Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
iScience ; 26(7): 107219, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37529320

ABSTRACT

The vast spectrum of clinical features of COVID-19 keeps challenging scientists and clinicians. Low resistance to infection might result in long-term viral persistence, but the underlying mechanisms remain unclear. Here, we studied the immune response of immunocompetent COVID-19 patients with prolonged SARS-CoV-2 infection by immunophenotyping, cytokine and serological analysis. Despite viral loads and symptoms comparable to regular mildly symptomatic patients, long-term carriers displayed weaker systemic IFN-I responses and fewer circulating pDCs and NK cells at disease onset. Type 1 cytokines remained low, while type-3 cytokines were in turn enhanced. Of interest, we observed no defects in antigen-specific cytotoxic T cell responses, and circulating antibodies displayed higher affinity against different variants of SARS-CoV-2 Spike protein in these patients. The identification of distinct immune responses in long-term carriers adds up to our understanding of essential host protective mechanisms to ensure tissue damage control despite prolonged viral infection.

2.
J Immunol ; 205(10): 2795-2805, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33037139

ABSTRACT

Hemolysis causes an increase of intravascular heme, oxidative damage, and inflammation in which macrophages play a critical role. In these cells, heme can act as a prototypical damage-associated molecular pattern, inducing TLR4-dependent cytokine production through the MyD88 pathway, independently of TRIF. Heme promotes reactive oxygen species (ROS) generation independently of TLR4. ROS and TNF production contribute to heme-induced necroptosis and inflammasome activation; however, the role of ROS in proinflammatory signaling and cytokine production remains unknown. In this study, we demonstrate that heme activates at least three signaling pathways that contribute to a robust MAPK phosphorylation and cytokine expression in mouse macrophages. Although heme did not induce a detectable Myddosome formation, the TLR4/MyD88 axis was important for phosphorylation of p38 and secretion of cytokines. ROS generation and spleen tyrosine kinase (Syk) activation induced by heme were critical for most proinflammatory signaling pathways, as the antioxidant N-acetyl-l-cysteine and a Syk inhibitor differentially blocked heme-induced ROS, MAPK phosphorylation, and cytokine production in macrophages. Early generated mitochondrial ROS induced by heme was Syk dependent, selectively promoted the phosphorylation of ERK1/2 without affecting JNK or p38, and contributed to CXCL1 and TNF production. Finally, lethality caused by sterile hemolysis in mice required TLR4, TNFR1, and mitochondrial ROS, supporting the rationale to target these pathways to mitigate tissue damage of hemolytic disorders.


Subject(s)
Heme/metabolism , Hemolysis/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , Animals , Chemokine CXCL1/metabolism , Disease Models, Animal , Humans , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/immunology , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Syk Kinase/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
An Acad Bras Cienc ; 88(3): 1485-99, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27627069

ABSTRACT

Alphaviruses among the viruses that cause arthritis, consisting in a public health problem worldwide by causing localized outbreaks, as well as large epidemics in humans. Interestingly, while the Old World alphaviruses are arthritogenic, the New World alphaviruses cause encephalitis. One exception is Mayaro virus (MAYV), which circulates exclusively in South America but causes arthralgia and is phylogenetically related to the Old World alphaviruses. Although MAYV-induced arthritis in humans is well documented, the molecular and cellular factors that contribute to its pathogenesis are completely unknown. In this study, we demonstrated for the first time that macrophages, key players in arthritis development, are target cells for MAYV infection, which leads to cell death through apoptosis. We showed that MAYV replication in macrophage induced the expression of TNF, a cytokine that would contribute to pathogenesis of MAYV fever, since TNF promotes an inflammatory profile characteristic of arthritis. We also found a significant increase in the production of reactive oxygen species (ROS) at early times of infection, which coincides with the peak of virus replication and precedes TNF secretion. Treatment of the cells with antioxidant agents just after infection completely abolished TNF secretion, indicating an involvement of ROS in inflammation induced during MAYV infection.


Subject(s)
Alphavirus Infections , Arthritis/virology , Macrophages/virology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Virus Replication , Alphavirus Infections/complications , Humans , South America
4.
Proc Natl Acad Sci U S A ; 111(39): E4110-8, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25225402

ABSTRACT

The increase of extracellular heme is a hallmark of hemolysis or extensive cell damage. Heme has prooxidant, cytotoxic, and inflammatory effects, playing a central role in the pathogenesis of malaria, sepsis, and sickle cell disease. However, the mechanisms by which heme is sensed by innate immune cells contributing to these diseases are not fully characterized. We found that heme, but not porphyrins without iron, activated LPS-primed macrophages promoting the processing of IL-1ß dependent on nucleotide-binding domain and leucine rich repeat containing family, pyrin domain containing 3 (NLRP3). The activation of NLRP3 by heme required spleen tyrosine kinase, NADPH oxidase-2, mitochondrial reactive oxygen species, and K(+) efflux, whereas it was independent of heme internalization, lysosomal damage, ATP release, the purinergic receptor P2X7, and cell death. Importantly, our results indicated the participation of macrophages, NLRP3 inflammasome components, and IL-1R in the lethality caused by sterile hemolysis. Thus, understanding the molecular pathways affected by heme in innate immune cells might prove useful to identify new therapeutic targets for diseases that have heme release.


Subject(s)
Heme/metabolism , Hemolysis/physiology , Inflammasomes/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/metabolism , Heme/chemistry , Heme/immunology , Hemolysis/immunology , Humans , Inflammasomes/immunology , Interleukin-1beta/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NADPH Oxidase 2 , NADPH Oxidases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Potassium/metabolism , Protoporphyrins/chemistry , Protoporphyrins/metabolism , Reactive Oxygen Species/metabolism
5.
J Clin Invest ; 122(7): 2531-42, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22728935

ABSTRACT

Oxidative damage contributes to microbe elimination during macrophage respiratory burst. Nuclear factor, erythroid-derived 2, like 2 (NRF2) orchestrates antioxidant defenses, including the expression of heme-oxygenase-1 (HO-1). Unexpectedly, the activation of NRF2 and HO-1 reduces infection by a number of pathogens, although the mechanism responsible for this effect is largely unknown. We studied Trypanosoma cruzi infection in mice in which NRF2/HO-1 was induced with cobalt protoporphyrin (CoPP). CoPP reduced parasitemia and tissue parasitism, while an inhibitor of HO-1 activity increased T. cruzi parasitemia in blood. CoPP-induced effects did not depend on the adaptive immunity, nor were parasites directly targeted. We also found that CoPP reduced macrophage parasitism, which depended on NRF2 expression but not on classical mechanisms such as apoptosis of infected cells, induction of type I IFN, or NO. We found that exogenous expression of NRF2 or HO-1 also reduced macrophage parasitism. Several antioxidants, including NRF2 activators, reduced macrophage parasite burden, while pro-oxidants promoted it. Reducing the intracellular labile iron pool decreased parasitism, and antioxidants increased the expression of ferritin and ferroportin in infected macrophages. Ferrous sulfate reversed the CoPP-induced decrease in macrophage parasite burden and, given in vivo, reversed their protective effects. Our results indicate that oxidative stress contributes to parasite persistence in host tissues and open a new avenue for the development of anti-T. cruzi drugs.


Subject(s)
Chagas Disease/parasitology , Oxidative Stress , Parasitemia/parasitology , Trypanosoma cruzi/physiology , Animals , Antioxidants/metabolism , Antiparasitic Agents/pharmacology , Antiparasitic Agents/therapeutic use , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Chagas Disease/drug therapy , Ferritins/genetics , Ferritins/metabolism , Ferrous Compounds/pharmacology , Heart/parasitology , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Host-Parasite Interactions , Iron/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/parasitology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Parasitemia/drug therapy , Protoporphyrins/pharmacology , Protoporphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Receptors, Immunologic/metabolism , Respiratory Burst , Trypanosoma cruzi/drug effects
6.
Blood ; 119(10): 2368-75, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22262768

ABSTRACT

Diseases that cause hemolysis or myonecrosis lead to the leakage of large amounts of heme proteins. Free heme has proinflammatory and cytotoxic effects. Heme induces TLR4-dependent production of tumor necrosis factor (TNF), whereas heme cytotoxicity has been attributed to its ability to intercalate into cell membranes and cause oxidative stress. We show that heme caused early macrophage death characterized by the loss of plasma membrane integrity and morphologic features resembling necrosis. Heme-induced cell death required TNFR1 and TLR4/MyD88-dependent TNF production. Addition of TNF to Tlr4(-/-) or to Myd88(-/-) macrophages restored heme-induced cell death. The use of necrostatin-1, a selective inhibitor of receptor-interacting protein 1 (RIP1, also known as RIPK1), or cells deficient in Rip1 or Rip3 revealed a critical role for RIP proteins in heme-induced cell death. Serum, antioxidants, iron chelation, or inhibition of c-Jun N-terminal kinase (JNK) ameliorated heme-induced oxidative burst and blocked macrophage cell death. Macrophages from heme oxygenase-1 deficient mice (Hmox1(-/-)) had increased oxidative stress and were more sensitive to heme. Taken together, these results revealed that heme induces macrophage necrosis through 2 synergistic mechanisms: TLR4/Myd88-dependent expression of TNF and TLR4-independent generation of ROS.


Subject(s)
Heme/pharmacology , Macrophages/drug effects , Reactive Oxygen Species/metabolism , Tumor Necrosis Factors/metabolism , Animals , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Imidazoles/pharmacology , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/cytology , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NIH 3T3 Cells , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factors/pharmacology
7.
J Leukoc Biol ; 85(5): 855-61, 2009 May.
Article in English | MEDLINE | ID: mdl-19188484

ABSTRACT

Deposition of immune complexes (IC) triggers Fc gamma R-dependent inflammation, leading to tissue damage in rheumatoid arthritis, systemic lupus erythematous, immune glomerulonephritis, and several immune vasculitides. Evidences support a role for macrophage migration inhibitory factor (MIF) in a number of inflammatory diseases, but the triggering of its secretion and its physiopathological role upon IC deposition remain elusive. Herein, we show that human macrophages secreted MIF after IC recognition, which in turn controlled the secretion of TNF. Macrophages from Mif-/- mice produced smaller amounts of TNF when stimulated with IgG-opsonized erythrocytes than wild-type (WT) cells. Using passive reverse Arthus reaction in the peritoneum and lungs as a model for IC-induced inflammation, we demonstrated that Mif-/- mice had a milder response, observed by reduced neutrophil recruitment, vascular leakage, and secretion of TNF, MIP-2, and keratinocyte-derived chemokine compared with WT controls. Adoptive transfer of alveolar macrophages from WT to Mif-/- mice rescued pulmonary neutrophil recruitment and TNF production upon passive reverse Arthus reaction. Our study indicates that Arthus inflammatory reaction is largely dependent on MIF and poses macrophages as a source of the MIF released upon IC recognition. These results give experimental support to the proposition that blockade of MIF might constitute an adjunctive, therapeutic approach to IC disease.


Subject(s)
Arthus Reaction/immunology , Inflammation/immunology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Macrophages, Alveolar/immunology , Tumor Necrosis Factors/biosynthesis , Adoptive Transfer , Animals , Antigen-Antibody Complex/immunology , Cells, Cultured , Female , Humans , Inflammation/pathology , Lung/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, IgG/immunology
8.
J Biol Chem ; 282(33): 24430-6, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17581818

ABSTRACT

Hemolysis or extensive cell damage can lead to high concentrations of free heme, causing oxidative stress and inflammation. Considering that heme induces neutrophil chemotaxis, we hypothesize that heme activates a G protein-coupled receptor. Here we show that similar to heme, several heme analogs were able to induce neutrophil migration in vitro and in vivo. Mesoporphyrins, molecules lacking the vinyl groups in their rings, were not chemotactic for neutrophils and selectively inhibited heme-induced migration. Moreover, migration of neutrophils induced by heme was abolished by pretreatment with pertussis toxin, an inhibitor of Galpha inhibitory protein, and with inhibitors of phosphoinositide 3-kinase, phospholipase Cbeta, mitogen-activated protein kinases, or Rho kinase. The induction of reactive oxygen species by heme was dependent of Galpha inhibitory protein and phosphoinositide 3-kinase and partially dependent of phospholipase Cbeta, protein kinase C, mitogen-activated protein kinases, and Rho kinase. Together, our results indicate that heme activates neutrophils through signaling pathways that are characteristic of chemoattractant molecules and suggest that mesoporphyrins might prove valuable in the treatment of the inflammatory consequences of hemorrhagic and hemolytic disorders.


Subject(s)
Chemotaxis , Heme/physiology , Neutrophils/physiology , Reactive Oxygen Species/metabolism , Receptors, Chemokine , Signal Transduction , Animals , Humans , Mice , Mice, Inbred C57BL , Neutrophil Infiltration
9.
Toxicon ; 50(2): 270-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17537475

ABSTRACT

Snake venom is a complex mixture containing diverse protein components with different structures and functions that are used for prey immobilization and death. Snake venoms from the family Viperidae cause pronounced local and systemic effects, such as pain, edema, hemorrhage and necrosis. Here, we investigated the enzymatic and biological activities of venoms from two Amazonian snakes, Bothriopsis bilineata and Bothriopsis taeniata. Both venoms presented high enzymatic activities for proteases kallikrein, thrombin and plasmin, low levels of trypsin, cathepsin C and leucine aminopeptidase activities, while lacked acetylcholinesterase activity. B. taeniata and B. bilineata crude venoms caused inflammation inducing neutrophil recruitment into peritoneal cavity of mice 4h after injection. Neutrophil recruitment induced by B. taeniata venom was accompanied by hemorrhage. EDTA treatment profoundly impaired neutrophil recruitment, suggesting the involvement of a metalloproteinase on venoms-induced neutrophil recruitment. Pretreatment with dexamethasone and zileuton, a 5-lipoxygenase inhibitor, significantly reduced neutrophil migration, but indomethacin and montelukast, a cysteinyl leukotriene receptor antagonist, had no effect, suggesting the involvement of lipoxygenase-derived metabolites, probably LTB(4). Together, these results show that B. bilineata and B. taeniata venoms induce a marked inflammatory reaction, with leukocyte recruitment, and hemorrhage, which parallels to a high proteolytic activity found in these venoms.


Subject(s)
Bothrops/physiology , Crotalid Venoms/toxicity , Animals , Cell Movement/drug effects , Crotalid Venoms/chemistry , Crotalid Venoms/enzymology , Edetic Acid/pharmacology , Freeze Drying , Inflammation/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , South America , Species Specificity
10.
J Biol Chem ; 282(28): 20221-9, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17502383

ABSTRACT

Heme is an ancient and ubiquitous molecule present in organisms of all kingdoms, composed of an atom of iron linked to four ligand groups of porphyrin. A high amount of free heme, a potential amplifier of the inflammatory response, is a characteristic feature of diseases with increased hemolysis or extensive cell damage. Here we demonstrate that heme, but not its analogs/precursors, induced tumor necrosis factor-alpha (TNF-alpha) secretion by macrophages dependently on MyD88, TLR4, and CD14. The activation of TLR4 by heme is exquisitely strict, requiring its coordinated iron and the vinyl groups of the porphyrin ring. Signaling of heme through TLR4 depended on an interaction distinct from the one established between TLR4 and lipopolysaccharide (LPS) since anti-TLR4/MD2 antibody or a lipid A antagonist inhibited LPS-induced TNF-alpha secretion but not heme activity. Conversely, protoporphyrin IX antagonized heme without affecting LPS-induced activation. Moreover, heme induced TNF-alpha and keratinocyte chemokine but was ineffective to induce interleukin-6, interleukin-12, and interferon-inducible protein-10 secretion or co-stimulatory molecule expression. These findings support the concept that the broad ligand specificity of TLR4 and the different activation profiles might in part reside in its ability to recognize different ligands in different binding sites. Finally, heme induced oxidative burst, neutrophil recruitment, and heme oxygenase-1 expression independently of TLR4. Thus, our results presented here reveal a previous unrecognized role of heme as an extracellular signaling molecule that affects the innate immune response through a receptor-mediated mechanism.


Subject(s)
Heme/immunology , Immunity, Innate/immunology , Macrophage Activation/immunology , Macrophages/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Cells, Cultured , Cytokines/immunology , Drug Antagonism , Gene Expression Regulation, Enzymologic , Heme/antagonists & inhibitors , Heme/pharmacology , Heme Oxygenase-1/immunology , Hemolysis/genetics , Hemolysis/immunology , Immunity, Innate/drug effects , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/immunology , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Neutrophils/immunology , Photosensitizing Agents/antagonists & inhibitors , Photosensitizing Agents/pharmacology , Protoporphyrins/antagonists & inhibitors , Protoporphyrins/pharmacology , Respiratory Burst/drug effects , Respiratory Burst/genetics , Respiratory Burst/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 4/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...