019 (COVID-19) pandemic. Here, we aim to achieve the rapid and sensitive detection of SARS-CoV-2 nucleocapsid protein antigen by enhancing the performance of optical biosensing based on optical frequency combs (OFC). The virus-concentration-dependent optical spectrum shift produced by antigen-antibody interactions is transformed into a photonic radio-frequency (RF) shift by a frequency conversion between the optical and RF regions in the OFC, facilitating rapid and sensitive detection with well-established electrical frequency measurements. Furthermore, active-dummy temperature-drift compensation with a dual-comb configuration enables the very small change in the virus-concentration-dependent signal to be extracted from the large, variable background signal caused by temperature disturbance. The achieved performance of dual-comb biosensing will greatly enhance the applicability of biosensors to viruses, biomarkers, environmental hormones, and so on.


Assuntos
Técnicas Biossensoriais , COVID-19 , Vírus , Humanos , COVID-19/diagnóstico , SARS-CoV-2 , Teste para COVID-19 , Antígenos Virais
15.
Eur J Immunol ; 41(5): 1309-20, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21469122

RESUMO

Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) required for T-cell activation and are classified into several subtypes by phenotypic and functional characteristics. However, it remains unclear if distinct transcription factors control the development of each DC subpopulation. In this report, we demonstrate that Notch signaling controls the development of a novel DC subtype that expresses Thy1 (Thy1(+) DCs). Overstimulation of bone marrow cells with the Notch ligand Delta-like 1 promoted the development of Thy1(+) DCs. Thy1(+) DCs are characterized as CD11c(+) MHC class II(+) NK1.1(-) B220(-) CD8α(+) , and are present in the thymus but not in the spleen and lymph nodes. Thymic Thy1(+) DCs are able to capture exogenous proteins and delete CD4(+) CD8(+) T cells. Transplantation experiments demonstrated that CD44(+) CD25(-) and CD44(+) CD25(+) thymocytes can differentiate into Thy1(+) DCs. Recombination signal binding protein for immunoglobulin kappa J region (RBP-J) deficiency in lineage-negative bone marrow cells, but not CD11c(+) cells, disrupted Thy1(+) DC development in the thymus. Our data indicate that Notch signaling controls the development of a novel type of Thy1-expressing DC in the thymus that possibly controls negative selection, and indicates that there may be highly regulated, differential transcriptional control of DC development. Furthermore, our findings suggest that Notch signaling regulates T-cell development not only by intrinsically inducing T-cell lineage-specific gene programs, but also by regulating negative selection through Thy1(+) DCs.


Assuntos
Células Dendríticas/imunologia , Receptores Notch/metabolismo , Antígenos Thy-1/genética , Timo/imunologia , Animais , Apresentação de Antígeno , Western Blotting , Células da Medula Óssea/metabolismo , Antígeno CD11c/análise , Diferenciação Celular , Células Dendríticas/classificação , Células Dendríticas/citologia , Citometria de Fluxo , Receptores de Hialuronatos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Subunidade alfa de Receptor de Interleucina-2/análise , Peptídeos e Proteínas de Sinalização Intracelular , Ativação Linfocitária , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Receptores Notch/genética , Transdução de Sinais , Linfócitos T , Antígenos Thy-1/metabolismo , Timo/citologia , Timo/embriologia
16.
Eur J Immunol ; 41(10): 2828-39, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21953641

RESUMO

IL-23 plays a critical role in the expansion of highly proinflammatory Th17 cells secreting IL-17 and IL-22. Recently, we demonstrated that Notch signaling drives IL-22 secretion through the aryl hydrocarbon receptor (AHR) and plays a protective role in Con A-induced hepatitis. In this study, we investigated the role of IL-23 in hepatitis using IL-23p19- and IL-17-deficient mice. In WT mice, the injection of Con A induced the upregulation of various cytokines, which included IL-23, IL-22, IL-17, IFN-γ and TNF-α. In IL-23p19-deficient mice, exacerbated hepatitis was observed and serum IL-22 and IL-17 levels were greatly reduced, whereas in IL-17-deficient mice, ameliorated hepatitis was observed. The injection of exogenous IL-22 protected p19-deficient mice from hepatitis, whereas the injection of exogenous IL-23 significantly increased the serum levels of not only IL-22 but also IL-17, and less effectively protected against hepatitis in IL-17-dependent and -independent manners. Finally, it was revealed that STAT3, STAT4 and Notch contributed to the production of both the cytokines, and that the AHR was important only for IL-22 production in response to Con A and IL-23 in liver mononuclear cells. These results suggest that IL-23 plays a protective role in hepatitis through IL-22 production and also a pathological role via IL-17-dependent and -independent mechanisms.


Assuntos
Hepatite Animal/imunologia , Hepatite Animal/metabolismo , Interleucina-17/metabolismo , Subunidade p19 da Interleucina-23/metabolismo , Interleucina-23 , Interleucinas/metabolismo , Animais , Concanavalina A , Citocinas/biossíntese , Interleucina-17/sangue , Interleucina-17/genética , Interleucina-23/administração & dosagem , Interleucina-23/biossíntese , Interleucina-23/metabolismo , Interleucina-23/farmacologia , Subunidade p19 da Interleucina-23/genética , Interleucinas/administração & dosagem , Interleucinas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Th17/imunologia , Células Th17/metabolismo , Interleucina 22
17.
Biochem Biophys Res Commun ; 418(4): 701-7, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22293205

RESUMO

B cells are classified into several subsets depending on their functions, marker expression pattern and localization. Marginal zone B (MZB) cells are a distinct lineage from follicular B cells, and regulate host defenses against blood-borne pathogens. Notch2/RBP-J signaling regulates the development of MZB cells by interacting with delta-like 1 ligand, although the target genes for Notch2 signaling remain unclear. We identified Fos as an upregulated gene in LPS-stimulated B cells that received Notch2 signaling. Fos is expressed in CD21(high)CD23(low) MZB cells at a higher level compared to CD21(Int)CD23(high) follicular B cells. Deleting the Notch2 gene in CD19(+) B cells decreased Fos expression in B cells. Overexpression of Fos in Notch2-deficient B cells or bone marrow cells partially restored MZB development. Fos promoter activity was upregulated by Notch2 signaling, indicating that Notch2 directly controls Fos transcription associated with MZB development. These data identify Fos as one of the target genes for Notch2 signaling that is crucial for MZB development.


Assuntos
Linfócitos B/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Oncogênicas v-fos/genética , Receptor Notch2/metabolismo , Animais , Linfócitos B/metabolismo , Genes Reporter , Luciferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Receptor Notch2/genética , Baço/citologia , Transcrição Gênica
18.
Int Arch Allergy Immunol ; 157(1): 65-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21912175

RESUMO

BACKGROUND: Notch signaling pathways govern immune function and the regulation of Th1 and Th2 differentiation. We previously demonstrated essential interactions between Notch on CD4+ T cells and Jagged1 on antigen-presenting cells in Th2 differentiation for the full development of allergen-induced airway hyperresponsiveness (AHR) and allergic airway inflammation. METHODS: Bone marrow-derived dendritic cells (BMDCs) were differentiated and incubated with different preparations of ovalbumin (OVA), including lipopolysaccharide (LPS)-depleted and LPS-spiked preparations. In some experiments recipient mice also received soluble Jagged1-Fc in addition to allergen-pulsed BMDCs. Ten days following transfer of BMDCs, mice were exposed to three airway challenges with OVA, and airway responsiveness to inhaled methacholine, airway inflammation and cytokine production were monitored 48 h later. Notch ligand expression was assessed by real-time PCR. RESULTS: Induction of Jagged1 expression on antigen-pulsed BMDCs was dependent on low-dose endotoxin. In vivo, transfer of endotoxin-free, antigen-pulsed BMDCs failed to induce AHR or airway eosinophilia on allergen challenge. However, administration of exogenous Jagged1-Fc together with endotoxin-free, allergen-pulsed BMDCs fully restored the responses to allergen challenge. CONCLUSIONS: These data demonstrate that LPS regulates the expression of Jagged1 on BMDCs, which is essential for the full development of lung allergic responses.


Assuntos
Antígenos/imunologia , Asma/imunologia , Asma/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Células Dendríticas/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lipopolissacarídeos/imunologia , Proteínas de Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Transferência Adotiva , Animais , Modelos Animais de Doenças , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Jagged-1 , Ligantes , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Receptores Notch/metabolismo , Proteínas Serrate-Jagged
19.
Am J Hematol ; 87(6): 637-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22488407

RESUMO

Familial hemophagocytic lymphohistiocytosis (FHL) is a disorder of immune homeostasis characterized by fever, cytopenias, hepatosplenomegaly, and coagulopathy. We studied the outcomes of 13 FHL patients who underwent the first unrelated cord blood transplantation (UCBT) after non-myeloablative conditionings. The major regimen consisted of fludarabine (FLU; n = 12)+melphalan (MEL; n = 11)± low-dose total body irradiation (TBI 2-4 Gy; n = 6). The median age at presentation and period to UCBT were 6 and 5 months, respectively. Central nervous system (CNS) disease developed in one infant at diagnosis, and in two others until UCBT. HLH activity was controlled in all but one at the time of UCBT. Ten patients had early engraftment on median day 21 with no grade >2 treatment-related toxicity and two controllable grade >2 acute GVHD. Two patients with early rejection successfully underwent subsequent UCBT after myeloablative conditioning. Two others had late graft failure following mixed donor chimerism. Two deaths occurred from HLH; early liver failure and late CNS disease. Of 11 FLU+MEL-conditioned patients, the frequency of disease-free complete engraftment was higher for MEL (≥120 mg/m(2) )+TBI, or high-dose MEL (180 mg/m(2) ) than for others (83% vs. 25%, p = 0.036). The FLU+MEL-based non-myeloablative regimen was acceptable for FHL infants undergoing UCBT, although further studies will be needed for confirmation.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Linfo-Histiocitose Hemofagocítica/cirurgia , Condicionamento Pré-Transplante/métodos , Transfusão de Componentes Sanguíneos , Doadores de Sangue , Pré-Escolar , Terapia Combinada , Transplante de Células-Tronco de Sangue do Cordão Umbilical/efeitos adversos , Intervalo Livre de Doença , Feminino , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro , Humanos , Lactente , Recém-Nascido , Japão , Linfo-Histiocitose Hemofagocítica/terapia , Masculino , Melfalan/administração & dosagem , Reoperação , Estudos Retrospectivos , Terapia de Salvação , Resultado do Tratamento , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados , Irradiação Corporal Total
20.
J Immunol ; 184(9): 4673-8, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20351182

RESUMO

CD8(+) T cells play a central role in cancer immunosurveillance, and the efficient induction of CTLs against tumor Ags is required for successful immunotherapy for cancer patients. Notch signaling directly regulates the transcription of effector molecules in CTLs. However, it remains unclear whether Notch signaling in CD8(+) T cells is required for antitumor CTL responses and whether modulation of Notch signaling can augment antitumor CTL responses. In this study, we demonstrate that signaling by Notch2 but not Notch1 in CD8(+) T cells is required for antitumor CTL responses. Notch2(flox/flox) mice crossed with E8I-cre transgenic (N2F/F-E8I) mice, in which the Notch2 gene is absent only in CD8(+) T cells, die earlier than control mice after inoculation with OVA-expressing EG7 thymoma cells. In contrast, Notch1(flox/flox) mice crossed with E8I-cre transgenic mice inoculated with EG7 cells die comparable to control mice, indicating that Notch2 is crucial for exerting antitumor CTL responses. Injection of anti-Notch2 agonistic Ab or delta-like 1-overexpressing dendritic cells augmented the antitumor response in C57BL/6 mice inoculated with EG7 cells. These findings indicate that Notch2 signaling in CD8(+) T cells is required for generating potent antitumor CTLs, thus providing a crucial target for augmenting tumor immune responses.


Assuntos
Receptor Notch2/fisiologia , Transdução de Sinais/imunologia , Timoma/imunologia , Timoma/prevenção & controle , Neoplasias do Timo/imunologia , Neoplasias do Timo/prevenção & controle , Animais , Anticorpos Monoclonais/administração & dosagem , Células CHO , Linhagem Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Imunoterapia Adotiva , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptor Notch1/deficiência , Receptor Notch1/genética , Receptor Notch1/fisiologia , Receptor Notch2/agonistas , Receptor Notch2/deficiência , Receptor Notch2/imunologia , Transdução de Sinais/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Timoma/genética , Timoma/patologia , Neoplasias do Timo/genética , Neoplasias do Timo/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA