Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38916186

RESUMEN

Significance Fidelity of intercellular communication depends on unambiguous interactions between protein ligands and membrane receptors. Most proteins destined to the extracellular space adopt the required three-dimensional shape as they travel through the endoplasmic reticulum (ER) and Golgi complex and other organelles of the exocytic pathway. However, some proteins, many of which involved in inflammation, avoid this classical secretory route and follow unconventional pathways to leave the cell. Recent advance Stringent quality control systems operate in the ER and cis-Golgi, restricting transport to native conformers, devoid of non-native disulfides and/or reactive thiols. However, some proteins released by living cells require reduced cysteines to exert their extracellular function(s). Remarkably, these proteins lack the secretory signal sequence normally required by secretory proteins for translocation into the ER lumen. Critical issues Why do IL-1ß, HMGB1 and other proinflammatory proteins avoid the ER-Golgi route to reach the intercellular space? These proteins require reactive cysteines for exerting their function. Therefore, eluding thiol-mediated quality control along the exocytic pathway is likely one of the main reasons why extracellular proteins that need to be reduced utilize unconventional pathways of secretion, where a quality control aimed at oxidating native cysteines is not present. Future directions Particularly under stress conditions, cells release redox-active enzymes and non-protein thiol compounds that exert an extracellular control of redox-sensitive protein activity, shaping inflammatory responses. This post-secretion, redox-dependent editing of protein messages is still largely undefined. Understanding the underlying mechanistic events will hopefully provide new tools to control inflammation.

2.
J Clin Immunol ; 44(2): 49, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38231350

RESUMEN

Syndrome of undifferentiated recurrent fever (SURF) is characterized by recurrent fevers, a lack of confirmed molecular diagnosis, and a complete or partial response to colchicine. Despite the clinical similarities to familial Mediterranean fever (FMF), the underlying inflammatory mechanisms of SURF are not yet understood. We here analyzed the in vitro activation of the pyrin inflammasome in a cohort of SURF patients compared to FMF and PFAPA patients. Peripheral blood mononuclear cells (PBMC) were collected from SURF (both colchicine-treated and untreated), FMF, PFAPA patients, and healthy donors. PBMC were stimulated ex vivo with Clostridium difficile toxin A (TcdA) and a PKC inhibitor (UCN-01), in the presence or absence of colchicine. The assembly of the pyrin inflammasome was evaluated by measuring the presence of apoptosis-associated Speck-like protein containing caspase recruitment domain (ASC) specks in monocytes using flow cytometry. IL-1ß secretion was quantified using an ELISA assay. No differences in TcdA-induced activation of pyrin inflammasome were observed among FMF, PFAPA, and healthy donors. Untreated SURF patients showed a reduced response to TcdA, which was normalized after colchicine treatment. In contrast to FMF, SURF patients, similar to PFAPA patients and healthy donors, did not exhibit pyrin inflammasome activation in response to UCN-01-mediated pyrin dephosphorylation. These data demonstrate that in vitro functional analysis of pyrin inflammasome activation can differentiate SURF from FMF and PFAPA patients, suggesting the involvement of the pyrin inflammasome in the pathophysiology of SURF.


Asunto(s)
Colchicina , Fiebre Mediterránea Familiar , Humanos , Colchicina/farmacología , Colchicina/uso terapéutico , Fiebre Mediterránea Familiar/diagnóstico , Fiebre Mediterránea Familiar/tratamiento farmacológico , Inflamasomas , Leucocitos Mononucleares , Pirina/genética
3.
Methods Mol Biol ; 2696: 115-122, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37578719

RESUMEN

Autoinflammatory diseases are a group of inherited and multifactorial disorders characterized by an over-activation of innate immune response. In most cases, the clinical manifestations are due to increased activity of the NLRP3 inflammasome resulting in increased IL-1ß secretion. Investigating inflammatory cells from subjects affected by autoinflammatory diseases presents a number of technical difficulties related to the rarity of the diseases, to the young age of most patients, to the difficult modulation of gene expression in primary cells. However, since cell stress is involved in the pathophysiology of these diseases, the study of freshly drawn blood monocytes from patients affected by IL-1-mediated diseases strongly increases the chances that the observed phenomena is indeed pertinent to the pathogenesis of the disease and not influenced by the long-term cell culture conditions (e.g., the high O2 tension) or gene transfection in continuous cell lines that may lead to artifacts.


Asunto(s)
Enfermedades Autoinflamatorias Hereditarias , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Monocitos/metabolismo , Interleucina-1beta/metabolismo , Inflamasomas/metabolismo , Enfermedades Autoinflamatorias Hereditarias/metabolismo
4.
Methods Mol Biol ; 2696: 281-297, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37578730

RESUMEN

Autoinflammatory diseases are a group of distinct disorders characterized by recurrent fever and inflammatory manifestations predominantly mediated by cytokines of the innate immune system, particularly IL-1ß, without involvement of autoantibodies or autoreactive T lymphocytes. Cryopyrin-associated periodic syndromes (CAPS), due to NLRP3 gene mutations, represent the prototype of these diseases. Owing to their genetic nature, most of these disorders have an early onset, ranging from the first hours to the first decade of life. Due to the rarity of CAPS patients and to the limitations of working with pediatric samples, the development of animal models of this disease is of great help for studying both pathophysiology and therapeutic strategies. In this chapter, we review the generation and characterization of a knock-in mouse bearing the NLRP3 gene with the N475K mutation, associated with CINCA, the most severe form of human CAPS.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina , Humanos , Ratones , Animales , Niño , Síndromes Periódicos Asociados a Criopirina/genética , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Interleucina-1beta/genética , Citocinas/genética , Mutación , Modelos Animales de Enfermedad
5.
Curr Opin Chem Biol ; 76: 102339, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37295350

RESUMEN

There is considerable interest in the role of oxygen-derived oxidants (often termed generically reactive oxygen species), and the potential effect of exogenous antioxidants, in the pathogenesis of infectious disease. Most of the published research focuses on the inflammatory response and the concept that oxidants are pro-inflammatory and antioxidants are anti-inflammatory. The present review discusses the evidence that both oxidants and thiol antioxidants are important in the various processes of innate and adaptive immunity, focusing on the function of the immune system in the defense against pathogens, rather than its pathogenic role in inflammatory and autoimmune disease.


Asunto(s)
Antioxidantes , Oxidantes , Antioxidantes/farmacología , Oxidación-Reducción , Oxidantes/farmacología , Especies Reactivas de Oxígeno , Estrés Oxidativo
6.
J Allergy Clin Immunol ; 150(4): 796-805, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35835255

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may result in a severe pneumonia associated with elevation of blood inflammatory parameters, reminiscent of cytokine storm syndrome. Steroidal anti-inflammatory therapies have shown efficacy in reducing mortality in critically ill patients; however, the mechanisms by which SARS-CoV-2 triggers such an extensive inflammation remain unexplained. OBJECTIVES: To dissect the mechanisms underlying SARS-CoV-2-associated inflammation in patients with severe coronavirus disease 2019 (COVID-19), we studied the role of IL-1ß, a pivotal cytokine driving inflammatory phenotypes, whose maturation and secretion are regulated by inflammasomes. METHODS: We analyzed nod-like receptor protein 3 pathway activation by means of confocal microscopy, plasma cytokine measurement, cytokine secretion following in vitro stimulation of blood circulating monocytes, and whole-blood RNA sequencing. The role of open reading frame 3a SARS-CoV-2 protein was assessed by confocal microscopy analysis following nucleofection of a monocytic cell line. RESULTS: We found that circulating monocytes from patients with COVID-19 display ASC (adaptor molecule apoptotic speck like protein-containing a CARD) specks that colocalize with nod-like receptor protein 3 inflammasome and spontaneously secrete IL-1ß in vitro. This spontaneous activation reverts following patient's treatment with the IL-1 receptor antagonist anakinra. Transfection of a monocytic cell line with cDNA coding for the ORF3a SARS-CoV-2 protein resulted in ASC speck formation. CONCLUSIONS: These results provide further evidence that IL-1ß targeting could represent an effective strategy in this disease and suggest a mechanistic explanation for the strong inflammatory manifestations associated with COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Inflamasomas , Antiinflamatorios , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Citocinas/metabolismo , ADN Complementario , Humanos , Inflamasomas/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas NLR , Receptores de Interleucina-1 , SARS-CoV-2
7.
Cancer Med ; 11(1): 183-193, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34796694

RESUMEN

Triple negative breast cancers (TNBCs) are very aggressive and have a poor prognosis due to lack of efficacious therapies. The only effective treatment is chemotherapy that however is frequently hindered by the occurrence of drug resistance. We approached this problem in vitro and in vivo on a triple negative and a hormone sensitive breast cancer cell lines: 4T1 and TS/A. A main defense mechanism of tumors is the extrusion of intracellular protons derived from the metabolic shift to glycolysis, and necessary to maintain an intracellular pH compatible with life. The resulting acidic extracellular milieu bursts the malignant behavior of tumors and impairs chemotherapy. Therefore, we investigated the efficacy of combined therapies that associate cisplatin (Cis) with proton exchanger inhibitors, such as esomeprazole (ESO) and 5-(N-ethyl-N-isopropyl)amiloride (EIPA). Our results demonstrate that in the 4T1 triple negative model the combined therapy Cis plus EIPA is significantly more effective than the other treatments. Instead, in the TS/A tumor the best therapeutic result is obtained with ESO alone. Remarkably, in both 4T1 and TS/A tumors these treatments correlate with increase of CD8+  T lymphocytes and dendritic cells, and a dramatic reduction of M2 macrophages and other suppressor myeloid cells (MDSC) in the tumor infiltrates.


Asunto(s)
Amilorida/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/uso terapéutico , Esomeprazol/uso terapéutico , Inhibidores de la Bomba de Protones/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Amilorida/uso terapéutico , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones Endogámicos BALB C , Intercambiador 1 de Sodio-Hidrógeno/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Macrófagos Asociados a Tumores/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
8.
Sci Transl Med ; 13(596)2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34078746

RESUMEN

Muscular dystrophies (MDs) are a group of genetic diseases characterized by progressive muscle wasting associated to oxidative stress and persistent inflammation. It is essential to deepen our knowledge on the mechanism connecting these two processes because current treatments for MDs have limited efficacy and/or are associated with side effects. Here, we identified the alarmin high-mobility group box 1 (HMGB1) as a functional link between oxidative stress and inflammation in MDs. The oxidation of HMGB1 cysteines switches its extracellular activities from the orchestration of tissue regeneration to the exacerbation of inflammation. Extracellular HMGB1 is present at high amount and undergoes oxidation in patients with MDs and in mouse models of Duchenne muscular dystrophy (DMD) and limb-girdle muscular dystrophy 3 (LGMDR3) compared to controls. Genetic ablation of HMGB1 in muscles of DMD mice leads to an amelioration of the dystrophic phenotype as evidenced by the reduced inflammation and muscle degeneration, indicating that HMGB1 oxidation is a detrimental process in MDs. Pharmacological treatment with an engineered nonoxidizable variant of HMGB1, called 3S, improves functional performance, muscle regeneration, and satellite cell engraftment in dystrophic mice while reducing inflammation and fibrosis. Overall, our data demonstrate that the balance between HMGB1 redox isoforms dictates whether skeletal muscle is in an inflamed or regenerating state, and that the nonoxidizable form of HMGB1 is a possible therapeutic approach to counteract the progression of the dystrophic phenotype. Rebalancing the HMGB1 redox isoforms may also be a therapeutic strategy for other disorders characterized by chronic oxidative stress and inflammation.


Asunto(s)
Proteína HMGB1 , Distrofia Muscular de Duchenne , Animales , Proteína HMGB1/metabolismo , Humanos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Oxidación-Reducción , Isoformas de Proteínas/metabolismo
9.
EMBO Mol Med ; 13(6): e12344, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33956406

RESUMEN

Boosting antitumor immunity has emerged as a powerful strategy in cancer treatment. While releasing T-cell brakes has received most attention, tumor recognition by T cells is a pre-requisite. Radiotherapy and certain cytotoxic drugs induce the release of damage-associated molecular patterns, which promote tumor antigen cross-presentation and T-cell priming. Antibodies against the "do not eat me" signal CD47 cause macrophage phagocytosis of live tumor cells and drive the emergence of antitumor T cells. Here we show that CXCR4 activation, so far associated only with tumor progression and metastasis, also flags tumor cells to immune recognition. Both CXCL12, the natural CXCR4 ligand, and BoxA, a fragment of HMGB1, promote the release of DAMPs and the internalization of CD47, leading to protective antitumor immunity. We designate as Immunogenic Surrender the process by which CXCR4 turns in tumor cells to macrophages, thereby subjecting a rapidly growing tissue to immunological scrutiny. Importantly, while CXCL12 promotes tumor cell proliferation, BoxA reduces it, and might be exploited for the treatment of malignant mesothelioma and a variety of other tumors.


Asunto(s)
Antígeno CD47 , Mesotelioma , Animales , Línea Celular Tumoral , Inmunización , Macrófagos , Mesotelioma/inmunología , Mesotelioma/metabolismo , Mesotelioma/terapia , Ratones , Fagocitosis
10.
J Biol Chem ; 295(22): 7799-7811, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32332096

RESUMEN

Members of the interleukin (IL)-1 family are key determinants of inflammation. Despite their role as intercellular mediators, most lack the leader peptide typically required for protein secretion. This lack is a characteristic of dozens of other proteins that are actively and selectively secreted from living cells independently of the classical endoplasmic reticulum-Golgi exocytic route. These proteins, termed leaderless secretory proteins (LLSPs), comprise proteins directly or indirectly involved in inflammation, including cytokines such as IL-1ß and IL-18, growth factors such as fibroblast growth factor 2 (FGF2), redox enzymes such as thioredoxin, and proteins most expressed in the brain, some of which participate in the pathogenesis of neurodegenerative disorders. Despite much effort, motifs that promote LLSP secretion remain to be identified. In this review, we summarize the mechanisms and pathophysiological significance of the unconventional secretory pathways that cells use to release LLSPs. We place special emphasis on redox regulation and inflammation, with a focus on IL-1ß, which is secreted after processing of its biologically inactive precursor pro-IL-1ß in the cytosol. Although LLSP externalization remains poorly understood, some possible mechanisms have emerged. For example, a common feature of LLSP pathways is that they become more active in response to stress and that they involve several distinct excretion mechanisms, including direct plasma membrane translocation, lysosome exocytosis, exosome formation, membrane vesiculation, autophagy, and pyroptosis. Further investigations of unconventional secretory pathways for LLSP secretion may shed light on their evolution and could help advance therapeutic avenues for managing pathological conditions, such as diseases arising from inflammation.


Asunto(s)
Evolución Molecular , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Procesamiento Proteico-Postraduccional , Vías Secretoras , Animales , Autofagia , Exocitosis , Exosomas/genética , Exosomas/metabolismo , Exosomas/patología , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-18/genética , Interleucina-1beta/genética , Lisosomas/genética , Lisosomas/metabolismo , Lisosomas/patología , Piroptosis
11.
J Nucl Cardiol ; 27(6): 2183-2194, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-30737636

RESUMEN

BACKGROUND: Oxidative stress and its interference on myocardial metabolism play a major role in Doxorubicin (DXR) cardiotoxic cascade. METHODS: Mice models of neuroblastoma (NB) were treated with 5 mg DXR/kg, either free (Free-DXR) or encapsulated in untargeted (SL[DXR]) or in NB-targeting Stealth Liposomes (pep-SL[DXR] and TP-pep-SL[DXR]). Control mice received saline. FDG-PET was performed at baseline (PET1) and 7 days after therapy (PET2). At PET2 Troponin-I and NT-proBNP were assessed. Explanted hearts underwent biochemical, histological, and immunohistochemical analyses. Finally, FDG uptake and glucose consumption were simultaneously measured in cultured H9c2 in the presence/absence of Free-DXR (1 µM). RESULTS: Free-DXR significantly enhanced the myocardial oxidative stress. Myocardial-SUV remained relatively stable in controls and mice treated with liposomal formulations, while it significantly increased at PET2 with respect to baseline in Free-DXR. At this timepoint, myocardial-SUV was directly correlated with both myocardial redox stress and hexose-6-phosphate-dehydrogenase (H6PD) enzymatic activity, which selectively sustain cellular anti-oxidant mechanisms. Intriguingly, in vitro, Free-DXR selectively increased FDG extraction fraction without altering the corresponding value for glucose. CONCLUSION: The direct correlation between cardiac FDG uptake and oxidative stress indexes supports the potential role of FDG-PET as an early biomarker of DXR oxidative damage.


Asunto(s)
Doxorrubicina/química , Fluorodesoxiglucosa F18/farmacocinética , Corazón/efectos de los fármacos , Miocardio/patología , Estrés Oxidativo , Animales , Antioxidantes , Biomarcadores/metabolismo , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Glucosa/química , Glucosa/farmacocinética , Humanos , Inmunohistoquímica , Cinética , Ratones , Ratones Desnudos , Neuroblastoma/tratamiento farmacológico , Oxidación-Reducción , Tomografía de Emisión de Positrones
12.
Curr Med Chem ; 27(25): 4233-4248, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30182839

RESUMEN

Tumor cells and tumor-associated stromal cells such as immune, endothelial and mesenchimal cells create a Tumor Microenvironment (TME) which allows tumor cell promotion, growth and dissemination while dampening the anti-tumor immune response. Efficient anti-tumor interventions have to keep into consideration the complexity of the TME and take advantage of immunotherapy and chemotherapy combined approaches. Thus, the aim of tumor therapy is to directly hit tumor cells and reverse endothelial and immune cell anergy. Selective targeting of tumor vasculature using TNFα-associated peptides or antibody fragments in association with chemotherapeutic agents, has been shown to exert a potent stimulatory effect on endothelial cells as well as on innate and adaptive immune responses. These drug combinations reducing the dose of single agents employed have led to minimize the associated side effects. In this review, we will analyze different TNFα-mediated tumor vesseltargeted therapies in both humans and tumor mouse models, with emphasis on the role played by the cross-talk between natural killer and dendritic cells and on the ability of TNFα to trigger tumor vessel activation and normalization. The improvement of the TNFα-based therapy with anti-angiogenic immunomodulatory drugs that may convert the TME from immunosuppressive to immunostimulant, will be discussed as well.


Asunto(s)
Neoplasias , Animales , Células Endoteliales , Humanos , Inmunoterapia , Microambiente Tumoral , Factor de Necrosis Tumoral alfa
13.
J Allergy Clin Immunol ; 145(1): 368-378.e13, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31194989

RESUMEN

BACKGROUND: Cryopyrin-associated periodic syndromes (CAPS) are a group of autoinflammatory diseases linked to gain-of-function mutations in the NOD-like receptor family, pyrin domain containing 3 (NLRP3) gene, which cause uncontrolled IL-1ß secretion. Proton pump inhibitors (PPIs), which are commonly used as inhibitors of gastric acid production, also have anti-inflammatory properties, protect mice from sepsis, and prevent IL-1ß secretion by monocytes from patients with CAPS. OBJECTIVE: We sought to develop a novel Nlrp3 knock-in (KI) mouse model of CAPS to study amyloidosis, a severe CAPS complication, and test novel therapeutic approaches. METHODS: We generated KI mice by engineering the N475K mutation, which is associated with the CAPS phenotype, into the mouse Nlrp3 gene. KI and wild-type mice received PPIs or PBS intraperitoneally and were analyzed for survival, inflammation, cytokine secretion, and amyloidosis development. RESULTS: Mutant Nlrp3 KI mice displayed features that recapitulate the immunologic and clinical phenotype of CAPS. They showed systemic inflammation with high levels of serum proinflammatory cytokines, inflammatory infiltrates in various organs, and amyloid deposits in the spleen, liver, and kidneys. Toll-like receptor stimulated macrophages from KI mice secreted high levels of IL-1ß, IL-18, and IL-1α but low amounts of IL-1 receptor antagonist. Treatment of KI mice with PPIs had a clear clinical effect, showing a reduction in inflammatory manifestations, regression of amyloid deposits, and normalization of proinflammatory and anti-inflammatory cytokine production by macrophages. CONCLUSION: Nlrp3 KI mice displayed a CAPS phenotype with many characteristics of autoinflammation, including amyloidosis. The therapeutic effectiveness of PPIs associated with a lack of toxicity indicates that these drugs could represent relevant adjuvants to the anti-IL-1 drugs in patients with CAPS and other IL-1-driven diseases.


Asunto(s)
Amiloidosis , Síndromes Periódicos Asociados a Criopirina , Proteína con Dominio Pirina 3 de la Familia NLR , Inhibidores de la Bomba de Protones/farmacología , Amiloidosis/tratamiento farmacológico , Amiloidosis/genética , Amiloidosis/inmunología , Animales , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Síndromes Periódicos Asociados a Criopirina/genética , Síndromes Periódicos Asociados a Criopirina/inmunología , Síndromes Periódicos Asociados a Criopirina/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Mutantes , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
14.
J Biol Chem ; 294(10): 3634-3646, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30635405

RESUMEN

Amyloid deposits of apolipoprotein A-I (apoA-I) and inflammation are common in atherosclerotic arteries. In this study, we investigated the interplay between oxidation of apoA-I methionine residues (Met(O)-ApoA-I), a known amyloidogenic modification of apoA-I, and the inflammatory response of immune cells. Soluble pre-fibrillar Met(O)-ApoA-I, but not apoA-I, induced intracellular accumulation of pro-interleukin (IL)-1ß and secretion of the pro-inflammatory cytokines tumor necrosis factor α (TNFα) and IL-6 in mouse bone marrow-derived macrophages (BMDMs) and human primary monocytes. Additionally, secretion of mature IL-1ß was also activated in human monocytes. The pro-inflammatory activity of Met(O)-ApoA-I was Toll-like receptor 4 (TLR4)-dependent and CD36-independent and was solely determined by oxidation of apoA-I methionine residues, in particular Met-86 and Met-148. In contrast, amyloid fibrils or reconstituted high-density lipoproteins (HDLs) generated from Met(O)-ApoA-I did not induce cytokine production in BMDMs. Although lipid-free Met(O)-ApoA-I remained functional in extracting lipids from cells and generating HDL, it gained strong pro-inflammatory properties that may aggravate local inflammation in the arteries and atherosclerosis. Our study indicates that oxidation of apoA-I methionine residues produces a potent danger-associated molecular pattern capable of stimulating pro-inflammatory cytokine secretion at levels similar to those induced by known pathogen-associated molecular patterns, such as lipopolysaccharide.


Asunto(s)
Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Metionina/metabolismo , Animales , Humanos , Inflamación/metabolismo , Ratones , Oxidación-Reducción , Receptor Toll-Like 4/metabolismo
15.
Front Immunol ; 9: 2118, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30283452

RESUMEN

Chemokine synergy-inducing molecules are emerging as regulating factors in cell migration. The alarmin HMGB1, in its reduced form, can complex with CXCL12 enhancing its activity on monocytes via the chemokine receptor CXCR4, while the form containing a disulfide bond, by binding to TLR2 or TLR4, initiates a cascade of events leading to production of cytokines and chemokines. So far, the possibility that the CXCL12/HMGB1 heterocomplex could be maintained in chronic inflammation was debated, due to the release of reactive oxygen species. Therefore, we have assessed if the heterocomplex could remain active in Rheumatoid Arthritis (RA) and its relevance in the disease assessment. Monocytes from RA patients with active disease require a low concentration of HMGB1 to enhance CXCL12-induced migration, in comparison to monocytes from patients in clinical remission or healthy donors. The activity of the heterocomplex depends on disease activity, on the COX2 and JAK/STAT pathways, and is determined by the redox potential of the microenvironment. In RA, the presence of an active thioredoxin system correlates with the enhanced cell migration, and with the presence of the heterocomplex in the synovial fluid. The present study highlights how, in an unbalanced microenvironment, the activity of the thioredoxin system plays a crucial role in sustaining inflammation. Prostaglandin E2 stimulation of monocytes from healthy donors is sufficient to recapitulate the response observed in patients with active RA. The activation of mechanisms counteracting the oxidative stress in the extracellular compartment preserves HMGB1 in its reduced form, and contributes to fuel the influx of inflammatory cells. Targeting the heterocomplex formation and its activity could thus be an additional tool for dampening the inflammation sustained by cell recruitment, for those patients with chronic inflammatory conditions who poorly respond to current therapies.


Asunto(s)
Artritis Reumatoide/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Proteína HMGB1/farmacología , Monocitos/efectos de los fármacos , Adulto , Anciano , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Movimiento Celular/inmunología , Células Cultivadas , Dinoprostona/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Oxidación-Reducción , Unión Proteica/efectos de los fármacos , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
16.
Cell Death Dis ; 9(11): 1088, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30352992

RESUMEN

IL-1ß is an essential cytokine, but its release needs to be strictly controlled to avoid severe inflammatory manifestations. Lacking a signal sequence, IL-1ß does not follow the endoplasmic reticulum-Golgi route. Several pathways have been proposed to mediate its release. One involves the translocation of pro-IL-1ß into intracellular vesicles of lysosomal origin that eventually fuse with the plasma membrane. Another exploits pores formed on the plasma membrane upon proteolytic cleavage of gasdermin D (GSDMD). Here we investigated how primary monocytes-the main source of IL-1ß in humans-control IL-1ß release in response to pro-inflammatory stimuli of increasing intensity and found that two different routes are induced depending on the strength of activation. Triggering of Toll-like receptor 4 (TLR4) by LPS induces slow IL-1ß release through LAMP2A+ vesicles. In contrast, the simultaneous stimulation of TLR2, TLR4 and TLR7/8 drives high levels of ROS, GSDMD cleavage and faster IL-1ß secretion. Drugs blocking ROS production prevent GSDMD cleavage supporting a role of oxidative stress in GSDMD-mediated secretion. Singly stimulated monocytes undergo apoptosis, whereas triple stimulation triggers pyroptosis, which might amplify inflammation. In both cases, however, IL-1ß secretion precedes cell death. Inhibition of caspases 4/5 prevents GSDMD cleavage and pore-mediated secretion, but not vesicular release. The two pathways also display other distinct pharmacologic sensitivities that reflect the underlying mechanisms. Remarkably, single TLR4 stimulation is sufficient to activate massive, GSDMD-mediated IL-1ß secretion in monocytes from patients affected by Cryopyrin Associated Periodic Syndrome (CAPS), an autoinflammatory disease linked to NLRP3 mutations. The exaggerated sensitivity to activation correlates with high basal ROS levels in CAPS monocytes. In conclusion, the vesicular pathway limits IL-1ß release upon low pathogen load while stronger stimulation or concomitant cell stress induce instead uncontrolled secretion via GSDMD leading to detrimental inflammatory manifestations.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Monocitos/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Vías Secretoras , Vesículas Transportadoras/metabolismo , Adolescente , Adulto , Apoptosis , Células Cultivadas , Niño , Preescolar , Síndromes Periódicos Asociados a Criopirina/sangre , Síndromes Periódicos Asociados a Criopirina/patología , Femenino , Humanos , Lipopolisacáridos/farmacología , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Monocitos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Piroptosis , Especies Reactivas de Oxígeno/metabolismo , Receptores Toll-Like/metabolismo , Adulto Joven , Zimosan/farmacología
17.
Sci Adv ; 4(5): eaar5770, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29732408

RESUMEN

Upon engagement of tyrosine kinase receptors, nicotinamide adenine dinucleotide phosphate (NADPH)-oxidases release H2O2 in the extracellular space. We reported previously that aquaporin-8 (AQP8) transports H2O2 across the plasma membrane and is reversibly gated during cell stress, modulating signal strength and duration. We show that AQP8 gating is mediated by persulfidation of cysteine 53 (C53). Treatment with H2S is sufficient to block H2O2 entry in unstressed cells. Silencing cystathionine ß-synthase (CBS) prevents closure, suggesting that this enzyme is the main source of H2S. Molecular modeling indicates that C53 persulfidation displaces a nearby histidine located in the narrowest part of the channel. We propose that H2O2 molecules transported through AQP8 sulfenylate C53, making it susceptible to H2S produced by CBS. This mechanism tunes H2O2 transport and may control signaling and limit oxidative stress.


Asunto(s)
Acuaporinas/metabolismo , Sulfuros/metabolismo , Secuencia de Aminoácidos , Acuaporinas/química , Transporte Biológico , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Peróxido de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/metabolismo , Modelos Biológicos , Conformación Molecular , Oxidación-Reducción , Estrés Fisiológico , Sulfuros/química
18.
Semin Cell Dev Biol ; 83: 12-21, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29571971

RESUMEN

Interleukin 1ß (IL-1ß) is a major mediator of inflammation, with a causative role in many diseases. Unlike most other cytokines, however, it lacks a secretory signal sequence, raising intriguing mechanistic, functional and evolutionary questions. Despite decades of strenuous efforts in many laboratories, how IL-1ß is secreted is still a matter of intense debate. Here, we summarize the different mechanisms and pathways that have been proposed for IL-1ß secretion. At least two of them, namely the endolysosomal vesicle-based and gasdermin D-dependent pathways (types III and I in the recent Rabouille's classification of unconventional protein secretion), can be triggered in monocytes, the main source of IL-1ß in humans, according to the type and strength of the pro-inflammatory stimuli. As during the escalation of human conflicts, monocytes deploy secretory mechanisms of increasing efficiency and dangerousness, shifting from the specific and controlled type III pathway to the much faster release of type I. Thus, the different mechanisms are activated depending on the severity of the conditions, from the self-limiting type III pathways in response of low pathogen load or small trauma, to the uncontrolled responses that underlie autoinflammatory disorders and sepsis.


Asunto(s)
Autofagia/fisiología , Interleucina-1beta/metabolismo , Lisosomas/metabolismo , Humanos
19.
Proc Natl Acad Sci U S A ; 115(7): E1530-E1539, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29378952

RESUMEN

Activation of the NLRP3 inflammasome induces maturation of IL-1ß and IL-18, both validated targets for treating acute and chronic inflammatory diseases. Here, we demonstrate that OLT1177, an orally active ß-sulfonyl nitrile molecule, inhibits activation of the NLRP3 inflammasome. In vitro, nanomolar concentrations of OLT1177 reduced IL-1ß and IL-18 release following canonical and noncanonical NLRP3 inflammasome activation. The molecule showed no effect on the NLRC4 and AIM2 inflammasomes, suggesting specificity for NLRP3. In LPS-stimulated human blood-derived macrophages, OLT1177 decreased IL-1ß levels by 60% and IL-18 by 70% at concentrations 100-fold lower in vitro than plasma concentrations safely reached in humans. OLT1177 also reduced IL-1ß release and caspase-1 activity in freshly obtained human blood neutrophils. In monocytes isolated from patients with cryopyrin-associated periodic syndrome (CAPS), OLT1177 inhibited LPS-induced IL-1ß release by 84% and 36%. Immunoprecipitation and FRET analysis demonstrated that OLT1177 prevented NLRP3-ASC, as well as NLRP3-caspase-1 interaction, thus inhibiting NLRP3 inflammasome oligomerization. In a cell-free assay, OLT1177 reduced ATPase activity of recombinant NLRP3, suggesting direct targeting of NLRP3. Mechanistically, OLT1177 did not affect potassium efflux, gene expression, or synthesis of the IL-1ß precursor. Steady-state levels of phosphorylated NF-κB and IkB kinase were significantly lowered in spleen cells from OLT1177-treated mice. We observed reduced IL-1ß content in tissue homogenates, limited oxidative stress, and increased muscle oxidative metabolism in OLT1177-treated mice challenged with LPS. Healthy humans receiving 1,000 mg of OLT1177 daily for 8 d exhibited neither adverse effects nor biochemical or hematological changes.


Asunto(s)
Antiinflamatorios/farmacología , Inflamasomas/antagonistas & inhibidores , Inflamación/prevención & control , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nitrilos/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Caspasa 1/metabolismo , Células Cultivadas , Humanos , Inflamación/inducido químicamente , Inflamación/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nitrilos/química , Nitrilos/uso terapéutico
20.
Oncotarget ; 8(40): 67482-67496, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978047

RESUMEN

Neoplastic cells live in a stressful context and survive thanks to their ability to overcome stress. Thus, tumor cell responses to stress are potential therapeutic targets. We selected two such responses in melanoma and sarcoma cells: the xc- antioxidant system, that opposes oxidative stress, and surface v-ATPases that counteract the low pHi by extruding protons, and targeted them with the xc- blocker sulfasalazine and the proton pump inhibitor esomeprazole. Sulfasalazine inhibited the cystine/cysteine redox cycle and esomeprazole decreased pHi while increasing pHe in tumor cell lines. Although the single treatment with either drug slightly inhibited cell proliferation and motility, the association of sulfasalazine and esomeprazole powerfully decreased sarcoma and melanoma growth and migration. In the 3-methylcholanthrene (3-MCA)-induced sarcoma model, the combined therapy strongly reduced the tumor burden and increased the survival time: notably, 22 % of double-treated mice recovered and survived off therapy. Tumor-associated macrophages (TAM) displaying M2 markers, that abundantly infiltrate sarcoma and melanoma, overexpress xc- and membrane v-ATPases and were drastically decreased in tumors from mice undergone the combined therapy. Thus, the double targeting of tumor cells and macrophages by sulfasalazine and esomeprazole has a double therapeutic effect, as decreasing TAM infiltration deprives tumor cells of a crucial allied. Sulfasalazine and esomeprazole may therefore display unexpected therapeutic values, especially in case of hard-to-treat cancers.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...