Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 35(7): e21684, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34159634

RESUMO

Adenine nucleotides represent crucial immunomodulators in the extracellular environment. The ectonucleotidases CD39 and CD73 are responsible for the sequential catabolism of ATP to adenosine via AMP, thus promoting an anti-inflammatory milieu induced by the "adenosine halo". AMPD2 intracellularly mediates AMP deamination to IMP, thereby both enhancing the degradation of inflammatory ATP and reducing the formation of anti-inflammatory adenosine. Here, we show that this enzyme is expressed on the surface of human immune cells and its predominance may modify inflammatory states by altering the extracellular milieu. Surface AMPD2 (eAMPD2) expression on monocytes was verified by immunoblot, surface biotinylation, mass spectrometry, and immunofluorescence microscopy. Flow cytometry revealed enhanced monocytic eAMPD2 expression after TLR stimulation. PBMCs from patients with rheumatoid arthritis displayed significantly higher levels of eAMPD2 expression compared with healthy controls. Furthermore, the product of AMPD2-IMP-exerted anti-inflammatory effects, while the levels of extracellular adenosine were not impaired by an increased eAMPD2 expression. In summary, our study identifies eAMPD2 as a novel regulator of the extracellular ATP-adenosine balance adding to the immunomodulatory CD39-CD73 system.


Assuntos
5'-Nucleotidase/metabolismo , AMP Desaminase/metabolismo , Adenina/metabolismo , Trifosfato de Adenosina/metabolismo , Leucócitos/metabolismo , Apirase , Células Cultivadas , Proteínas Ligadas por GPI/metabolismo , Humanos
2.
Front Immunol ; 10: 1744, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396235

RESUMO

Glucocorticoids regulate fundamental processes of the human body and control cellular functions such as cell metabolism, growth, differentiation, and apoptosis. Moreover, endogenous glucocorticoids link the endocrine and immune system and ensure the correct function of inflammatory events during tissue repair, regeneration, and pathogen elimination via genomic and rapid non-genomic pathways. Due to their strong immunosuppressive, anti-inflammatory and anti-allergic effects on immune cells, tissues and organs, glucocorticoids significantly improve the quality of life of many patients suffering from diseases caused by a dysregulated immune system. Despite the multitude and seriousness of glucocorticoid-related adverse events including diabetes mellitus, osteoporosis and infections, these agents remain indispensable, representing the most powerful, and cost-effective drugs in the treatment of a wide range of rheumatic diseases. These include rheumatoid arthritis, vasculitis, and connective tissue diseases, as well as many other pathological conditions of the immune system. Depending on the therapeutically affected cell type, glucocorticoid actions strongly vary among different diseases. While immune responses always represent complex reactions involving different cells and cellular processes, specific immune cell populations with key responsibilities driving the pathological mechanisms can be identified for certain autoimmune diseases. In this review, we will focus on the mechanisms of action of glucocorticoids on various leukocyte populations, exemplarily portraying different autoimmune diseases as heterogeneous targets of glucocorticoid actions: (i) Abnormalities in the innate immune response play a crucial role in the initiation and perpetuation of giant cell arteritis (GCA). (ii) Specific types of CD4+ T helper (Th) lymphocytes, namely Th1 and Th17 cells, represent important players in the establishment and course of rheumatoid arthritis (RA), whereas (iii) B cells have emerged as central players in systemic lupus erythematosus (SLE). (iv) Allergic reactions are mainly triggered by several different cytokines released by activated Th2 lymphocytes. Using these examples, we aim to illustrate the versatile modulating effects of glucocorticoids on the immune system. In contrast, in the treatment of lymphoproliferative disorders the pro-apoptotic action of glucocorticoids prevails, but their mechanisms differ depending on the type of cancer. Therefore, we will also give a brief insight into the current knowledge of the mode of glucocorticoid action in oncological treatment focusing on leukemia.


Assuntos
Artrite Reumatoide/imunologia , Arterite de Células Gigantes/imunologia , Glucocorticoides/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Células Th1/imunologia , Células Th17/imunologia , Artrite Reumatoide/patologia , Arterite de Células Gigantes/patologia , Humanos , Lúpus Eritematoso Sistêmico/patologia , Células Th1/patologia , Células Th17/patologia
3.
Int J Mol Sci ; 19(8)2018 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-30087255

RESUMO

Mesenchymal stem/stromal cells (MSCs) are stem cells of the connective tissue, possess a plastic phenotype, and are able to differentiate into various tissues. Besides their role in tissue regeneration, MSCs perform additional functions as a modulator or inhibitor of immune responses. Due to their pleiotropic function, MSCs have also gained therapeutic importance for the treatment of autoimmune diseases and for improving fracture healing and cartilage regeneration. However, the therapeutic/immunomodulatory mode of action of MSCs is largely unknown. Here, we describe that MSCs express the inhibitory receptor CTLA-4 (cytotoxic T lymphocyte antigen 4). We show that depending on the environmental conditions, MSCs express different isoforms of CTLA-4 with the secreted isoform (sCTLA-4) being the most abundant under hypoxic conditions. Furthermore, we demonstrate that the immunosuppressive function of MSCs is mediated mainly by the secretion of CTLA-4. These findings open new ways for treatment when tissue regeneration/fracture healing is difficult.


Assuntos
Antígeno CTLA-4/imunologia , Células-Tronco Mesenquimais/imunologia , Adipogenia , Antígeno CTLA-4/análise , Células Cultivadas , Humanos , Tolerância Imunológica , Células-Tronco Mesenquimais/citologia , Osteogênese
4.
Microvasc Res ; 116: 34-44, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28993199

RESUMO

Hypoxia driven angiogenesis is a prominent feature of tissue regeneration, inflammation and tumor growth and is regulated by hypoxia-inducible factor (HIF)-1 and -2. The distinct functions of HIFs in the hypoxia-induced angiogenesis and metabolic switch of endothelial cells are still unknown and therefore aim of this study. We investigated the role of HIF-1 and -2 in the adaptation of immortalized human microvascular endothelial cells (HMEC-1) to hypoxic conditions (1% O2) in terms of angiogenesis, cytokine secretion, gene expression and ATP/ADP-ratio using shRNA-mediated reduction of the oxygen sensitive α-subunits of either HIF-1 or HIF-2 or the combination of both. Reduction of HIF-1α diminished cellular energy, hypoxia-induced glycolytic gene expression, and angiogenesis not altering pro-angiogenic factors. Reduction of HIF-2α diminished hypoxia-induced pro-angiogenic factors, enhanced anti-angiogenic factors and attenuated angiogenesis not altering glycolytic gene expression. Reduction of both HIFs reduced cell survival, gene expression of glycolytic enzymes and pro-angiogenic factors as compared to the corresponding control. Finally, we identified the macrophage migration inhibitory factor (MIF) to be redundantly regulated by HIF-1 and HIF-2 and to be essential in the process of hypoxia-driven angiogenesis. Our results demonstrate a major impact of HIF-1 and HIF-2 on hypoxia-induced angiogenesis indicating distinct but also overlapping functions of HIF-1 and HIF-2. These findings open new possibilities for therapeutic approaches by specifically targeting the HIF-1 and HIF-2 or their target MIF.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Endoteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Microvasos/metabolismo , Neovascularização Fisiológica , Adaptação Fisiológica , Antígenos de Diferenciação de Linfócitos B/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular , Linhagem Celular , Microambiente Celular , Regulação Enzimológica da Expressão Gênica , Glicólise , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/genética , Microvasos/citologia , Neovascularização Fisiológica/genética , Transdução de Sinais
5.
Int J Mol Sci ; 18(3)2017 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-28282868

RESUMO

Immunologically restricted patients such as those with autoimmune diseases or malignancies often suffer from delayed or insufficient fracture healing. In human fracture hematomas and the surrounding bone marrow obtained from immunologically restricted patients, we analyzed the initial inflammatory phase on cellular and humoral level via flow cytometry and multiplex suspension array. Compared with controls, we demonstrated higher numbers of immune cells like monocytes/macrophages, natural killer T (NKT) cells, and activated T helper cells within the fracture hematomas and/or the surrounding bone marrow. Also, several pro-inflammatory cytokines such as Interleukin (IL)-6 and Tumor necrosis factor α (TNFα), chemokines (e.g., Eotaxin and RANTES), pro-angiogenic factors (e.g., IL-8 and Macrophage migration inhibitory factor: MIF), and regulatory cytokines (e.g., IL-10) were found at higher levels within the fracture hematomas and/or the surrounding bone marrow of immunologically restricted patients when compared to controls. We conclude here that the inflammatory activity on cellular and humoral levels at fracture sites of immunologically restricted patients considerably exceeds that of control patients. The initial inflammatory phase profoundly differs between these patient groups and is probably one of the reasons for prolonged or insufficient fracture healing often occurring within immunologically restricted patients.


Assuntos
Consolidação da Fratura/imunologia , Hospedeiro Imunocomprometido , Inflamação/imunologia , Indutores da Angiogênese/metabolismo , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Estudos de Casos e Controles , Citocinas/metabolismo , Feminino , Fraturas Ósseas/imunologia , Fraturas Ósseas/patologia , Hematoma/imunologia , Hematoma/patologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Leucócitos/patologia , Masculino , Neovascularização Fisiológica , Fenótipo
6.
Int J Nanomedicine ; 11: 5883-5896, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27877036

RESUMO

Combined individually tailored methods for diagnosis and therapy (theragnostics) could be beneficial in destructive diseases, such as rheumatoid arthritis. Nanoparticles are promising candidates for theragnostics due to their excellent biocompatibility. Nanoparticle modifications, such as improved surface coating, are in development to meet various requirements, although safety concerns mean that modified nanoparticles require further review before their use in medical applications is permitted. We have previously demonstrated that iron oxide nanoparticles with amino-polyvinyl alcohol (a-PVA) adsorbed on their surfaces have the unwanted effect of increasing human immune cell cytokine secretion. We hypothesized that this immune response was caused by free-floating PVA. The aim of the present study was to prevent unwanted immune reactions by further surface modification of the a-PVA nanoparticles. After cross-linking of PVA to nanoparticles to produce PVA-grafted nanoparticles, and reduction of their zeta potential, the effects on cell viability and cytokine secretion were analyzed. PVA-grafted nanoparticles still stimulated elevated cytokine secretion from human immune cells; however, this was inhibited after reduction of the zeta potential. In conclusion, covalent cross-linking of PVA to nanoparticles and adjustment of the surface charge rendered them nontoxic to immune cells, nonimmunogenic, and potentially suitable for use as theragnostic agents.


Assuntos
Células Sanguíneas/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Compostos Férricos/química , Nanopartículas de Magnetita/administração & dosagem , Álcool de Polivinil/química , Adsorção , Células Sanguíneas/metabolismo , Citocinas/metabolismo , Citometria de Fluxo , Humanos , Nanopartículas de Magnetita/química
7.
Int J Nanomedicine ; 10: 3429-45, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26056442

RESUMO

Nanotechnology provides new opportunities in human medicine, mainly for diagnostic and therapeutic purposes. The autoimmune disease rheumatoid arthritis (RA) is often diagnosed after irreversible joint structural damage has occurred. There is an urgent need for a very early diagnosis of RA, which can be achieved by more sensitive imaging methods. Superparamagnetic iron oxide nanoparticles (SPION) are already used in medicine and therefore represent a promising tool for early diagnosis of RA. The focus of our work was to investigate any potentially negative effects resulting from the interactions of newly developed amino-functionalized amino-polyvinyl alcohol coated (a-PVA) SPION (a-PVA-SPION), that are used for imaging, with human immune cells. We analyzed the influence of a-PVA-SPION with regard to cell survival and cell activation in human whole blood in general, and in human monocytes and macrophages representative of professional phagocytes, using flow cytometry, multiplex suspension array, and transmission electron microscopy. We found no effect of a-PVA-SPION on the viability of human immune cells, but cytokine secretion was affected. We further demonstrated that the percentage of viable macrophages increased on exposure to a-PVA-SPION. This effect was even stronger when a-PVA-SPION were added very early in the differentiation process. Additionally, transmission electron microscopy analysis revealed that both monocytes and macrophages are able to endocytose a-PVA-SPION. Our findings demonstrate an interaction between human immune cells and a-PVA-SPION which needs to be taken into account when considering the use of a-PVA-SPION in human medicine.


Assuntos
Artrite Reumatoide/sangue , Nanopartículas de Magnetita/química , Álcool de Polivinil/química , Sobrevivência Celular/efeitos dos fármacos , Citocinas/sangue , Endocitose/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Nanopartículas de Magnetita/efeitos adversos , Microscopia Eletrônica de Transmissão , Monócitos/efeitos dos fármacos , Testes de Toxicidade/métodos
8.
Int Rev Immunol ; 34(1): 34-49, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25259409

RESUMO

Energy homeostasis is a hallmark of cell survival and maintenance of cell function. Here we focus on the impact of cellular energy metabolism on T-lymphocyte differentiation, activation, and function in health and disease. We describe the role of transcriptional and posttranscriptional regulation of lymphocyte metabolism on immune functions of T cells. We also summarize the current knowledge about T-lymphocyte adaptations to inflammation and hypoxia, and the impact on T-cell behavior of pathophysiological hypoxia (as found in tumor tissue, chronically inflamed joints in rheumatoid arthritis and during bone regeneration). A better understanding of the underlying mechanisms that control immune cell metabolism and immune response may provide therapeutic opportunities to alter the immune response under conditions of either immunosuppression or inflammation, potentially targeting infections, vaccine response, tumor surveillance, autoimmunity, and inflammatory disorders.


Assuntos
Artrite Reumatoide/metabolismo , Metabolismo Energético/genética , Hipóxia/metabolismo , Neoplasias/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Diferenciação Celular , Proliferação de Células , Anergia Clonal/genética , Regulação da Expressão Gênica , Humanos , Hipóxia/tratamento farmacológico , Hipóxia/imunologia , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Fatores Imunológicos/uso terapêutico , Ativação Linfocitária , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/imunologia , Transdução de Sinais , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia
9.
Clin Exp Rheumatol ; 33(1): 34-43, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25535886

RESUMO

OBJECTIVES: The circadian rhythm of clinical symptoms in rheumatoid arthritis (RA) has been primarily attributed to circadian variations in humoral factors and hormones. In this study, we investigated circadian rhythms of cellular immunity in RA (CiRA study). METHODS: Peripheral blood of female postmenopausal patients with active RA (DAS 28 ≥ 4.2) (n=5) and female postmenopausal non-RA controls (n=5) was collected every 2 hours for 24 hours and analysed by flow cytometry, cytokine multiplex suspension array and quantitative RT-PCR of clock gene expression in isolated CD14+ monocytes. Endogenous circadian rhythms of macrophages were investigated by BMAL1-luciferase bioluminescence. Significance of circadian rhythms was tested by Cosinor analysis. RESULTS: We found (i) circadian rhythms in the relative frequency of peripheral blood cell populations that were present in postmenopausal non-RA controls but absent in patients with active RA, (ii) circadian rhythms that were absent in non-RA controls but present in patients with RA and (iii) circadian rhythms that were present in both groups but with differences in peak phase or amplitude or amplitude/magnitude. The circadian rhythm in expression of the clock genes PER2 and PER3 in CD14+ monocytes was lost in patients with RA. The amplitude of BMAL1-luciferase bioluminescence tended to be lower in patients with RA than in non-RA controls. CONCLUSIONS: We conclude that (i) in RA some immune cell populations lose their normal circadian rhythms whereas others establish new 'inflammatory' circadian rhythms and (ii) these findings provide a good basis for further identifying pathophysiological aspects of RA chronobiology with potential therapeutic implications.


Assuntos
Artrite Reumatoide/imunologia , Ritmo Circadiano , Imunidade Celular , Macrófagos/imunologia , Monócitos/imunologia , Adaptação Fisiológica , Idoso , Artrite Reumatoide/sangue , Artrite Reumatoide/genética , Artrite Reumatoide/fisiopatologia , Biomarcadores/sangue , Estudos de Casos e Controles , Ritmo Circadiano/genética , Citocinas/sangue , Feminino , Humanos , Imunidade Humoral , Mediadores da Inflamação/sangue , Receptores de Lipopolissacarídeos/sangue , Macrófagos/metabolismo , Pessoa de Meia-Idade , Monócitos/metabolismo , Proteínas Circadianas Period/genética , Projetos Piloto , Pós-Menopausa/imunologia , RNA Mensageiro/sangue , Fatores de Tempo
10.
Ann N Y Acad Sci ; 1318: 1-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24611742

RESUMO

Glucocorticoids (GCs) are routinely used to treat a wide range of rheumatic and other inflammatory diseases. GCs are steroidal drugs that exert their strong anti-inflammatory and immunosuppressive effects via genomic mechanisms, primarily by signaling through the cytosolic glucocorticoid receptor. In addition, rapid, nongenomic responses following GC treatment have been reported to involve signaling via the membrane-bound glucocorticoid receptor (mGR). Since an important clinical role of this receptor has been proposed, investigations regarding the origin and function of the mGR are currently performed in order to understand rapid GC signaling and to optimize treatment strategies with GCs. Here, we summarize the current knowledge on the mGR and compare these findings to results obtained for other membrane-bound receptors, such as membrane forms of the estrogen and progesterone receptors.


Assuntos
Proteínas de Membrana/fisiologia , Receptores de Glucocorticoides/fisiologia , Humanos , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/fisiologia
11.
J Leukoc Biol ; 95(2): 233-41, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24168857

RESUMO

Hypoxia is an important feature of inflamed tissue, such as the RA joint. Activated monocytes/macrophages and endothelial cells play a pivotal role in the pathogenesis of RA, implicated in the mechanism of inflammation and erosion. During development, myeloid progenitor cells sequentially give rise to monoblasts, promonocytes, and monocytes that are released from the bone marrow into the bloodstream. After extravasation, monocytes differentiate into long-lived, tissue-specific macrophages or DCs. The effect of different oxygen concentrations experienced by these cells during maturation represents a novel aspect of this developmental process. In inflamed joint tissue, the microvascular architecture is highly dysregulated; thus, efficiency of oxygen supply to the synovium is poor. Therefore, invading cells must adapt instantaneously to changes in the oxygen level of the microenvironment. Angiogenesis is an early event in the inflammatory joint, which is important in enabling activated monocytes to enter via endothelial cells by active recruitment to expand the synovium into a "pannus", resulting in cartilage degradation and bone destruction. The increased metabolic turnover of the expanding synovial pannus outpaces the dysfunctional vascular supply, resulting in hypoxia. The abnormal bioenergetics of the microenvironment further promotes synovial cell invasiveness. In RA, joint hypoxia represents a potential threat to cell function and survival. Notably, oxygen availability is a crucial parameter in the cellular energy metabolism, itself an important factor in determining the function of immune cells.


Assuntos
Hipóxia/patologia , Macrófagos/patologia , Monócitos/patologia , Oxigênio/farmacologia , Animais , Artrite Reumatoide/patologia , Artrite Reumatoide/fisiopatologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Humanos , Hipóxia/sangue , Hipóxia/fisiopatologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos
12.
Bone ; 55(1): 93-101, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23571050

RESUMO

Radiation of the hip is an established method to prevent heterotopic ossification (HO) following total hip arthroplasty (THA) but the precise mechanism is unclear. As inflammatory processes are suggested to be involved in the pathogenesis of HO, we hypothesized that the preoperative irradiation impacts local immune components. Therefore, we quantified immune cell populations and cytokines in hematomas resulting from the transection of the femur in two groups of patients receiving THA: patients irradiated preoperatively (THA-X-hematoma: THA-X-H group) in the hip region (7 Gy) in order to prevent HO and patients who were not irradiated (THA-H group) but were postoperatively treated with non-steroidal anti-inflammatory drugs (NSAIDs). Radiation resulted in significantly increased frequencies of T cells, cytotoxic T cells, NKT cells and CD25+CD127- Treg cells, whereas the number of naive CD45RA-expressing cytotoxic T cells was reduced. These results indicate differential immune cell activation, corroborated by our findings of significantly higher concentrations of pro-inflammatory cytokines (e.g., IL-6, IFNγ) and chemokines (e.g., MCP-1, RANTES) in the THA-X-H group as compared to THA-H group. In contrast, the concentration of the angiogenic VEGF was significantly suppressed in the THA-X-H group. We conclude that preoperative irradiation results in significant changes in immune cell composition and cytokine secretion in THA-hematomas, establishing a specific - rather proinflammatory - milieu. This increase of inflammatory activity together with the observed suppression in VEGF secretion may contribute to the prevention of HO.


Assuntos
Artroplastia de Quadril/efeitos adversos , Quadril/efeitos da radiação , Inflamação/patologia , Ossificação Heterotópica/etiologia , Ossificação Heterotópica/prevenção & controle , Cuidados Pré-Operatórios , Anti-Inflamatórios não Esteroides/uso terapêutico , Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Quimiocinas/sangue , Citometria de Fluxo , Hematoma/etiologia , Hematoma/imunologia , Hematoma/patologia , Humanos , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Contagem de Linfócitos , Modelos Biológicos , Células T Matadoras Naturais/imunologia , Ossificação Heterotópica/sangue , Ossificação Heterotópica/tratamento farmacológico , Cuidados Pós-Operatórios , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/sangue
13.
Eur J Immunol ; 43(6): 1588-97, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23519896

RESUMO

Inflamed areas are characterized by infiltration of immune cells, local hypoxia and alterations of cellular redox states. We investigated the impact of hypoxia on survival, proliferation, cytokine secretion, intracellular energy and redox state of human CD4(+) T cells. We found that pathophysiological hypoxia (<2% O2 ) significantly decreased CD4(+) T-cell survival after mitogenic stimulation. This effect was not due to an increased caspase-3/7-mediated apoptosis or adenosine-5'-triphosphate (ATP) consumption/depletion. However, the ability of stimulated T cells to proliferate was reduced under hypoxic conditions, despite increased expression of CD25. Pathophysiological hypoxia was also found to modify intracellular ROS (iROS) levels in stimulated T cells over time as compared with levels found in normoxia. Physiological hypoxia (5% O2 ) did not decrease CD4(+) T-cell survival and proliferation or modify iROS levels as compared with normoxia. We conclude that pathophysiological hypoxia affects T-cell proliferation and viability via disturbed IL-2R signalling downstream of STAT5a phosphorylation, but not as a result of impaired cellular energy homeostasis. We suggest iROS links early events in T-cell stimulation to the inhibition of the lymphoproliferative response under pathophysiological hypoxic conditions. The level of iROS may therefore act as a mediator of immune functions leading to down-regulation of long-term T-cell activity in inflamed tissues.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Hipóxia Celular/imunologia , Sobrevivência Celular , Células Cultivadas , Humanos , Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
14.
PLoS One ; 7(9): e46483, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029528

RESUMO

BACKGROUND: Bone fracture initiates a series of cellular and molecular events including the expression of hypoxia-inducible factor (HIF)-1. HIF-1 is known to facilitate recruitment and differentiation of multipotent human mesenchymal stromal cells (hMSC). Therefore, we analyzed the impact of hypoxia and HIF-1 on the competitive differentiation potential of hMSCs towards adipogenic and osteogenic lineages. METHODOLOGY/PRINCIPAL FINDINGS: Bone marrow derived primary hMSCs cultured for 2 weeks either under normoxic (app. 18% O(2)) or hypoxic (less than 2% O(2)) conditions were analyzed for the expression of MSC surface markers and for expression of the genes HIF1A, VEGFA, LDHA, PGK1, and GLUT1. Using conditioned medium, adipogenic or osteogenic differentiation as verified by Oil-Red-O or von-Kossa staining was induced in hMSCs under either normoxic or hypoxic conditions. The expression of HIF1A and VEGFA was measured by qPCR. A knockdown of HIF-1α by lentiviral transduction was performed, and the ability of the transduced hMSCs to differentiate into adipogenic and osteogenic lineages was analyzed. Hypoxia induced HIF-1α and HIF-1 target gene expression, but did not alter MSC phenotype or surface marker expression. Hypoxia (i) suppressed adipogenesis and associated HIF1A and PPARG gene expression in hMSCs and (ii) enhanced osteogenesis and associated HIF1A and RUNX2 gene expression. shRNA-mediated knockdown of HIF-1α enhanced adipogenesis under both normoxia and hypoxia, and suppressed hypoxia-induced osteogenesis. CONCLUSIONS/SIGNIFICANCE: Hypoxia promotes osteogenesis but suppresses adipogenesis of human MSCs in a competitive and HIF-1-dependent manner. We therefore conclude that the effects of hypoxia are crucial for effective bone healing, which may potentially lead to the development of novel therapeutic approaches.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Células-Tronco Mesenquimais/fisiologia , Adipogenia , Antígenos CD/metabolismo , Hipóxia Celular , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , PPAR gama/genética , PPAR gama/metabolismo , Fenótipo , Interferência de RNA , Ativação Transcricional , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Arthritis Res Ther ; 14(4): R181, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22870988

RESUMO

INTRODUCTION: Inflammatory arthritis is a progressive disease with chronic inflammation of joints, which is mainly characterized by the infiltration of immune cells and synovial hyperproliferation. Monocytes migrate towards inflamed areas and differentiate into macrophages. In inflamed tissues, much lower oxygen levels (hypoxia) are present in comparison to the peripheral blood. Hence, a metabolic adaptation process must take place. Other studies suggest that Hypoxia Inducible Factor 1-alpha (HIF-1α) may regulate this process, but the mechanism involved for human monocytes is not yet clear. To address this issue, we analyzed the expression and function of HIF-1α in monocytes and macrophages, but also considered alternative pathways involving nuclear factor of kappa light polypeptide gene enhancer in B-cells (NFκB). METHODS: Isolated human CD14⁺ monocytes were incubated under normoxia and hypoxia conditions with or without phorbol 12-myristate 13-acetate (PMA) stimulation, respectively. Nuclear and cytosolic fractions were prepared in order to detect HIF-1α and NFκB by immunoblot. For the experiments with macrophages, primary human monocytes were differentiated into human monocyte derived macrophages (hMDM) using human macrophage colony-stimulating factor (hM-CSF). The effects of normoxia and hypoxia on gene expression were compared between monocytes and hMDMs using quantitative PCR (quantitative polymerase chain reaction). RESULTS: We demonstrate, using primary human monocytes and hMDM, that the localization of transcription factor HIF-1α during the differentiation process is shifted from the cytosol (in monocytes) into the nucleus (in macrophages), apparently as an adaptation to a low oxygen environment. For this localization change, protein kinase C alpha/beta 1 (PKC-α/ß1) plays an important role. In monocytes, it is NFκB1, and not HIF-1α, which is of central importance for the expression of hypoxia-adjusted genes. CONCLUSIONS: These data demonstrate that during differentiation of monocytes into macrophages, crucial cellular adaptation mechanisms are decisively changed.


Assuntos
Adaptação Fisiológica/fisiologia , Macrófagos/metabolismo , Monócitos/metabolismo , Hipóxia Celular/fisiologia , Células Cultivadas , Células HL-60 , Humanos , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA