Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Comp Physiol B ; 194(2): 105-119, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38573502

ABSTRACT

The innate immune system, a cornerstone for organismal resilience against environmental and microbial insults, is highly conserved across the evolutionary spectrum, underpinning its pivotal role in maintaining homeostasis and ensuring survival. This review explores the evolutionary parallels between mammalian and insect innate immune systems, illuminating how investigations into these disparate immune landscapes have been reciprocally enlightening. We further delve into how advancements in mammalian immunology have enriched our understanding of insect immune responses, highlighting the intertwined evolutionary narratives and the shared molecular lexicon of immunity across these organisms. Therefore, this review posits a holistic understanding of innate immune mechanisms, including immunometabolism, autophagy and cell death. The examination of how emerging insights into mammalian and vertebrate immunity inform our understanding of insect immune responses and their implications for vector-borne disease transmission showcases the imperative for a nuanced comprehension of innate immunity's evolutionary tale. This understanding is quintessential for harnessing innate immune mechanisms' potential in devising innovative disease mitigation strategies and promoting organismal health across the animal kingdom.


Subject(s)
Biological Evolution , Immunity, Innate , Insecta , Mammals , Animals , Insecta/immunology , Mammals/immunology , Autophagy/immunology
2.
Cancers (Basel) ; 15(19)2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37835434

ABSTRACT

Glioblastoma (GB) is the most aggressive primary malignant brain tumor and is associated with short survival. O-GlcNAcylation is an intracellular glycosylation that regulates protein function, enzymatic activity, protein stability, and subcellular localization. Aberrant O-GlcNAcylation is related to the tumorigenesis of different tumors, and mounting evidence supports O-GlcNAc transferase (OGT) as a potential therapeutic target. Here, we used two human GB cell lines alongside primary human astrocytes as a non-tumoral control to investigate the role of O-GlcNAcylation in cell proliferation, cell cycle, autophagy, and cell death. We observed that hyper O-GlcNAcylation promoted increased cellular proliferation, independent of alterations in the cell cycle, through the activation of autophagy. On the other hand, hypo O-GlcNAcylation inhibited autophagy, promoted cell death by apoptosis, and reduced cell proliferation. In addition, the decrease in O-GlcNAcylation sensitized GB cells to the chemotherapeutic temozolomide (TMZ) without affecting human astrocytes. Combined, these results indicated a role for O-GlcNAcylation in governing cell proliferation, autophagy, cell death, and TMZ response, thereby indicating possible therapeutic implications for treating GB. These findings pave the way for further research and the development of novel treatment approaches which may contribute to improved outcomes and increased survival rates for patients facing this challenging disease.

3.
Biochem Eng J ; 186: 108537, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35874089

ABSTRACT

Serological tests detect antibodies generated by infection or vaccination, and are indispensable tools along different phases of a pandemic, from early monitoring of pathogen spread up to seroepidemiological studies supporting immunization policies. This work discusses the development of an accurate and affordable COVID-19 antibody test, from production of a recombinant protein antigen up to test validation and economic analysis. We first developed a cost-effective, scalable technology to produce SARS-COV-2 spike protein and then used this antigen to develop an enzyme-linked immunosorbent assay (ELISA). A receiver operator characteristic (ROC) analysis allowed optimizing the cut-off and confirmed the high accuracy of the test: 98.6% specificity and 95% sensitivity for 11+ days after symptoms onset. We further showed that dried blood spots collected by finger pricking on simple test strips could replace conventional plasma/serum samples. A cost estimate was performed and revealed a final retail price in the range of one US dollar, reflecting the low cost of the ELISA test platform and the elimination of the need for venous blood sampling and refrigerated sample handling in clinical laboratories. The presented workflow can be completed in 4 months from first antigen expression to final test validation. It can be applied to other pathogens and in future pandemics, facilitating reliable and affordable seroepidemiological surveillance also in remote areas and in low-income countries.

5.
Front Cell Infect Microbiol ; 11: 668034, 2021.
Article in English | MEDLINE | ID: mdl-33996638

ABSTRACT

The ability to sense and adequately respond to variable environmental conditions is central for cellular and organismal homeostasis. Eukaryotic cells are equipped with highly conserved stress-response mechanisms that support cellular function when homeostasis is compromised, promoting survival. Two such mechanisms - the unfolded protein response (UPR) and autophagy - are involved in the cellular response to perturbations in the endoplasmic reticulum, in calcium homeostasis, in cellular energy or redox status. Each of them operates through conserved signaling pathways to promote cellular adaptations that include re-programming transcription of genes and translation of new proteins and degradation of cellular components. In addition to their specific functions, it is becoming increasingly clear that these pathways intersect in many ways in different contexts of cellular stress. Viral infections are a major cause of cellular stress as many cellular functions are coopted to support viral replication. Both UPR and autophagy are induced upon infection with many different viruses with varying outcomes - in some instances controlling infection while in others supporting viral replication and infection. The role of UPR and autophagy in response to coronavirus infection has been a matter of debate in the last decade. It has been suggested that CoV exploit components of autophagy machinery and UPR to generate double-membrane vesicles where it establishes its replicative niche and to control the balance between cell death and survival during infection. Even though the molecular mechanisms are not fully elucidated, it is clear that UPR and autophagy are intimately associated during CoV infections. The current SARS-CoV-2 pandemic has brought renewed interest to this topic as several drugs known to modulate autophagy - including chloroquine, niclosamide, valinomycin, and spermine - were proposed as therapeutic options. Their efficacy is still debatable, highlighting the need to better understand the molecular interactions between CoV, UPR and autophagy.


Subject(s)
COVID-19 , Autophagy , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Humans , SARS-CoV-2 , Unfolded Protein Response
6.
PLoS Pathog ; 16(7): e1008599, 2020 07.
Article in English | MEDLINE | ID: mdl-32692767

ABSTRACT

Heme oxygenase (HO-1) mediates the enzymatic cleavage of heme, a molecule with proinflammatory and prooxidant properties. HO-1 activity deeply impacts host capacity to tolerate infection through reduction of tissue damage or affecting resistance, the ability of the host to control pathogen loads. In this Review, we will discuss the contribution of HO-1 in different and complex protozoan infections, such as malaria, leishmaniasis, Chagas disease, and toxoplasmosis. The complexity of these infections and the pleiotropic effects of HO-1 constitute an interesting area of study and an opportunity for drug development.


Subject(s)
Heme Oxygenase-1/metabolism , Protozoan Infections/enzymology , Animals , Humans , Immune Tolerance/physiology
7.
Eur J Cell Biol ; 99(1): 151060, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31812279

ABSTRACT

Trypanosoma cruzi causes Chagas disease, a neglected illness that affects millions of people worldwide, especially in Latin America. The balance between biochemical pathways triggered by the parasite and host cells response will ultimately define the progression of a life-threatening disease, justifying the efforts to understand cellular mechanisms for infection restrain. In this interaction, parasite and host cells are affected by different physiological responses as autophagy modulation, which could be under intense cellular stress, such as nutrient deprivation, hormone depletion, or infection. Autophagy is a constitutive pathway that leads to degradation of macromolecules and cellular structures and may induce cell death. In Trypanosoma cruzi infection, the relevance of host autophagy is controversial regarding in vitro parasite intracellular life cycle. In the present study, we evaluated host cell autophagy during T. cruzi infection in phagocytic and non-professional phagocytic cells. We described that the presence of the parasite increased the number of LC3 puncta, a marker for autophagy, in cardiac cells and peritoneal macrophages in vitro. The induction of host autophagy decreased infection in macrophages in early and late time-periods. We suggest that starved phagocytic cells reduced internalization, also confirmed by inert particles and dead trypomastigotes. Whereas, in cardiac cells, starvation-induced autophagy decreased lipid droplets and infection in later time-point, by reducing parasite differentiation/proliferation. In ATG5 knockout MEF cells, we confirmed our hypothesis of autophagy machinery activation during parasite internalization, increasing infection. Our data suggest that host autophagy downregulates T. cruzi infection through impairing parasite intracellular life cycle, reducing the infection in primary culture cells.


Subject(s)
Autophagy , Chagas Disease/metabolism , Animals , Cell Survival , Cells, Cultured , Chagas Disease/pathology , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Trypanosoma cruzi/isolation & purification , Trypanosoma cruzi/metabolism
8.
Respir Physiol Neurobiol ; 259: 30-36, 2019 01.
Article in English | MEDLINE | ID: mdl-29997055

ABSTRACT

Acute lung injury (ALI) remains a major cause of mortality. In lipopolysaccharide (LPS)-stimulated macrophages, eugenol reduces cyclooxygenase-2 expression, NF-κB activation, and inflammatory mediators. We examined the anti-inflammatory and anti-oxidative action of eugenol in an in vivo model of LPS-induced lung injury. Lung mechanics and histology were analyzed in mice 24 h after LPS exposure, with and without eugenol treatment at different doses. Additional animals, submited to the same protocol, were treated with eugenol at 150 mg/kg to determine its effect on inflammatory cytokines (ELISA) and oxidative markers. LPS-induced lung functional and histological changes were significantly improved by eugenol, in a dose-dependent way. Furthermore, eugenol (150 mg/kg) was able to inhibit the release of inflammatory cytokines (TNF-α, IL-1ß and IL-6), NADPH oxidase activity, as well as antioxidant enzymes activity (superoxide dismutase, catalase and glutathione peroxidase). Finally, eugenol reduced LPS-induced protein oxidation. In conclusion, eugenol improved in vivo LPS-induced ALI through both anti-inflammatory and anti-oxidative effects, avoiding damage to lung structure.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Eugenol/therapeutic use , Inflammation/drug therapy , Inflammation/etiology , Lung Injury/complications , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Lipopolysaccharides/toxicity , Lung Injury/chemically induced , Male , Mice , Mice, Inbred BALB C , NADPH Oxidases/metabolism , Pulmonary Medicine/methods , Statistics, Nonparametric
9.
Front Immunol ; 9: 935, 2018.
Article in English | MEDLINE | ID: mdl-29875765

ABSTRACT

Cellular responses to stress can be defined by the overwhelming number of changes that cells go through upon contact with and stressful conditions such as infection and modifications in nutritional status. One of the main cellular responses to stress is autophagy. Much progress has been made in the understanding of the mechanisms involved in the induction of autophagy during infection by intracellular bacteria. This review aims to discuss recent findings on the role of autophagy as a cellular response to intracellular bacterial pathogens such as, Streptococcus pyogenes, Mycobacterium tuberculosis, Shigella flexneri, Salmonella typhimurium, Listeria monocytogenes, and Legionella pneumophila, how the autophagic machinery senses these bacteria directly or indirectly (through the detection of bacteria-induced nutritional stress), and how some of these bacterial pathogens manage to escape from autophagy.


Subject(s)
Autophagy , Bacterial Infections/microbiology , Bacterial Physiological Phenomena , Host-Pathogen Interactions , Intracellular Space/microbiology , AMP-Activated Protein Kinases/metabolism , Amino Acids/metabolism , Animals , Autophagosomes/immunology , Autophagosomes/metabolism , Autophagosomes/microbiology , Autophagy/immunology , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Physiological Phenomena/immunology , Biological Transport , Biomarkers , Host-Pathogen Interactions/immunology , Humans , Intracellular Space/immunology , Intracellular Space/metabolism , Signal Transduction
10.
Autophagy ; 13(3): 625-626, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28055290

ABSTRACT

Heme is an essential molecule expressed in many tissues where it plays key roles as the prosthetic group of several proteins involved in vital physiological and metabolic processes such as gas and electron transport. Structurally, heme is a tetrapyrrole ring containing an atom of iron (Fe) in its center. When released into the extracellular milieu, heme exerts several deleterious effects, which make it an important player in infectious and noninfectious hemolytic diseases where large amounts of free heme are observed such as malaria, dengue fever, ß-thalassemia, sickle cell disease and ischemia-reperfusion. Our recent work has uncovered an unappreciated cellular response triggered by heme or Fe, one of its degradation products, on macrophages, which is the formation of protein aggregates known as aggresome-like induced structres (ALIS). This response was shown to be fully dependent on ROS production and the activation of the transcription factor NFE2L2/NRF2. In addition, we have demonstrated that heme degradation by HMOX1/HO-1 (heme oxygenase 1) is required and that Fe is essential for the formation of ALIS, as heme analogs lacking the central atom of Fe are not able to induce these structures. ALIS formation is also observed in vivo, in a model of phenylhydrazine (PHZ)-induced hemolysis, indicating that it is an integral part of the host response to excessive free heme and that it may play a role in cellular homeostasis.


Subject(s)
Heme/pharmacology , Iron/pharmacology , Protein Aggregates/drug effects , Animals , Humans , Models, Biological
11.
Proc Natl Acad Sci U S A ; 113(47): E7474-E7482, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821769

ABSTRACT

Hemolytic diseases include a variety of conditions with diverse etiologies in which red blood cells are destroyed and large amounts of hemeproteins are released. Heme has been described as a potent proinflammatory molecule that is able to induce multiple innate immune responses, such as those triggered by TLR4 and the NLRP3 inflammasome, as well as necroptosis in macrophages. The mechanisms by which eukaryotic cells respond to the toxic effects induced by heme to maintain homeostasis are not fully understood, however. Here we describe a previously uncharacterized cellular response induced by heme: the formation of p62/SQTM1 aggregates containing ubiquitinated proteins in structures known as aggresome-like induced structures (ALIS). This action is part of a response driven by the transcription factor NRF2 to the excessive generation of reactive oxygen species induced by heme that results in the expression of genes involved in antioxidant responses, including p62/SQTM1. Furthermore, we show that heme degradation by HO-1 is required for ALIS formation, and that the free iron released on heme degradation is necessary and sufficient to induce ALIS. Moreover, ferritin, a key protein in iron metabolism, prevents excessive ALIS formation. Finally, in vivo, hemolysis promotes an increase in ALIS formation in target tissues. Our data unravel a poorly understood aspect of the cellular responses induced by heme that can be explored to better understand the effects of free heme and free iron during hemolytic diseases such as sickle cell disease, dengue fever, malaria, and sepsis.


Subject(s)
Heme Oxygenase-1/metabolism , Heme/metabolism , Iron/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Animals , Ferritins/metabolism , HEK293 Cells , Heme/chemistry , Humans , Mice , Oxidative Stress , Protein Aggregates , Proteolysis , RAW 264.7 Cells , Sequestosome-1 Protein/chemistry , Ubiquitination , Up-Regulation
12.
Nat Commun ; 7: 13344, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27882934

ABSTRACT

Diabetes mellitus (DM) encompasses a multitude of secondary disorders, including heart disease. One of the most frequent and potentially life threatening disorders of DM-induced heart disease is ventricular tachycardia (VT). Here we show that toll-like receptor 2 (TLR2) and NLRP3 inflammasome activation in cardiac macrophages mediate the production of IL-1ß in DM mice. IL-1ß causes prolongation of the action potential duration, induces a decrease in potassium current and an increase in calcium sparks in cardiomyocytes, which are changes that underlie arrhythmia propensity. IL-1ß-induced spontaneous contractile events are associated with CaMKII oxidation and phosphorylation. We further show that DM-induced arrhythmias can be successfully treated by inhibiting the IL-1ß axis with either IL-1 receptor antagonist or by inhibiting the NLRP3 inflammasome. Our results establish IL-1ß as an inflammatory connection between metabolic dysfunction and arrhythmias in DM.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Interleukin-1beta/immunology , Macrophages/immunology , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Tachycardia, Ventricular/immunology , Toll-Like Receptor 2/immunology , Action Potentials , Animals , Antirheumatic Agents/pharmacology , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Caspase 1/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Inflammasomes/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Transgenic , Myocardial Contraction , Myocytes, Cardiac/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Potassium/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/metabolism , Toll-Like Receptor 2/genetics
13.
Microbes Infect ; 18(3): 169-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26774331

ABSTRACT

Despite a long battle that was started by Oswaldo Cruz more than a century ago, in 1903, Brazil still struggles to fight Aedes aegypti and Aedes albopictus, the mosquito vectors of dengue virus (DENV), Chikungynya virus (CHIKV) and Zika virus (ZIKV). Dengue fever has been a serious public health problem in Brazil for decades, with recurrent epidemic outbreaks occurring during summers. In 2015, until November, 1,534,932 possible cases were reported to the Ministry of Healthv. More recently, the less studied CHIKV and ZIKV have gained attention because of a dramatic increase in their incidence (around 400% for CHIKV) and the association of ZIKV infection with a 11-fold increase in the number of cases of microcephaly from 2014 to 2015 in northeast Brazil (1761 cases until December 2015). The symptoms of these three infections are very similar, which complicates the diagnosis. These include fever, headache, nausea, fatigue, and joint pain. In some cases, DENV infection develops into dengue hemorrhagic fever, a life threatening condition characterized by bleeding and decreases in platelet numbers in the blood. As for CHIKV, the most important complication is joint pain, which can last for months.


Subject(s)
Aedes/virology , Autophagy , Chikungunya Fever/transmission , Dengue/transmission , Disease Transmission, Infectious/prevention & control , Host-Pathogen Interactions , Zika Virus Infection/transmission , Animals , Brazil/epidemiology , Chikungunya Fever/epidemiology , Dengue/epidemiology , Humans , Incidence , Insect Vectors , Zika Virus Infection/epidemiology
14.
Immunity ; 39(5): 858-73, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24238340

ABSTRACT

The peptidoglycan sensor Nod2 and the autophagy protein ATG16L1 have been linked to Crohn's disease (CD). Although Nod2 and the related sensor, Nod1, direct ATG16L1 to initiate anti-bacterial autophagy, whether ATG16L1 affects Nod-driven inflammation has not been examined. Here, we uncover an unanticipated autophagy-independent role for ATG16L1 in negatively regulating Nod-driven inflammatory responses. Knockdown of ATG16L1 expression, but not that of ATG5 or ATG9a, specifically enhanced Nod-driven cytokine production. In addition, autophagy-incompetent truncated forms of ATG16L1 regulated Nod-driven cytokine responses. Mechanistically, we demonstrated that ATG16L1 interfered with poly-ubiquitination of the Rip2 adaptor and recruitment of Rip2 into large signaling complexes. The CD-associated allele of ATG16L1 was impaired in its ability to regulate Nod-driven inflammatory responses. Overall, these results suggest that ATG16L1 is critical for Nod-dependent regulation of cytokine responses and that disruption of this Nod1- or Nod2-ATG16L1 signaling axis could contribute to the chronic inflammation associated with CD.


Subject(s)
Autophagy/physiology , Carrier Proteins/physiology , Cytokines/biosynthesis , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Animals , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/genetics , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Predisposition to Disease , Humans , Inflammation , Intestinal Mucosa/cytology , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitination
15.
Front Immunol ; 4: 361, 2013.
Article in English | MEDLINE | ID: mdl-24273538

ABSTRACT

Since they were first described as cytosolic sensors of microbial molecules a decade ago, the Nod-like receptors (NLRs) have been shown to have many different and important roles in various aspects of immune and inflammatory responses, ranging from antimicrobial mechanisms to control of adaptive responses. In this review, we focus on the interplay between NLRs and autophagy, an evolutionarily conserved mechanism that is crucial for homeostasis and has recently been shown to be involved in the protective response against infections. Furthermore, the association between mutations of NLRs as well as proteins that form the autophagic machinery and inflammatory diseases such as Crohn's disease highlight the importance of these proteins and their interactions in the regulation of inflammation.

16.
Proc Natl Acad Sci U S A ; 108(36): 14896-901, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21856952

ABSTRACT

Although a number of studies have examined the development of T-helper cell type 2 (Th2) immunity in different settings, the mechanisms underlying the initiation of this arm of adaptive immunity are not well understood. We exploited the fact that immunization with antigen plus either nucleotide-binding oligomerization domain-containing proteins 1 (Nod1) or 2 (Nod2) agonists drives Th2 induction to understand how these pattern-recognition receptors mediate the development of systemic Th2 immune responses. Here, we show in bone-marrow chimeric mice that Nod1 and Nod2 expression within the stromal compartment is necessary for priming of effector CD4(+) Th2 responses and specific IgG1 antibodies. In contrast, sensing of these ligands by dendritic cells was not sufficient to induce Th2 immunity, although these cells contribute to the response. Moreover, we determined that CD11c(+) cells were the critical antigen-presenting cells, whereas basophils and B cells did not affect the capacity of Nod ligands to induce CD4(+) Th2 effector function. Finally, we found that full Th2 induction upon Nod1 and Nod2 activation was dependent on both thymic stromal lymphopoietin production by the stromal cells and the up-regulation of the costimulatory molecule, OX40 ligand, on dendritic cells. This study provides in vivo evidence of how systemic Th2 immunity is induced in the context of Nod stimulation. Such understanding will influence the rational design of therapeutics that could reprogram the immune system during an active Th1-mediated disease, such as Crohn's disease.


Subject(s)
Cytokines/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Th2 Cells/immunology , Animals , B-Lymphocytes/immunology , Basophils/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/therapy , Cytokines/genetics , Dendritic Cells/immunology , Immunity, Cellular/physiology , Immunization , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , OX40 Ligand , Protein Structure, Tertiary , Th1 Cells/immunology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology , Thymic Stromal Lymphopoietin
17.
J Biol Chem ; 285(43): 32844-32851, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20729208

ABSTRACT

Infectious diseases that cause hemolysis are among the most threatening human diseases, because of severity and/or global distribution. In these conditions, hemeproteins and heme are released, but whether heme affects the inflammatory response to microorganism molecules remains to be characterized. Here, we show that heme increased the lethality and cytokine secretion induced by LPS in vivo and enhanced the secretion of cytokines by macrophages stimulated with various agonists of innate immune receptors. Activation of nuclear factor κB (NF-κB) and MAPKs and the generation of reactive oxygen species were essential to the increase in cytokine production induced by heme plus LPS. This synergistic effect of heme and LPS was blocked by a selective inhibitor of spleen tyrosine kinase (Syk) and was abrogated in dendritic cells deficient in Syk. Moreover, inhibition of Syk and the downstream molecules PKC and PI3K reduced the reactive oxygen species generation by heme. Our results highlight a mechanism by which heme amplifies the secretion of cytokines triggered by microbial molecule activation and indicates possible pathways for therapeutic intervention during hemolytic infectious diseases.


Subject(s)
Heme/immunology , Immunity, Innate/physiology , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Protein-Tyrosine Kinases/immunology , Reactive Oxygen Species/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Heme/agonists , Heme/metabolism , Heme/pharmacology , Humans , Immunity, Innate/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/agonists , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/genetics , Protein Kinase C/immunology , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , Syk Kinase
18.
Autophagy ; 6(3): 409-11, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200479

ABSTRACT

Autophagy is one of the main cellular degradation systems in eukaryotes, responsible for the elimination of long-lived proteins and damaged organelles. Besides its well-documented role as a housekeeping mechanism, autophagy has recently caught the attention of groups working in the fields of microbiology and immunology, especially those working in innate immunity. In particular, the highly specific segregation and degradation of intracellular bacteria by the autophagic machinery was a matter of great interest. However, it was still unclear how the autophagy machinery could target intracellular bacteria with such specificity. We have recently analyzed the role of the intracellular peptidoglycan (PG) receptors Nod1 and Nod2 as a link between intracellular bacterial sensing and the induction of autophagy. Our results demonstrated that Nod2 recruits the critical autophagy protein ATG16L1 to the plasma membrane during bacterial invasion and that cells expressing mutations in these proteins--two of the most important associated with Crohn disease--autophagy is defective upon infection or stimulation with the bacterial peptidoglycan fragment MDP. Thus, our findings put together two genes previously reported as independent risk factors for the development of Crohn disease and open a venue in the study of new therapies to cure the disease.


Subject(s)
Autophagy/immunology , Bacteria/metabolism , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Signal Transduction/physiology , Animals , Autophagy-Related Proteins , Carrier Proteins/genetics , Carrier Proteins/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/microbiology , Humans , Mice , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics
19.
Gut Microbes ; 1(5): 307-315, 2010 Sep.
Article in English | MEDLINE | ID: mdl-21327039

ABSTRACT

Autophagy is a homeostatic pathway that processes and recycles damaged organelles and other cytoplasmic contents. While studies have implicated autophagy in the immune response to infection, the understanding of how the autophagic machinery specifically targets intracellular pathogens has remained elusive. Two recent studies have uncovered an autophagy-mediated immune response to bacteria through their detection by Nod receptors. In particular, Nod1 and Nod2 recruit the autophagic protein ATG16L1 to the plasma membrane at the bacterial entry site to promote an autophagy-dependent elimination of bacteria. In addition, Nod2 and ATG16L1 synergize to initiate an adaptive immune response to bacterial invasion by enhancing major histocompatibility complex (MHC) class II antigen presentation. These findings link two Crohn disease-associated susceptibility genes and reveal that cells expressing the risk-associated variants of ATG16L1 are defective in autophagy-mediated bacterial handling and antigen presentation. This could lead to bacterial persistence and contribute to the pathogenesis of the disease.

20.
Nat Immunol ; 11(1): 55-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19898471

ABSTRACT

Autophagy is emerging as a crucial defense mechanism against bacteria, but the host intracellular sensors responsible for inducing autophagy in response to bacterial infection remain unknown. Here we demonstrated that the intracellular sensors Nod1 and Nod2 are critical for the autophagic response to invasive bacteria. By a mechanism independent of the adaptor RIP2 and transcription factor NF-kappaB, Nod1 and Nod2 recruited the autophagy protein ATG16L1 to the plasma membrane at the bacterial entry site. In cells homozygous for the Crohn's disease-associated NOD2 frameshift mutation, mutant Nod2 failed to recruit ATG16L1 to the plasma membrane and wrapping of invading bacteria by autophagosomes was impaired. Our results link bacterial sensing by Nod proteins to the induction of autophagy and provide a functional link between Nod2 and ATG16L1, which are encoded by two of the most important genes associated with Crohn's disease.


Subject(s)
Autophagy , Carrier Proteins/metabolism , Cell Membrane/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Animals , Autophagy-Related Proteins , Bacteria/metabolism , Carrier Proteins/genetics , Cell Line , Cell Membrane/microbiology , Cell Membrane/ultrastructure , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence , Mutation , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...