Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Microbiol Immunol ; 67(2): 79-89, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36345699

RESUMO

A well preserved immune system is a powerful tool to prevent foreign invasion or to suppress internal mutation, which must be tightly controlled by co-stimulatory molecules in different pathophysiological conditions. One such critical molecule is OX40L expressed on activated antigen-presenting cells (APCs). Consistently, its abnormality is associated with various immunological disorders such as autoinflammatory diseases and allergy. However, a comprehensive analysis of the immune-moderating role of OX40L in dendritic cells (DCs), the most powerful APCs to initiate immune responses in vivo, and investigation of its anti-tumor efficacy in the disease setting have not been performed properly. In this study, genetic approaches for both gain-of-function and reduction-of-function were employed to reveal that OX40L was required for the efficient presentation, but not uptake, of antigens by DCs to stimulate CD4+ , as well as CD8+ T cells in vivo. As a result, CD4+ T cells were promoted towards Th1, but inhibited on Treg differentiation, by the LPS-induced OX40L on DCs, which was supported by their altered expression of co-inhibitory receptor, PD-L1. CD8+ T cells, on the other hand, also enhanced their cytotoxicity towards target cells in response to OX40L expression on the DCs transferred in vivo. Finally, in a DC-mediated tumor immunity model, the strong immunogenic roles of OX40L on DCs led to better metastasis inhibition in vivo. Collectively, our results demonstrate that OX40L could serve as a potential target in the DC-based vaccine for enhanced anti-tumor efficacy in vivo.


Assuntos
Linfócitos T CD8-Positivos , Células Dendríticas , Camundongos , Animais , Ativação Linfocitária
2.
Clin Exp Immunol ; 206(1): 12-27, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34109619

RESUMO

Atherosclerosis is an inflammatory disease with break-down of homeostatic immune regulation of vascular tissues. As a critical initiator of host immunity, dendritic cells (DCs) have also been identified in the aorta of healthy individuals and atherosclerotic patients, whose roles in regulating arterial inflammation aroused great interest. Accumulating evidence has now pointed to the fundamental roles for DCs in every developmental stage of atherosclerosis due to their myriad of functions in immunity and tolerance induction, ranging from lipid uptake, efferocytosis and antigen presentation to pro- and anti-inflammatory cytokine or chemokine secretion. In this study we provide a timely summary of the published works in this field, and comprehensively discuss both the direct and indirect roles of DCs in atherogenesis. Understanding the pathogenic roles of DCs during the development of atherosclerosis in vascular tissues would certainly help to open therapeutic avenue to the treatment of cardiovascular diseases.


Assuntos
Apresentação de Antígeno , Aorta/imunologia , Aterosclerose/imunologia , Células Dendríticas/imunologia , Tolerância Imunológica , Animais , Humanos
3.
Cell Immunol ; 349: 104049, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32057353

RESUMO

Pathogenic microorganisms utilize multiple approaches to break down host immunity in favor of their invasion, of which, cystatin C is one of the soluble factors secreted by parasites reported to affect host immunity in vivo. The cellular targets and mechanisms of action in vivo of cystatin C, however, are far from clear. As professional antigen-presenting cells, dendritic cells (DCs) are first immune cells that contact foreign pathogenic agents or their products to initiate immune responses. We previously reported that cystatin C can regulate the functions of DCs in terms of suppressed CD4+ T cell activation but enhanced Th1/Th17 differentiation via different mechanisms. Here, we further verified these regulatory effects of cystatin C on DCs in vivo. We found that the suppressive role of DC-mediated CD4+ T cell proliferation by cystatin C was partly cell-contact independent and extended to CD8+ T cells in vivo. Although cystatin C-overexpressing DCs trafficked equally as their mock-transduced counterparts, their adoptive transfer suppressed CD8+ T cell immunity and resulted in compromised tumor rejection in both vaccination and treatment regimes. Compared with their role in promoting Th17 differentiation in vivo, cystatin C-transduced DCs had far greater ability to induce T regulatory cells (Tregs), leading to collectively a higher Treg/Th17 ratio in an adoptively transferred disease model, and thus relieved Th17-dependent autoimmunity. Collectively, these data demonstrated strong in vivo evidences for immune regulatory roles of cystatin C in DCs and provided theoretical basis for the application of cystatin C-transduced cell therapy in the treatment or remission of certain autoimmune diseases. (246).


Assuntos
Transferência Adotiva , Artrite Experimental/terapia , Doenças Autoimunes/terapia , Cistatina C/fisiologia , Células Dendríticas/imunologia , Evasão Tumoral/imunologia , Transferência Adotiva/efeitos adversos , Animais , Comunicação Celular , Células Cultivadas , Cistatina C/genética , Células Dendríticas/transplante , Regulação para Baixo , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Granzimas/biossíntese , Granzimas/genética , Imunoterapia Adotiva , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Ativação Linfocitária , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Ovalbumina/imunologia , Proteínas Citotóxicas Formadoras de Poros/biossíntese , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Recombinantes/metabolismo , Organismos Livres de Patógenos Específicos , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Transdução Genética
4.
Immunol Cell Biol ; 97(10): 916-930, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31513306

RESUMO

Cystatin C is a ubiquitously expressed cysteine protease inhibitor that protects cells from either improper hydrolysis by endogenous proteases or pathogen growth/virulence by exogenous proteases. Although commonly used as a serum biomarker for evaluating renal function, cystatin C is associated with many immunological disorders under various pathophysiological conditions. How cystatin C affects immune cells, especially dendritic cells (DCs), however, is far from clear. In this study, we found that pharmacological treatment with or genetic overexpression of cystatin C in bone marrow-derived DCs (BMDCs) reduced their capacity to stimulate CD4+ T-cell proliferation, despite increased antigen uptake. This reduced capacity corresponded with reduced major histocompatibility complex-II presentation owing to diminished levels of the chaperon H2-DM in BMDCs. Instead of promoting proliferation, cystatin C promoted skewing of T cells toward proinflammatory T-helper (Th)1/Th17 differentiation. This was mediated by augmented extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase phosphorylation in BMDCs, leading to secretion of polarizing cytokines, which in turn led to the Th deviation. Collectively, our study explained the cellular and molecular basis of how this protease inhibitor can regulate immune responses, namely by affecting BMDCs and their cytokine pathway. Our results might open up an avenue for the development of therapeutic agents for the treatment of cystatin C-related immunological diseases.


Assuntos
Apresentação de Antígeno , Células da Medula Óssea/citologia , Cistatina C/metabolismo , Citocinas/biossíntese , Células Dendríticas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Peptídeos/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Polaridade Celular , Proliferação de Células , Galinhas , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Células Th1/imunologia , Células Th17/imunologia
5.
Immunol Cell Biol ; 96(9): 912-921, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29624724

RESUMO

As professional antigen-presenting cells, dendritic cells (DCs) initiate and regulate immune responses against inflammation. The invasion of pathogens into the body, however, can in turn cause the change of DCs in both activity and viability, which ultimately affect immune homeostasis. The exact mechanisms that the bacteria utilize to alter the lifespan of DCs, however, are far from clear. In this study, we found that the bacterial wall compound lipopolysaccharide (LPS) can promote the survival of GM-CSF-differentiated DCs (GM-DCs). At molecular levels, we demonstrated that GM-DCs had distinct pattern of mRNA expression for anti-apoptotic BCL-2 family members, of which, Bcl-x increased significantly following LPS stimulation. Interestingly, specific inhibition of BCL-XL protein alone was sufficient to remove the anti-apoptotic effects of LPS on BM-DCs. Further study of the signaling mechanisms revealed that although LPS can activate both Erk MAP kinase and PI3 kinase pathways, only blocking of PI3K abolished both Bcl-x upregulation and the enhanced survival phenotype, suggesting that the PI3K signaling mediated the upregulation of Bcl-x for the LPS-induced pro-survival in GM-DCs. Collectively, this study unveils a molecular mechanism that DCs adapt to maintain innate immunity against the invasion of pathogens.


Assuntos
Sobrevivência Celular , Células Dendríticas/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Apoptose , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Feminino , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais , Ativação Transcricional , Regulação para Cima
6.
Cell Immunol ; 331: 100-109, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29909072

RESUMO

Dendritic cells (DCs) contain heterogeneous populations, with classical DCs developed at steady state and monocyte-derived DCs mobilized under inflammatory conditions, although their total numbers in vivo are scares. To obtain enough quantity for immunological study or clinical application, we have previously established that bone marrow-derived DCs in the presence of Flt-3L (FL-DCs) or GM-CSF (GM-DCs) in vitro are equivalent to the steady state DCs and inflammatory DCs in vivo respectively. What difference, however, exists between these two most commonly used culture systems in DC functions and survival, and how does it correlate to the division of works by their corresponding counterparts in vivo remain ill-defined. In this study, we found that GM-DCs of inflammatory nature were more phagocytic, potent at inducing Th2 and Th17 differentiation, and had longer survival rate, whereas FL-DCs of steady state characters were stronger T cell activator and better at directing Th1 differentiation. Mechanistically, NO production induced by the LPS-activated GM-DCs could partly explain for their failure to improve T cell proliferation, and the distinct T cell differentiation profiles and viability demonstrated by the two types of DCs were underpinned by their preferential secretion of T cell polarizing cytokines and expression of anti-apoptotic genes. Such disparate functionalities and survival potentials of steady state and inflammatory DCs in vitro fit in well with their respective roles in vivo in particular immunological settings and have serious implications in translational applications.


Assuntos
Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Células Dendríticas/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Ativação Linfocitária/imunologia , Proteínas de Membrana/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
7.
Arch Microbiol ; 199(5): 775-785, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28283680

RESUMO

Non-hemolytic enterotoxin (NHE), a tri-partite, proteinaceous toxin encoded by contiguous nheA, nheB and nheC genes of Bacillus cereus sensu lato (B. cereus s.l.), is considered to be associated with the foodborne diarrheic syndrome. However, B. cereus s.l. includes a number of closely related strains, and the occurrence of NHE among them, and other members of Bacillus is unclear. Consequently, we aimed to determine the distribution and evolution of NHE within Bacillus by confirming the presence of the nheA, B and C sequences and variation within them using published data, and to analyze the genomic and genetic diversity. The phylogenetic tree of NHE proteins (NheA, NheB and NheC) from 81 different B. cereus s.l. strains was constructed. And on the genetic determinants of the NHE toxin did not bring any obvious link between the nheABC genes sequence of a strain and its virulence in the diarrhoeal pathogenesis. Analysis of the genomic diversity of the nheA, B and C loci revealed that their upstream regions were more conserved than the downstream sequences. Multilocus sequence typing schemes (MLST) based on seven concatenated housekeeping genes and nheA, B and C genes of the 75 strains were developed. The neighbor joining phylogenetic tree based on seven housekeeping genes together with nheA, B and C genes was similiar to published Bacillus phylogenetic trees. And on the genetic determinants of the NHE toxin did not bring any obvious link between the nheABC genes sequence of a strain and its virulence in the diarrhoeal pathogenesis.The results indicate that nheA, B and C genes do not affect the diversity of housekeeping genes, and this specific NHE protein does not participate in the diarrheic syndrome.


Assuntos
Bacillus cereus/genética , Bacillus cereus/patogenicidade , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Enterotoxinas/genética , Bacillus cereus/classificação , Proteínas de Bactérias/biossíntese , Toxinas Bacterianas/biossíntese , Enterotoxinas/biossíntese , Doenças Transmitidas por Alimentos/microbiologia , Humanos , Tipagem de Sequências Multilocus , Filogenia , Fatores de Virulência/genética
8.
J Biol Chem ; 289(14): 9730-40, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24570004

RESUMO

The cysteine protease inhibitor cystatin C is thought to be secreted by most cells and eliminated in the kidneys, so its concentration in plasma is diagnostic of kidney function. Low extracellular cystatin C is linked to pathologic protease activity in cancer, arthritis, atherosclerosis, aortic aneurism, and emphysema. Cystatin C forms non-inhibitory dimers and aggregates by a mechanism known as domain swapping, a property that reportedly protects against Alzheimer disease but can also cause amyloid angiopathy. Despite these clinical associations, little is known about the regulation of cystatin C production, dimerization, and secretion. We show that hematopoietic cells are major contributors to extracellular cystatin C levels in healthy mice. Among these cells, macrophages and dendritic cells (DC) are the predominant producers of cystatin C. Both cell types synthesize monomeric and dimeric cystatin C in vivo, but only secrete monomer. Dimerization occurs co-translationally in the endoplasmic reticulum and is regulated by the levels of reactive oxygen species (ROS) derived from mitochondria. Drugs or stimuli that reduce the intracellular concentration of ROS inhibit cystatin C dimerization. The extracellular concentration of inhibitory cystatin C is thus partly dependent on the abundance of macrophages and DC, and the ROS levels. These results have implications for the diagnostic use of serum cystatin C as a marker of kidney function during inflammatory processes that induce changes in DC or macrophage abundance. They also suggest an important role for macrophages, DC, and ROS in diseases associated with the protease inhibitory activity or amyloidogenic properties of cystatin C.


Assuntos
Cistatina C/biossíntese , Células-Tronco Hematopoéticas/metabolismo , Biossíntese de Proteínas/fisiologia , Multimerização Proteica/fisiologia , Animais , Cistatina C/genética , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Inflamação/genética , Inflamação/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Mutantes , Mitocôndrias/genética , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
9.
Immunol Cell Biol ; 93(5): 442-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25643616

RESUMO

A protease inhibitor, cystatin C (Cst C), is a secreted cysteine protease inhibitor abundantly expressed in body fluids. Clinically, it is mostly used to measure glomerular filtration rate as a marker for kidney function due to its relatively small molecular weight and easy detection. However, recent findings suggest that Cst C is regulated at both transcriptional and post-translational levels, and Cst C production from haematopoietic cell lineages contributes significantly to the systematic pools of Cst C. Furthermore, Cst C is directly linked to many pathologic processes through various mechanisms. Thus fluctuation of Cst C levels might have serious clinical implications rather than a mere reflection of kidney functions. Here, we summarize the pathophysiological roles of Cst C dependent and independent on its inhibition of proteases, outline its change of expression by various stimuli, and elucidate the regulatory mechanisms to control this disease-related protease inhibitor. Finally, we discuss the clinical implications of these findings for translational gains.


Assuntos
Aterosclerose/imunologia , Biomarcadores/metabolismo , Cistatina C/metabolismo , Infecções/imunologia , Metástase Neoplásica/imunologia , Animais , Linhagem da Célula , Cistatina C/genética , Regulação da Expressão Gênica , Taxa de Filtração Glomerular , Hematopoese , Humanos , Testes de Função Renal
10.
Biochem J ; 453(3): 381-91, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23692256

RESUMO

p32 [also known as HABP1 (hyaluronan-binding protein 1), gC1qR (receptor for globular head domains complement 1q) or C1qbp (complement 1q-binding protein)] has been shown previously to have both mitochondrial and non-mitochondrial localization and functions. In the present study, we show for the first time that endogenous p32 protein is a mitochondrial protein in HeLa cells under control and stress conditions. In defining the impact of altering p32 levels in these cells, we demonstrate that the overexpression of p32 increased mitochondrial fibrils. Conversely, siRNA-mediated p32 knockdown enhanced mitochondrial fragmentation accompanied by a loss of detectable levels of the mitochondrial fusion mediator proteins Mfn (mitofusin) 1 and Mfn2. More detailed ultrastructure analysis by transmission electron microscopy revealed aberrant mitochondrial structures with less and/or fragmented cristae and reduced mitochondrial matrix density as well as more punctate ER (endoplasmic reticulum) with noticeable dissociation of their ribosomes. The analysis of mitochondrial bioenergetics showed significantly reduced capacities in basal respiration and oxidative ATP turnover following p32 depletion. Furthermore, siRNA-mediated p32 knockdown resulted in differential stress-dependent effects on cell death, with enhanced cell death observed in the presence of hyperosmotic stress or cisplatin treatment, but decreased cell death in the presence of arsenite. Taken together, our studies highlight the critical contributions of the p32 protein to the morphology of mitochondria and ER under normal cellular conditions, as well as important roles of the p32 protein in cellular metabolism and various stress responses.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Transporte/genética , Retículo Endoplasmático/ultraestrutura , Células HeLa , Humanos , Immunoblotting , Microscopia Confocal , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/genética
11.
J Pharm Biomed Anal ; 248: 116338, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38971092

RESUMO

Tetrahydroxy stilbene glucoside (TSG) is a water-soluble natural product that has shown potential in treating atherosclerosis (AS). However, its underlying mechanisms remain unclear. Here, we demonstrate that an 8-week TSG treatment (100 mg/kg/d) significantly reduces atherosclerotic lesions and alleviates dyslipidemia symptoms in ApoE-/- mice. 1H nuclear magnetic resonance metabolomic analysis reveals differences in both lipid components and water-soluble metabolites in the livers of AS mice compared to control groups, and TSG treatment shifts the metabolic profiles of AS mice towards a normal state. At the transcriptional level, TSG significantly restores the expression of fatty acid metabolism-related genes (Srepb-1c, Fasn, Scd1, Gpat1, Dgat1, Pparα and Cpt1α), and regulates the expression levels of disturbed cholesterol metabolism-related genes (Srebp2, Hmgcr, Ldlr, Acat1, Acat2 and Cyp7a1) associated with lipid metabolism. Furthermore, at the cellular level, TSG remarkably polarizes aortic macrophages to their M2 phenotype. Our data demonstrate that TSG alleviates arthrosclerosis by dual-targeting to hepatic lipid metabolism and aortic M2 macrophage polarization in ApoE-/- mice, with significant implications for translational medicine and the treatment of AS using natural products.


Assuntos
Aorta , Apolipoproteínas E , Aterosclerose , Glucosídeos , Metabolismo dos Lipídeos , Fígado , Macrófagos , Estilbenos , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Camundongos , Glucosídeos/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Aorta/efeitos dos fármacos , Aorta/metabolismo , Estilbenos/farmacologia , Apolipoproteínas E/genética , Masculino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Camundongos Knockout
12.
Front Biosci (Landmark Ed) ; 29(4): 157, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38682203

RESUMO

Dendritic cells (DCs), the most efficient antigen-presenting cells (APCs), bridge the innate and adaptive immune systems. As such, the turn-over of DCs is critical during autoimmune responses, and the dysregulation of DC apoptosis could cause severe immune destruction in the host. For example, reduction of immunogenic DCs by increased apoptosis could lead to immune tolerance to pathogen infection that might allow exposure of nuclear autoantigens, whereas reduced apoptosis could result in long-term lymphocyte activation to break the immune tolerance for the development of autoimmune disease. Thus, keeping a balance between survival and apoptosis of DCs is crucial to maintain immune homeostasis. In this review, we summarize the recent development on the factors inducing DC apoptosis and their underlying mechanisms to provide insights into the immunopathogenesis of some autoimmune diseases, which could lead to effective therapeutic interventions in the clinics.


Assuntos
Apoptose , Doenças Autoimunes , Células Dendríticas , Células Dendríticas/imunologia , Humanos , Doenças Autoimunes/imunologia , Apoptose/imunologia , Animais , Tolerância Imunológica/imunologia
13.
Eur J Immunol ; 42(11): 2889-900, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22806691

RESUMO

Fms-like tyrosine kinase 3 ligand (Flt3L) is a major cytokine that drives development of dendritic cells (DCs) under steady state, whereas GM-CSF becomes a prominent influence on differentiation during inflammation. The influence GM-CSF exerts on Flt3L-induced DC development has not been thoroughly examined. Here, we report that GM-CSF alters Flt3L-induced DC development. When BM cells were cultured with both Flt3L and GM-CSF, few CD8⁺ equivalent DCs or plasmacytoid DCs developed compared to cultures supplemented with Flt3L alone. The disappearance of these two cell subsets in GM-CSF + Flt3L culture was not a result of simple inhibition of their development, but a diversion of the original differentiation trajectory to form a new cell population. As a consequence, both DC progeny and their functions were altered. The effect of GM-CSF on DC subset development was confirmed in vivo. First, the CD8⁺ DC numbers were increased under GM-CSF deficiency (when either GM-CSF or its receptor was ablated). Second, this population was decreased under GM-CSF hyperexpression (by transgenesis or by Listeria infection). Our finding that GM-CSF dominantly changes the regulation of DC development in vitro and in vivo has important implications for inflammatory diseases or GM-CSF therapy.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Animais , Apresentação de Antígeno/imunologia , Células Dendríticas/citologia , Citometria de Fluxo , Inflamação/imunologia , Inflamação/patologia , Interleucina-10/imunologia , Interleucina-12/imunologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Espécies Reativas de Oxigênio/imunologia , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/imunologia , Tirosina Quinase 3 Semelhante a fms/imunologia
14.
Expert Rev Mol Med ; 15: e6, 2013 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-23883595

RESUMO

Systemic lupus erythematosus (SLE) is a severe multi-system autoimmune disease, whereas interferon regulatory factor (IRF) 5 belongs to the family of transcription factors that modulate immune system activities. Recently, many lines of investigations suggested that IRF5 gene polymorphisms are closely associated with the disease onset of SLE. Indeed, expressed in B cells, dendritic cells (DCs), monocytes and macrophages, IRF5 could significantly affect these immune cells participating in the pathogenesis of SLE, and numerous studies implied that this transcription factor is mechanistically linked to the disease progression. Here, we comprehensively review the updated evidence indicating the roles of IRF5 in autoimmune lupus. Hopefully, the information obtained will lead to a better understanding of the pathogenesis and development of novel therapeutic strategies for the systemic autoimmune disease.


Assuntos
Fatores Reguladores de Interferon/metabolismo , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Progressão da Doença , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo
15.
J Immunol ; 186(6): 3666-73, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21300820

RESUMO

Cystatin C (CstC) is a cysteine protease inhibitor of major clinical importance. Low concentration of serum CstC is linked to atherosclerosis. CstC can prevent formation of amyloid ß associated with Alzheimer's disease and can itself form toxic aggregates. CstC regulates NO secretion by macrophages and is a TGF-ß antagonist. Finally, the serum concentration of CstC is an indicator of kidney function. Yet, little is known about the regulation of CstC expression in vivo. In this study, we demonstrate that the transcription factor IFN regulatory factor 8 (IRF-8) is critical for CstC expression in primary dendritic cells. Only those cells with IRF-8 bound to the CstC gene promoter expressed high levels of the inhibitor. Secretion of IL-10 in response to inflammatory stimuli downregulated IRF-8 expression and consequently CstC synthesis in vivo. Furthermore, the serum concentration of CstC decreased in an IL-10-dependent manner in mice treated with the TLR9 agonist CpG. CstC synthesis is therefore more tightly regulated than hitherto recognized. The mechanisms involved in this regulation might be targeted to alter CstC production, with potential therapeutic value. Our results also indicate that caution should be exerted when using the concentration of serum CstC as an indicator of kidney function in conditions in which inflammation may alter CstC production.


Assuntos
Cistatina C/biossíntese , Cistatina C/sangue , Regulação para Baixo/imunologia , Mediadores da Inflamação/fisiologia , Fatores Reguladores de Interferon/biossíntese , Interleucina-10/fisiologia , Animais , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/patologia , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Cistatina C/deficiência , Células Dendríticas/classificação , Células Dendríticas/imunologia , Células Dendríticas/patologia , Regulação para Baixo/genética , Mediadores da Inflamação/antagonistas & inibidores , Fatores Reguladores de Interferon/deficiência , Fatores Reguladores de Interferon/fisiologia , Interleucina-10/metabolismo , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
16.
Iran J Immunol ; 20(2): 145-158, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37246608

RESUMO

Background: The functions of dendritic cells (DCs) are influenced by their intracellular metabolism, in which liver kinase B1 (LKB1) plays an important role. However, due to the difficulty in isolating the DCs, the roles of LKB1 in DC maturation and functions in tumor settings have been poorly characterized. Objective: To investigate the roles of LKB1 in DC functions including phagocytosis and presentation of antigens, activation, T cell differentiation, and ultimately tumor eradication. Methods: Genetic modification of Lkb1 in the DCs was made by lentiviral transduction, and their impacts on T cell proliferation, differentiation, activity, or B16 melanoma metastasis were examined by flow cytometry, qPCR, or lung tumor nodule counting. Results: LKB1 did not affect antigen uptake and presentation by the DCs, but facilitated the stimulation of T cell proliferation. Interestingly, following T cell activation, Foxp3-expressing regulatory T cells (Treg) were increased (P=0.0267) or decreased (P=0.0195) in mice injected with Lkb1 knockdown DCs or overexpressing DCs, respectively. Further exploration revealed that LKB1 inhibited OX40L (P=0.0385) and CD86 (P=0.0111) expression, and these co-stimulatory molecules enhanced Treg proliferation, and downregulated immune suppressive cytokine IL-10 (P=0.0315). Moreover, we found that the injection of the DCs with limited LKB1 expression before tumor inoculation could reduce their production of granzyme B (P<0.0001) and perforin (P=0.0042) from CD8+T cells, thereby impairing their cytotoxicity and promoting tumor growth. Conclusion: Our data suggest that LKB1 can enhance DC-mediated T cell immunity by restraining Treg development and thereby suppressing tumor growth.


Assuntos
Neoplasias , Proteínas Serina-Treonina Quinases , Animais , Camundongos , Citocinas/metabolismo , Células Dendríticas , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T Reguladores
17.
Exp Ther Med ; 25(5): 234, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37114180

RESUMO

Both tolerogenicity and immunogenicity of dendritic cells (DCs) are regulated by their intracellular metabolism. As a rate-limiting enzyme of tryptophan (Trp) metabolism, indoleamine 2,3-dioxygenase (IDO) is involved in regulating the functions of numerous cell types, including DCs, a subset of which has a high capacity for producing IDO to control over-activated inflammation. To identify the mechanisms of IDO in DCs, stable DC lines with both gain- and reduction-of-function of IDO were established using a recombinant DNA technique. Although the IDO variation did not affect DC survival and migration, it altered Trp metabolism and other features of DCs analyzed by high-performance liquid chromatography and flow cytometry. On the surface of the DCs, IDO inhibited co-stimulatory CD86 but promoted co-inhibitory programmed cell death ligand 1 expression, and suppressed the antigen uptake, which ultimately led to the compromised ability of DCs to activate T cells. Furthermore, IDO also suppressed IL-12 secretion but enhanced that of IL-10 in DCs, which eventually induced T cells into tolerogenic phenotypes by inhibiting the differentiation of Th1 but promoting that of regulatory T cells. Collectively, the findings of the present study demonstrated that IDO is a key molecule for tolerogenic DC induction by metabolically regulating surface molecule and cytokine expression. This conclusion may lead to the targeted development of therapeutic drugs for autoimmune diseases.

18.
Int Immunopharmacol ; 116: 109758, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36706593

RESUMO

Atherosclerosis is a chronic inflammatory disease, in which immune disorders constitute an essential part of vascular pathogenesis. Accumulating evidence indicates that dendritic cells (DCs) and their tryptophan metabolisms regulate host immune responses. However, the mechanistic involvement of metabolic products from DCs in dysregulating vascular immunity during the development of atherosclerosis is far from clear. Flow cytometry examination showed immune cells were accumulated and gradually increased in the atherosclerotic lesions during the atherosclerosis progression, in which IDO+DCs were enriched. To study the role of DC-expressed IDO in the development of atherosclerosis, we made a stable IDO-overexpressing DC line (IDOoeDCs) by lentiviral infection for adoptive transfer into pro-atherosclerotic mice. Compared with DCs containing empty vector (VectorCtrlDC)-treated group, treatment of IDOoeDCs led to a significant reduction of atherosclerotic lesions in the aorta, with decreased aortic infiltration of Th1 immune cells and reduced vascular inflammation. Importantly, IDOoeDCs increased aortic kynurenine (Kyn) concentration and aryl hydrocarbon receptor (AHR) expression, concomitant with CD4+CD25+Foxp3+Treg expansion in the aortic tissues, which were abrogated by AHR antagonist treatment. These results indicate that DC-expressed IDO reduces atherosclerotic lesions by inducing aortic CD4+CD25+Foxp3+Treg expansion through IDO-Kyn-AHR axis, which may represent a novel possibility for treatment or prevention of atherosclerosis.


Assuntos
Aterosclerose , Cinurenina , Camundongos , Animais , Cinurenina/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Linfócitos T Reguladores , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Células Dendríticas , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Fatores de Transcrição Forkhead/metabolismo
19.
Cell Mol Immunol ; 20(1): 65-79, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36471114

RESUMO

The cytokine granulocyte-macrophage-colony stimulating factor (GM-CSF) possesses the capacity to differentiate monocytes into macrophages (MØs) with opposing functions, namely, proinflammatory M1-like MØs and immunosuppressive M2-like MØs. Despite the importance of these opposing biological outcomes, the intrinsic mechanism that regulates the functional polarization of MØs under GM-CSF signaling remains elusive. Here, we showed that GM-CSF-induced MØ polarization resulted in the expression of cytokine-inducible SH2-containing protein (CIS) and that CIS deficiency skewed the differentiation of monocytes toward immunosuppressive M2-like MØs. CIS deficiency resulted in hyperactivation of the JAK-STAT5 signaling pathway, consequently promoting downregulation of the transcription factor Interferon Regulatory Factor 8 (IRF8). Loss- and gain-of-function approaches highlighted IRF8 as a critical regulator of the M1-like polarization program. In vivo, CIS deficiency induced the differentiation of M2-like macrophages, which promoted strong Th2 immune responses characterized by the development of severe experimental asthma. Collectively, our results reveal a CIS-modulated mechanism that clarifies the opposing actions of GM-CSF in MØ differentiation and uncovers the role of GM-CSF in controlling allergic inflammation.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos , Macrófagos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Monócitos/metabolismo , Citocinas/metabolismo , Fatores Reguladores de Interferon/metabolismo , Diferenciação Celular
20.
Eur J Immunol ; 41(9): 2585-95, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21660938

RESUMO

Resident CD8(+) DCs perform several functions, including cross-presenting antigen and rapidly engulfing the Gram-positive intracellular pathogen Listeria monocytogenes. Little is known about how these functions of CD8(+) DCs are modulated. Here, we show that granulocyte-macrophage CSF (GM-CSF), a cytokine that exists at low levels at steady state but is elevated during infection and inflammation, enhances cross-presentation and rapid uptake of L. monocytogenes by resident CD8(+) DCs. This previously unrecognized functional enhancement of CD8(+) DCs by GM-CSF was independent of promoting DC survival in vitro. Enhancement of these functions by GM-CSF was also marked by CD103 expression on CD8(+) DCs that was strongly regulated by GM-CSF. Our findings not only identify GM-CSF as a key molecule regulating CD8(+) DC function, but also as a factor responsible for functional heterogeneity of CD8(+) DCs that is at least substantially demarcated by CD103 expression.


Assuntos
Antígenos CD/metabolismo , Células Dendríticas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Cadeias alfa de Integrinas/metabolismo , Listeria monocytogenes/imunologia , Listeriose/imunologia , Animais , Antígenos de Bactérias/imunologia , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos CD8/biossíntese , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/patologia , Células Cultivadas , Apresentação Cruzada/genética , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Regulação da Expressão Gênica/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Imunomodulação , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/imunologia , Listeria monocytogenes/patogenicidade , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Baço/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA