Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1168635, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37215144

RESUMO

Introduction: Macrophages significantly contribute to the regulation of vessel formation under physiological and pathological conditions. Although the angiogenesis-regulating role of alternatively polarized macrophages is quite controversial, a growing number of evidence shows that they can participate in the later phases of angiogenesis, including vessel sprouting and remodeling or regression. However, the epigenetic and transcriptional regulatory mechanisms controlling this angiogenesis-modulating program are not fully understood. Results: Here we show that IL-4 can coordinately regulate the VEGFA-VEGFR1 (FLT1) axis via simultaneously inhibiting the proangiogenic Vegfa and inducing the antiangiogenic Flt1 expression in murine bone marrow-derived macrophages, which leads to the attenuated proangiogenic activity of alternatively polarized macrophages. The IL-4-activated STAT6 and IL-4-STAT6 signaling pathway-induced EGR2 transcription factors play a direct role in the transcriptional regulation of the Vegfa-Flt1 axis. We demonstrated that this phenomenon is not restricted to the murine bone marrow-derived macrophages, but can also be observed in different murine tissue-resident macrophages ex vivo and parasites-elicited macrophages in vivo with minor cell type-specific differences. Furthermore, IL-4 exposure can modulate the hypoxic response of genes in both murine and human macrophages leading to a blunted Vegfa/VEGFA and synergistically induced Flt1/FLT1 expression. Discussion: Our findings establish that the IL-4-activated epigenetic and transcriptional program can determine angiogenesis-regulating properties in alternatively polarized macrophages under normoxic and hypoxic conditions.


Assuntos
Interleucina-4 , Fator A de Crescimento do Endotélio Vascular , Humanos , Camundongos , Animais , Interleucina-4/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Macrófagos/metabolismo , Transdução de Sinais , Regulação da Expressão Gênica , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
Mol Cell ; 83(1): 121-138.e7, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36521490

RESUMO

Cell cycle (CC) facilitates cell division via robust, cyclical gene expression. Protective immunity requires the expansion of pathogen-responsive cell types, but whether CC confers unique gene expression programs that direct the subsequent immunological response remains unclear. Here, we demonstrate that single macrophages (MFs) adopt different plasticity states in CC, which leads to heterogeneous cytokine-induced polarization, priming, and repolarization programs. Specifically, MF plasticity to interferon gamma (IFNG) is substantially reduced during S-G2/M, whereas interleukin 4 (IL-4) induces S-G2/M-biased gene expression, mediated by CC-biased enhancers. Additionally, IL-4 polarization shifts the CC-phase distribution of MFs toward the G2/M phase, providing a subpopulation-specific mechanism for IL-4-induced, dampened IFNG responsiveness. Finally, we demonstrate CC-dependent MF responses in murine and human disease settings in vivo, including Th2-driven airway inflammation and pulmonary fibrosis, where MFs express an S-G2/M-biased tissue remodeling gene program. Therefore, MF inflammatory and regenerative responses are gated by CC in a cyclical, phase-dependent manner.


Assuntos
Cromatina , Interleucina-4 , Humanos , Camundongos , Animais , Interleucina-4/genética , Interleucina-4/farmacologia , Cromatina/genética , Cromatina/metabolismo , Macrófagos/metabolismo , Interferon gama/genética , Interferon gama/farmacologia , Ciclo Celular/genética , Divisão Celular
3.
Immunity ; 55(11): 2006-2026.e6, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36323312

RESUMO

Prior exposure to microenvironmental signals could fundamentally change the response of macrophages to subsequent stimuli. It is believed that T helper-2 (Th2)-cell-type cytokine interleukin-4 (IL-4) and Toll-like receptor (TLR) ligand-activated transcriptional programs mutually antagonize each other, and no remarkable convergence has been identified between them. In contrast, here, we show that IL-4-polarized macrophages established a hyperinflammatory gene expression program upon lipopolysaccharide (LPS) exposure. This phenomenon, which we termed extended synergy, was supported by IL-4-directed epigenomic remodeling, LPS-activated NF-κB-p65 cistrome expansion, and increased enhancer activity. The EGR2 transcription factor contributed to the extended synergy in a macrophage-subtype-specific manner. Consequently, the previously alternatively polarized macrophages produced increased amounts of immune-modulatory factors both in vitro and in vivo in a murine Th2 cell-type airway inflammation model upon LPS exposure. Our findings establish that IL-4-induced epigenetic reprogramming is responsible for the development of inflammatory hyperresponsiveness to TLR activation and contributes to lung pathologies.


Assuntos
Interleucina-4 , Lipopolissacarídeos , Camundongos , Animais , Interleucina-4/metabolismo , Lipopolissacarídeos/metabolismo , Ligantes , Epigenômica , Macrófagos/metabolismo , Receptores Toll-Like/metabolismo , Epigênese Genética , NF-kappa B/metabolismo
4.
FEBS Open Bio ; 11(12): 3218-3229, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34358410

RESUMO

The changing extra- and intracellular microenvironment calls for rapid cell fate decisions that are precisely and primarily regulated at the transcriptional level. The cellular components of the immune system are excellent examples of how cells respond and adapt to different environmental stimuli. Innate immune cells such as macrophages are able to modulate their transcriptional programs and epigenetic regulatory networks through activation and repression of particular genes, allowing them to quickly respond to a rapidly changing environment. Tissue macrophages are essential components of different immune- and nonimmune cell-mediated physiological mechanisms in mammals and are widely used models for investigating transcriptional regulatory mechanisms. Therefore, it is critical to unravel the distinct sets of transcription activators, repressors, and coregulators that play roles in determining tissue macrophage identity and functions during homeostasis, as well as in diseases affecting large human populations, such as metabolic syndromes, immune-deficiencies, and tumor development. In this review, we will focus on transcriptional repressors that play roles in tissue macrophage development and function under physiological conditions.


Assuntos
Macrófagos/imunologia , Especificidade de Órgãos/imunologia , Transcrição Gênica/genética , Animais , Diferenciação Celular/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Mucosa Intestinal/imunologia , Macrófagos/fisiologia , Macrófagos Alveolares/imunologia , Microglia/imunologia , Baço/imunologia , Fatores de Transcrição/metabolismo
5.
iScience ; 24(4): 102312, 2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33855282

RESUMO

Mesenchymal stromal cell-like (MSCl) cells generated from human embryonic stem cells are considered to be an eligible cell line to model the immunomodulatory behavior of mesenchymal stromal cells (MSCs) in vitro. Dendritic cells (DCs) are essential players in the maintenance and restoration of the sensitive balance between tolerance and immunity. Here, the effects of MSCl cells on the in vitro differentiation of human monocytes into DCs were investigated. MSCl cells promote the differentiation of CTLA-4 expressing DCs via the production of all-trans retinoic acid (ATRA) functioning as a ligand of RARα, a key nuclear receptor in DC development. These semi-matured DCs exhibit an ability to activate allogeneic, naive T cells and polarize them into IL-10 + IL-17 + double-positive T helper cells in a CTLA-4-dependent manner. Mapping the molecular mechanisms of MSC-mediated indirect modulation of DC differentiation may help to expand MSCs' clinical application in cell-free therapies.

6.
Front Immunol ; 11: 572960, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33013932

RESUMO

To detect replicating viruses, dendritic cells (DCs) utilize cytoplasmic retinoic acid inducible gene-(RIG) I-like receptors (RLRs), which play an essential role in the subsequent activation of antiviral immune responses. In this study, we aimed to explore the role of the mammalian target of rapamycin (mTOR) in the regulation of RLR-triggered effector functions of human monocyte-derived DCs (moDCs) and plasmacytoid DCs (pDCs). Our results show that RLR stimulation increased the phosphorylation of the mTOR complex (mTORC) 1 and mTORC2 downstream targets p70S6 kinase and Akt, respectively, and this process was prevented by the mTORC1 inhibitor rapamycin as well as the dual mTORC1/C2 kinase inhibitor AZD8055 in both DC subtypes. Furthermore, inhibition of mTOR in moDCs impaired the RLR stimulation-triggered glycolytic switch, which was reflected by the inhibition of lactate production and downregulation of key glycolytic genes. Blockade of mTOR diminished the ability of RLR-stimulated moDCs and pDCs to secret type I interferons (IFNs) and pro-inflammatory cytokines, while it did not affect the phenotype of DCs. We also found that mTOR blockade decreased the phosphorylation of Tank-binding kinase 1 (TBK1), which mediates RLR-driven cytokine production. In addition, rapamycin abrogated the ability of both DC subtypes to promote the proliferation and differentiation of IFN-y and Granzyme B producing CD8 + T cells. Interestingly, AZD8055 was much weaker in its ability to decrease the T cell proliferation capacity of DCs and was unable to inhibit the DC-triggered production of IFN-y and Granyzme B by CD8 + T cells. Here we demonstrated for the first time that mTOR positively regulates the RLR-mediated antiviral activity of human DCs. Further, we show that only selective inhibition of mTORC1 but not dual mTORC1/C2 blockade suppresses effectively the T cell stimulatory capacity of DCs that should be considered in the development of new generation mTOR inhibitors and in the improvement of DC-based vaccines.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteína DEAD-box 58/metabolismo , Células Dendríticas/imunologia , Monócitos/imunologia , Receptores Imunológicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Vacinas/imunologia , Viroses/imunologia , Antineoplásicos/farmacologia , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Células Cultivadas , Humanos , Interferon Tipo I/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Morfolinas/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
7.
Nanotechnol Sci Appl ; 13: 11-22, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32280204

RESUMO

INTRODUCTION: In this work we selected components, developed technology and studied a number of parameters of polymer nanocomposite materials, remembering that the material would have high optical and good mechanical characteristics, good sorption ability in order to ensure high value of the optical signal for a short time while maintaining the initial geometric shape. In addition, if this nanocomposite is used for medicine and biology (biocompatible or biocidal materials or the creation of a sensor based on it), the material must be non-toxic and/or biocompatible. We study the creation of polymer nanocomposites which may be applied as biocompatible materials with new functional parameters. MATERIAL AND METHODS: A number of polymer nanocomposites based on various urethane-acrylate monomers and nanoparticles of gold, silicon oxides, zinc and/or titanium oxides are obtained, their mechanical (microhardness) properties and wettability (contact angle) are studied. The set of required, biology-related properties of these materials, such as toxicity and sorption of microorganisms are also investigated in order to prove their possible applicability. RESULTS AND DISCUSSION: The composition of the samples influences their microhardness and the value of contact angle, which means that varying with the monomer and the metallic, oxide nanoparticles composition, we could change these parameters. Besides it, the set of required, biology-related properties of these materials, such as toxicity and sorption of microorganisms were also investigated in order to prove their possible applicability. It was shown that the materials are non-toxic, the adhesion of microorganisms on their surface also could be varied by changing their composition. CONCLUSION: The presented polymer nanocomposites with different compositions of monomer and the presence of nanoparticles in them are prospective material for a possible bio-application as it is biocompatible, not toxic. The sorption of microorganism could be varied depending on the type of bacterias, the monomer composition, and nanoparticles.

8.
Front Immunol ; 8: 427, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28458670

RESUMO

Dendritic cells are considered as the main coordinators of both mucosal and systemic immune responses, thus playing a determining role in shaping the outcome of effector cell responses. However, it is still uncovered how primary human monocyte-derived DC (moDC) populations drive the polarization of helper T (Th) cells in the presence of commensal bacteria harboring unique immunomodulatory properties. Furthermore, the individual members of the gut microbiota have the potential to modulate the outcome of immune responses and shape the immunogenicity of differentiating moDCs via the activation of retinoic acid receptor alpha (RARα). Here, we report that moDCs are able to mediate robust Th1 and Th17 responses upon stimulation by Escherichia coli Schaedler or Morganella morganii, while the probiotic Bacillus subtilis strain limits this effect. Moreover, physiological concentrations of all-trans retinoic acid (ATRA) are able to re-program the differentiation of moDCs resulting in altered gene expression profiles of the master transcription factors RARα and interferon regulatory factor 4, and concomitantly regulate the cell surface expression levels of CD1 proteins and also the mucosa-associated CD103 integrin to different directions. It was also demonstrated that the ATRA-conditioned moDCs exhibited enhanced pro-inflammatory cytokine secretion while reduced their co-stimulatory and antigen-presenting capacity thus reducing Th1 and presenting undetectable Th17 type responses against the tested microbiota strains. Importantly, these regulatory circuits could be prevented by the selective inhibition of RARα functionality. These results altogether demonstrate that selected commensal bacterial strains are able to drive strong effector immune responses by moDCs, while in the presence of ATRA, they support the development of both tolerogenic and inflammatory moDC in a RARα-dependent manner.

9.
Front Microbiol ; 8: 321, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28326063

RESUMO

The vertebrate gut symbiont Lactobacillus reuteri exhibits strain-specific adhesion and health-promoting properties. Here, we investigated the role of the mucus adhesins, CmbA and MUB, upon interaction of L. reuteri ATCC PTA 6475 and ATCC 53608 strains with human monocyte-derived dendritic cells (moDCs). We showed that mucus adhesins increased the capacity of L. reuteri strains to interact with moDCs and promoted phagocytosis. Our data also indicated that mucus adhesins mediate anti- and pro-inflammatory effects by the induction of interleukin-10 (IL-10), tumor necrosis factor alpha (TNF-α), IL-1ß, IL-6, and IL-12 cytokines. L. reuteri ATCC PTA 6475 and ATCC 53608 were exclusively able to induce moDC-mediated Th1 and Th17 immune responses. We further showed that purified MUB activates moDCs and induces Th1 polarized immune responses associated with increased IFNγ production. MUB appeared to mediate these effects via binding to C-type lectin receptors (CLRs), as shown using cell reporter assays. Blocking moDCs with antibodies against DC-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN) or Dectin-2 did not affect the uptake of the MUB-expressing strain, but reduced the production of TNF-α and IL-6 by moDCs significantly, in line with the Th1 polarizing capacity of moDCs. The direct interaction between MUB and CLRs was further confirmed by atomic force spectroscopy. Taken together these data suggest that mucus adhesins expressed at the cell surface of L. reuteri strains may exert immunoregulatory effects in the gut through modulating the Th1-promoting capacity of DCs upon interaction with C-type lectins.

10.
Immunol Lett ; 171: 60-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26861999

RESUMO

Ion channels are crucially important for the activation and proliferation of T lymphocytes, and thus, for the function of the immune system. Previous studies on the effects of channel blockers on T cell proliferation reported variable effectiveness due to differing experimental systems. Therefore our aim was to investigate how the strength of the mitogenic stimulation influences the efficiency of cation channel blockers in inhibiting activation, cytokine secretion and proliferation of T cells under standardized conditions. Human peripheral blood lymphocytes were activated via monoclonal antibodies targeting the TCR-CD3 complex and the co-stimulator CD28. We applied the blockers of Kv1.3 (Anuroctoxin), KCa3.1 (TRAM-34) and CRAC (2-Apb) channels of T cells either alone or in combination with rapamycin, the inhibitor of the mammalian target of rapamycin (mTOR). Five days after the stimulation ELISA and flow cytometric measurements were performed to determine IL-10 and IFN-γ secretion, cellular viability and proliferation. Our results showed that ion channel blockers and rapamycin inhibit IL-10 and IFN-γ secretion and cell division in a dose-dependent manner. Simultaneous application of the blockers for each channel along with rapamycin was the most effective, indicating synergy among the various activation pathways. Upon increasing the extent of mitogenic stimulation the anti-proliferative effect of the ion channel blockers diminished. This phenomenon may be important in understanding the fine-tuning of T cell activation.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Ativados pela Liberação de Cálcio/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canal de Potássio Kv1.3/antagonistas & inibidores , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Compostos de Boro/farmacologia , Células Cultivadas , Sinergismo Farmacológico , Humanos , Imunossupressores/farmacologia , Interferon gama/metabolismo , Interleucina-4/metabolismo , Mitógenos/imunologia , Pirazóis/farmacologia , Receptores de Antígenos de Linfócitos T/metabolismo , Sirolimo/farmacologia , Linfócitos T/fisiologia
11.
J Leukoc Biol ; 92(1): 159-69, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22517920

RESUMO

Cytosolic RIG-I-like helicases (RLR) are PRRs involved in type I IFN production and antiviral immunity. This study focuses to the comparison of the expression, function, and signaling cascades associated to RLR in the previously identified CD14(-)DC-SIGN(+)PPARγ(low)CD1a(+) and CD14(low)DC-SIGN(+)PPARγ(high)CD1a(-) human moDC subsets. Our results revealed that the expression of RLR genes and proteins as well as the activity of the coupled signaling pathways are significantly higher in the CD1a(+) subset than in its phenotypically and functionally distinct counterpart. Specific activation of RLR in moDCs by poly(I:C) or influenza virus was shown to induce the secretion of IFN-ß via IRF3, whereas induction of proinflammatory cytokine responses were predominantly controlled by TLR3. The requirement of RLR-mediated signaling in CD1a(+) moDCs for priming naïve CD8(+) T lymphocytes and inducing influenza virus-specific cellular immune responses was confirmed by RIG-I/MDA5 silencing, which abrogated these functions. Our results demonstrate the subset-specific activation of RLR and the underlying mechanisms behind its cytokine secretion profile and identify CD1a(+) moDCs as an inflammatory subset with specialized functional activities. We also provide evidence that this migratory DC subset can be detected in human tonsil and reactive LNs.


Assuntos
RNA Helicases DEAD-box/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunidade Inata , Influenza Humana/imunologia , Interferon beta/metabolismo , Orthomyxoviridae/imunologia , Células Apresentadoras de Antígenos/imunologia , Antígenos CD1/genética , Antígenos CD1/metabolismo , Western Blotting , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Citocinas/metabolismo , RNA Helicases DEAD-box/genética , Células Dendríticas/citologia , Humanos , Técnicas Imunoenzimáticas , Influenza Humana/metabolismo , Influenza Humana/virologia , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Linfonodos/metabolismo , Tonsila Palatina/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...