Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 29(8): 1450-1465, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35031770

RESUMO

Heme is an erythrocyte-derived toxin that drives disease progression in hemolytic anemias, such as sickle cell disease. During hemolysis, specialized bone marrow-derived macrophages with a high heme-metabolism capacity orchestrate disease adaptation by removing damaged erythrocytes and heme-protein complexes from the blood and supporting iron recycling for erythropoiesis. Since chronic heme-stress is noxious for macrophages, erythrophagocytes in the spleen are continuously replenished from bone marrow-derived progenitors. Here, we hypothesized that adaptation to heme stress progressively shifts differentiation trajectories of bone marrow progenitors to expand the capacity of heme-handling monocyte-derived macrophages at the expense of the homeostatic generation of dendritic cells, which emerge from shared myeloid precursors. This heme-induced redirection of differentiation trajectories may contribute to hemolysis-induced secondary immunodeficiency. We performed single-cell RNA-sequencing with directional RNA velocity analysis of GM-CSF-supplemented mouse bone marrow cultures to assess myeloid differentiation under heme stress. We found that heme-activated NRF2 signaling shifted the differentiation of bone marrow cells towards antioxidant, iron-recycling macrophages, suppressing the generation of dendritic cells in heme-exposed bone marrow cultures. Heme eliminated the capacity of GM-CSF-supplemented bone marrow cultures to activate antigen-specific CD4 T cells. The generation of functionally competent dendritic cells was restored by NRF2 loss. The heme-induced phenotype of macrophage expansion with concurrent dendritic cell depletion was reproduced in hemolytic mice with sickle cell disease and spherocytosis and associated with reduced dendritic cell functions in the spleen. Our data provide a novel mechanistic underpinning of hemolytic stress as a driver of hyposplenism-related secondary immunodeficiency.


Assuntos
Anemia Falciforme , Células da Medula Óssea , Células Dendríticas , Heme , Macrófagos , Fator 2 Relacionado a NF-E2 , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Células Dendríticas/citologia , Eritropoese , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Hemólise , Ferro , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , RNA , Baço
2.
Front Immunol ; 12: 680855, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054870

RESUMO

Clearance of red blood cells and hemoproteins is a key metabolic function of macrophages during hemolytic disorders and following tissue injury. Through this archetypical phagocytic function, heme is detoxified and iron is recycled to support erythropoiesis. Reciprocal interaction of heme metabolism and inflammatory macrophage functions may modify disease outcomes in a broad range of clinical conditions. We hypothesized that acute hemolysis and heme induce acute anti-inflammatory signals in liver macrophages. Using a macrophage-driven model of sterile liver inflammation, we showed that phenylhydrazine (PHZ)-mediated acute erythrophagocytosis blocked the anti-CD40 antibody-induced pathway of macrophage activation. This process attenuated the inflammatory cytokine release syndrome and necrotizing hepatitis induced by anti-CD40 antibody treatment of mice. We further established that administration of heme-albumin complexes specifically delivered heme to liver macrophages and replicated the anti-inflammatory effect of hemolysis. The anti-inflammatory heme-signal was induced in macrophages by an increased intracellular concentration of the porphyrin independently of iron. Overall, our work suggests that induction of heme-signaling strongly suppresses inflammatory macrophage function, providing protection against sterile liver inflammation.


Assuntos
Anticorpos/imunologia , Antígenos CD40/antagonistas & inibidores , Antígenos CD40/imunologia , Hemólise/imunologia , Hepatite/etiologia , Albuminas/metabolismo , Animais , Anticorpos/efeitos adversos , Biópsia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Eritrócitos/patologia , Perfilação da Expressão Gênica , Heme/metabolismo , Hepatite/metabolismo , Hepatite/patologia , Ferro/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Fenil-Hidrazinas/efeitos adversos , Porfirinas/metabolismo , Ligação Proteica
3.
J Clin Invest ; 130(10): 5576-5590, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32663195

RESUMO

During hemolysis, macrophages in the liver phagocytose damaged erythrocytes to prevent the toxic effects of cell-free hemoglobin and heme. It remains unclear how this homeostatic process modulates phagocyte functions in inflammatory diseases. Using a genetic mouse model of spherocytosis and single-cell RNA sequencing, we found that erythrophagocytosis skewed liver macrophages into an antiinflammatory phenotype that we defined as MarcohiHmoxhiMHC class IIlo erythrophagocytes. This phenotype transformation profoundly mitigated disease expression in a model of an anti-CD40-induced hyperinflammatory syndrome with necrotic hepatitis and in a nonalcoholic steatohepatitis model, representing 2 macrophage-driven sterile inflammatory diseases. We reproduced the antiinflammatory erythrophagocyte transformation in vitro by heme exposure of mouse and human macrophages, yielding a distinctive transcriptional signature that segregated heme-polarized from M1- and M2-polarized cells. Mapping transposase-accessible chromatin in single cells by sequencing defined the transcription factor NFE2L2/NRF2 as a critical driver of erythrophagocytes, and Nfe2l2/Nrf2 deficiency restored heme-suppressed inflammation. Our findings point to a pathway that regulates macrophage functions to link erythrocyte homeostasis with innate immunity.


Assuntos
Hemólise/fisiologia , Fígado/citologia , Fígado/fisiologia , Macrófagos/citologia , Macrófagos/fisiologia , Fagócitos/citologia , Fagócitos/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Heme/metabolismo , Humanos , Técnicas In Vitro , Inflamação/prevenção & controle , Macrófagos/classificação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Fagócitos/classificação , Fagocitose/fisiologia , Fenótipo , RNA-Seq , Análise de Célula Única , Esferocitose Hereditária/genética , Esferocitose Hereditária/patologia , Esferocitose Hereditária/fisiopatologia
4.
Blood Adv ; 4(12): 2751-2761, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32559293

RESUMO

Hemophagocytic syndromes comprise a cluster of hyperinflammatory disorders, including hemophagocytic lymphohistiocytosis and macrophage activation syndrome. Overwhelming macrophage activation has long been considered a final common pathway in the pathophysiology of hemophagocytic syndromes leading to the characteristic cytokine storm, laboratory abnormalities, and organ injuries that define the clinical spectrum of the disease. So far, it is unknown whether primary macrophage activation alone can induce the disease phenotype. In this study, we established a novel mouse model of a hemophagocytic syndrome by treating mice with an agonistic anti-CD40 antibody (Ab). The response in wild-type mice is characterized by a cytokine storm, associated with hyperferritinemia, high soluble CD25, erythrophagocytosis, secondary endothelial activation with multiple organ vaso-occlusion, necrotizing hepatitis, and variable cytopenias. The disease is dependent on a tumor necrosis factor-α-interferon-γ-driven amplification loop. After macrophage depletion with clodronate liposomes or in mice with a macrophage-selective deletion of the CD40 gene (CD40flox/flox/LysMCre), the disease was abolished. These data provide a new preclinical model of a hemophagocytic syndrome and reinforce the key pathophysiological role of macrophages.


Assuntos
Linfo-Histiocitose Hemofagocítica , Síndrome de Ativação Macrofágica , Animais , Interferon gama , Ativação de Macrófagos , Macrófagos , Camundongos
5.
Pharmacol Res Perspect ; 6(2): e00392, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29610666

RESUMO

Activation of the innate immune system by free heme has been proposed as one of the principal consequences of cell-free hemoglobin (Hb) exposure. Nonetheless, in the absence of infection, heme exposures within a hematoma, during hemolysis, or upon systemic administration of Hb (eg, as a Hb-based oxygen carrier) are typically not accompanied by uncontrolled inflammation, challenging the assumption that heme is a major proinflammatory mediator in vivo. Because of its hydrophobic nature, heme liberated from oxidized hemoglobin is rapidly transferred to alternative protein-binding sites (eg, albumin) or to hydrophobic lipid compartments minimizing protein-free heme under in vivo equilibrium conditions. We demonstrate that the capacity of heme to activate human neutrophil granulocytes strictly depends on the availability of non protein-associated heme. In human endothelial cells as well as in mouse macrophage cell cultures and in mouse models of local and systemic heme exposure, protein-associated heme or Hb do not induce inflammatory gene expression over a broad range of exposure conditions. Only experiments in protein-free culture medium demonstrated a weak capacity of heme-solutions to induce toll-like receptor-(TLR4) dependent TNF-alpha expression in macrophages. Our data suggests that the equilibrium-state of free and protein-associated heme critically determines the proinflammatory capacity of the metallo-porphyrin. Based on these data it appears unlikely that inflammation-promoting equilibrium conditions could ever occur in vivo.


Assuntos
Heme/fisiologia , Inflamação , Macrófagos/imunologia , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/imunologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Relação Dose-Resposta a Droga , Perfilação da Expressão Gênica , Heme/farmacologia , Heme Oxigenase-1/metabolismo , Hemólise/efeitos dos fármacos , Hemólise/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/genética , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Transcriptoma/imunologia
6.
BMC Biotechnol ; 18(1): 15, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29544494

RESUMO

BACKGROUND: Preclinical studies have evaluated haptoglobin (Hp) polymers from pooled human plasma as a therapeutic protein to attenuate toxic effects of cell-free hemoglobin (Hb). Proof of concept studies have demonstrated efficacy of Hp in hemolysis associated with transfusion and sickle cell anemia. However, phenotype-specific Hp products might be desirable to exploit phenotype specific activities of Hp 1-1 versus Hp 2-2, offering opportunities for recombinant therapeutics. Prohaptoglobin (proHp) is the primary translation product of the Hp mRNA. ProHp is proteolytically cleaved by complement C1r subcomponent-like protein (C1r-LP) in the endoplasmic reticulum. Two main allelic Hp variants, HP1 and HP2 exist. The larger HP2 is considered to be the ancestor variant of all human Hp alleles and is characterized by an α2-chain, which contains an extra cysteine residue that pairs with additional α-chains generating multimers with molecular weights of 200-900 kDa. The two human HP1 alleles (HP1F and HP1S) differ by a two-amino-acid substitution polymorphism within the α-chain and are derived from HP2 by recurring exon deletions. RESULTS: In the present study, we describe a process for the production of recombinant phenotype specific Hp polymers in mammalian FS293F cells. This approach demonstrates that efficient expression of mature and fully functional protein products requires co-expression of active C1r-LP. The functional characterization of our proteins, which included monomer/polymer distribution, binding affinities as well as NO-sparing and antioxidant functions, demonstrated that C1r-LP-processed recombinant Hp demonstrates equal protective functions as plasma derived Hp in vitro as well as in animal studies. CONCLUSIONS: We present a recombinant production process for fully functional phenotype-specific Hp therapeutics. The proposed process could accelerate the development of Hb scavengers to treat patients with cell-free Hb associated disease states, such as sickle cell disease and other hemolytic conditions.


Assuntos
Haptoglobinas/genética , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Engenharia de Proteínas/métodos , Serina Endopeptidases/genética , Animais , Vasos Coronários/efeitos dos fármacos , Cobaias , Haptoglobinas/farmacologia , Heme/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Óxido Nítrico/metabolismo , Fenótipo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Serina Endopeptidases/metabolismo , Suínos
7.
Oxid Med Cell Longev ; 2013: 870472, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840921

RESUMO

The CD163 scavenger receptor pathway for Hb:Hp complexes is an essential mechanism of protection against the toxicity of extracellular hemoglobin (Hb), which can accumulate in the vasculature and within tissues during hemolysis. Chloroquine is a lysosomotropic agent, which has been extensively used as an antimalarial drug in the past, before parasite resistance started to limit its efficacy in most parts of the world. More recent use of chloroquine is related to its immunomodulatory activity in patients with autoimmune diseases, which may also involve hemolytic disease components. In this study we examined the effects of chloroquine on the human Hb clearance pathway. For this purpose we developed a new mass-spectrometry-based method to specifically quantify intracellular Hb peptides within the endosomal-lysosomal compartment by single reaction monitoring (SRM). We found that chloroquine exposure impairs trafficking of Hb:Hp complexes through the endosomal-lysosomal compartment after internalization by CD163. Relative quantification of intracellular Hb peptides by SRM confirmed that chloroquine blocked cellular Hb:Hp catabolism. This effect suppressed the cellular heme-oxygenase-1 (HO-1) response and shifted macrophage iron homeostasis towards inappropriately high expression of the transferrin receptor with concurrent inhibition of ferroportin expression. A functional deficiency of Hb detoxification and heme-iron recycling may therefore be an adverse consequence of chloroquine treatment during hemolysis.


Assuntos
Cloroquina/farmacologia , Endocitose/efeitos dos fármacos , Heme/metabolismo , Hemoglobinas/metabolismo , Homeostase/efeitos dos fármacos , Ferro/metabolismo , Macrófagos/metabolismo , Sequência de Aminoácidos , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antimaláricos/farmacologia , Células HEK293 , Haptoglobinas/metabolismo , Heme Oxigenase-1/metabolismo , Hemoglobinas/química , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Macrófagos/efeitos dos fármacos , Dados de Sequência Molecular , Peptídeos/metabolismo , Transporte Proteico/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo
8.
Blood ; 116(24): 5347-56, 2010 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-20739658

RESUMO

Glucocorticoids are used extensively to treat autoimmune hemolytic anemias. Some beneficial effects of glucocorticoid pulse therapy have also been reported in sickle cell disease and paroxysmal nocturnal hemoglobinuria. Based on established concepts of hemoglobin (Hb) toxicity and physiologic Hb scavenger systems, we evaluated whether glucocorticoids could support an adaptive response to extracellular Hb independently of their immunosuppressive activities. Using global proteome and transcriptome analysis with mass-spectrometry (isobaric tag for relative and absolute quantitation and liquid chromatography-mass spectrometry) and gene-array experiments, we found that glucocorticoid treatment in vitro and in patients on glucocorticoid-pulse therapy polarized monocytes into a M2/alternatively activated phenotype with high Hb-scavenger receptor (CD163) expression and enhanced Hb-clearance and detoxification capability. Monocytes concurrently exposed to the interactive activity of glucocorticoids and extracellular Hb were characterized by high expression of a group of antioxidant enzymes known to be regulated by the conserved oxidative response transcription factor nuclear factor E2-related factor. Further, suppressed transferrin receptor, together with high ferroportin expression, pointed to a shift in iron homeostasis directed toward an increased cellular export of heme-derived iron. Therefore, stimulating Hb-endocytosis by CD163 and enhancing antioxidative homeostasis and iron recycling may be an essential activity of glucocorticoids that helps alleviate the adverse effects of extracellular Hb.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Glucocorticoides/farmacologia , Monócitos/efeitos dos fármacos , Antígenos CD/farmacologia , Antígenos de Diferenciação Mielomonocítica/farmacologia , Antioxidantes , Endocitose , Glucocorticoides/uso terapêutico , Heme/metabolismo , Hemoglobinas/metabolismo , Homeostase , Humanos , Ferro/metabolismo , Monócitos/citologia , Fenótipo , Receptores de Superfície Celular
9.
J Leukoc Biol ; 83(2): 325-33, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17947394

RESUMO

Macrophages constitute the major cellular compartment for hemoglobin (Hb) degradation and subsequent recycling of heme-iron to erythropoiesis. Dysregulation of macrophage iron and heme metabolism is a major pathophysiologic determinant of anemia of chronic disease. In this study, we show that the heme transporter heme carrier protein 1 (HCP-1) is expressed in human macrophages. Within early endosomes, HCP-1 colocalizes with endocytosed Hb-haptoglobin (Hp) complexes, which are taken up via the CD163 scavenger receptor pathway. Hb-Hp passes the divalent metal transporter 1B/HCP-1-positive endosomal compartment on its route from the cell surface to lysosomes. HCP-1 mRNA and protein expression are down-regulated by stimulation of macrophages with various TLR agonists and IFN-gamma. The profound suppression of HCP-1 expression by inflammatory macrophage activation parallels the regulation of the iron exporter ferroportin. In contrast, dexamethasone enhanced HCP-1 expression significantly. Given the spatial relationship, we propose that the Hb scavenger receptor CD163 and HCP-1 constitute a linked pathway for Hb catabolism and heme-iron recycling in human macrophages.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Receptores de Superfície Celular/metabolismo , Antígenos CD/genética , Antígenos CD/isolamento & purificação , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/isolamento & purificação , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Dexametasona/farmacologia , Endocitose , Endossomos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Heme/metabolismo , Humanos , Interferon gama/farmacologia , Ferro/metabolismo , Rim , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/isolamento & purificação , Modelos Biológicos , Mapeamento de Interação de Proteínas , Transportador de Folato Acoplado a Próton , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Receptores Toll-Like/agonistas
10.
Eur J Haematol ; 77(5): 432-6, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17044836

RESUMO

OBJECTIVES: Uncontrolled macrophage activation with hemophagocytosis is a distinctive feature of hemophagocytic syndromes (HPS). We examined whether lympho-histiocytic infiltration of the bone marrow and liver, as well as hemo-/erythrophagocytosis also occurs during sepsis and whether this process could account for the increased production of anti-inflammatory heme-oxygenase (HO-1) products observed during sepsis. METHODS: Hemophagocytosis and expression of CD163, HO-1, ferritin as well as CD8 and granzyme-B were examined in post-mortem bone marrow samples from 28 patients with sepsis and from eight control patients. RESULTS: Comparison of samples from non-septic patients with samples from patients with fatal sepsis revealed that the latter group displayed dense lympho-histiocytic bone marrow infiltration with CD163(+)/HO-1(+)/ferritin(+) macrophages as well as with CD8(+) and granzyme-B(+) T-cells. Hemophagocytosis with prominent phagocytosis of erythroid cells was readily apparent in septic patients, implying that this process is a likely stimulus for the up-regulation of macrophage HO-1 expression. CONCLUSIONS: Lympho-histiocytic activation with hemophagocytosis is a shared pathophysiologic mechanism in HPS and sepsis. Furthermore, the association of hemophagocytosis with an increase in HO-1 expression may indicate a novel role for this apparently futile process as a negative regulator of inflammation.


Assuntos
Regulação Enzimológica da Expressão Gênica , Heme Oxigenase-1/biossíntese , Macrófagos/enzimologia , Fagocitose , Sepse/enzimologia , Regulação para Cima , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Medula Óssea/enzimologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Células Eritroides/enzimologia , Células Eritroides/patologia , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Heme Oxigenase-1/genética , Humanos , Fígado/enzimologia , Fígado/patologia , Macrófagos/patologia , Masculino , Pessoa de Meia-Idade , Fagocitose/genética , Sepse/genética , Sepse/patologia , Regulação para Cima/genética
11.
Circ Res ; 99(9): 943-50, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-17008602

RESUMO

Heme toxicity contributes to the pathogenesis of chronic inflammatory diseases, atherosclerosis, and hemolysis associated vasculopathy. Macrophage clearance of cell free hemoglobin (Hb) is thus an essential homeostatic function of these cells. We examined the transcriptional response of human PBMC derived macrophages to Hb by gene array analysis. The observed noninflammatory macrophage response was characterized by induction of an antioxidative and antiinflammatory gene expression pattern with most prominent induction of the inducible heme oxygenase (HO-1). The metabolically active Hb-CD163-HO-1 pathway resulted in synthesis of ferritin-1 of the antioxidative and antiinflammatory end products linked to heme breakdown by HO-1. This response was mediated by the Hb scavenger receptor CD163 and heme and was not related to Hb mediated depletion of reduced glutathione. In contrast to other cellular responses induced by CD163, there was no role of protein phosphorylation dependent CD163 signaling in the protective macrophage response to Hb. Instead, CD163 acted as an Hb transporter, which undergoes constitutive and ligand independent internalization and recycling between the cell surface and early endosomes. The expression of CD163 and HO-1 in macrophages of neovascularized atherosclerotic lesions suggests that the pathway described herein is active in vivo. Noninflammatory Hb clearance and intimately linked HO-1 expression may provide the long sought-after explanation for the antiinflammatory activity associated with CD163-positive macrophages.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Endocitose , Heme Oxigenase-1/biossíntese , Heme/metabolismo , Hemoglobinas/metabolismo , Macrófagos/metabolismo , Receptores de Superfície Celular/metabolismo , Antígenos CD/análise , Antígenos CD/fisiologia , Antígenos de Diferenciação Mielomonocítica/análise , Antígenos de Diferenciação Mielomonocítica/fisiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Células Cultivadas , Citoproteção , Endossomos/metabolismo , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Glutationa/metabolismo , Heme/fisiologia , Heme Oxigenase-1/genética , Hemoglobinas/farmacologia , Humanos , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Neovascularização Patológica/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/fisiologia , Receptores Depuradores/metabolismo , Transcrição Gênica
12.
Blood ; 107(1): 373-80, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16189277

RESUMO

CD163 mediates the internalization of hemoglobin-haptoglobin (Hb-Hp) complexes by macrophages. Because Hp binding capacity is exhausted during severe hemolysis, an Hp-independent Hb-clearance pathway is presumed to exist. We demonstrate that Hb interacts efficiently with CD163 in the absence of Hp. Not only is Hb internalized into an endosomal compartment by CD163 as a result of active receptor-dependent endocytosis; it also inhibits the uptake of Hb-Hp complexes, suggesting a common receptor-binding site. Free Hb further induces heme oxygenase mRNA expression in CD163+ HEK293 cells, but not in CD163- cells. Additional evidence for Hp-independent Hb-CD163 interaction is provided by the demonstration that CD163 mediates the uptake of alpha alpha-DBBF crosslinked Hb, a chemically modified Hb that forms minimal Hp complexes. Moreover, certain modifications to Hb, such as polymerization or the attachment of specific functional groups (3 lysyl residues) to the beta-Cys93 can reduce or enhance this pathway of uptake. In human macrophages, Hp-complex formation critically enhances Hb uptake at low (1 microg/mL), but not at high (greater than 100 microg/mL), ligand concentrations, lending support for a concentration-dependent biphasic model of macrophage Hb-clearance. These results identify CD163 as a scavenger receptor for native Hb and small-molecular-weight Hb-based blood substitutes after Hp depletion.


Assuntos
Antígenos CD/fisiologia , Antígenos de Diferenciação Mielomonocítica/fisiologia , Haptoglobinas/deficiência , Hemoglobinas/metabolismo , Receptores de Superfície Celular/fisiologia , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Aspirina/análogos & derivados , Substitutos Sanguíneos , Linhagem Celular , Endocitose , Endossomos/metabolismo , Regulação da Expressão Gênica , Heme Oxigenase (Desciclizante)/genética , Hemoglobina A , Hemoglobinas/química , Humanos , Macrófagos , Receptores de Superfície Celular/metabolismo , Receptores Depuradores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA