Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Curr Biol ; 33(1): R33-R36, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36626862

RESUMEN

A new study reveals how Mycobacterium tuberculosis evades anti-bacterial immunity by modifying the plasma membrane phospholipid composition of infected macrophages, thereby blocking the host's pyroptosis response and supporting chronic infection.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Inflamasomas/metabolismo , Tuberculosis/metabolismo , Tuberculosis/microbiología , Macrófagos/metabolismo
2.
EMBO Rep ; 23(10): e54277, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35899491

RESUMEN

Neutrophils are the most prevalent immune cells in circulation, but the repertoire of canonical inflammasomes in neutrophils and their respective involvement in neutrophil IL-1ß secretion and neutrophil cell death remain unclear. Here, we show that neutrophil-targeted expression of the disease-associated gain-of-function Nlrp3A350V mutant suffices for systemic autoinflammatory disease and tissue pathology in vivo. We confirm the activity of the canonical NLRP3 and NLRC4 inflammasomes in neutrophils, and further show that the NLRP1b, Pyrin and AIM2 inflammasomes also promote maturation and secretion of interleukin (IL)-1ß in cultured bone marrow neutrophils. Notably, all tested canonical inflammasomes promote GSDMD cleavage in neutrophils, and canonical inflammasome-induced pyroptosis and secretion of mature IL-1ß are blunted in GSDMD-knockout neutrophils. In contrast, GSDMD is dispensable for PMA-induced NETosis. We also show that Salmonella Typhimurium-induced pyroptosis is markedly increased in Nox2/Gp91Phox -deficient neutrophils that lack NADPH oxidase activity and are defective in PMA-induced NETosis. In conclusion, we establish the canonical inflammasome repertoire in neutrophils and identify differential roles for GSDMD and the NADPH complex in canonical inflammasome-induced neutrophil pyroptosis and mitogen-induced NETosis, respectively.


Asunto(s)
Trampas Extracelulares , Inflamasomas , Neutrófilos , Proteínas de Unión a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Piroptosis , Animales , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitógenos/metabolismo , NADP/metabolismo , NADPH Oxidasas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Neutrófilos/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Pirina/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34996874

RESUMEN

Lethal toxin (LeTx)-mediated killing of myeloid cells is essential for Bacillus anthracis, the causative agent of anthrax, to establish systemic infection and induce lethal anthrax. The "LeTx-sensitive" NLRP1b inflammasome of BALB/c and 129S macrophages swiftly responds to LeTx intoxication with pyroptosis and secretion of interleukin (IL)-1ß. However, human NLRP1 is nonresponsive to LeTx, prompting us to investigate B. anthracis host-pathogen interactions in C57BL/6J (B6) macrophages and mice that also lack a LeTx-sensitive Nlrp1b allele. Unexpectedly, we found that LeTx intoxication and live B. anthracis infection of B6 macrophages elicited robust secretion of IL-1ß, which critically relied on the NLRP3 inflammasome. TNF signaling through both TNF receptor 1 (TNF-R1) and TNF-R2 were required for B. anthracis-induced NLRP3 inflammasome activation, which was further controlled by RIPK1 kinase activity and LeTx-mediated proteolytic inactivation of MAP kinase signaling. In addition to activating the NLRP3 inflammasome, LeTx-induced MAPKK inactivation and TNF production sensitized B. anthracis-infected macrophages to robust RIPK1- and caspase-8-dependent apoptosis. In agreement, purified LeTx triggered RIPK1 kinase activity- and caspase-8-dependent apoptosis only in macrophages primed with TNF or following engagement of TRIF-dependent Toll-like receptors. Consistently, genetic and pharmacological inhibition of RIPK1 inhibited NLRP3 inflammasome activation and apoptosis of LeTx-intoxicated and B. anthracis-infected macrophages. Caspase-8/RIPK3-deficient mice were significantly protected from B. anthracis-induced lethality, demonstrating the in vivo pathophysiological relevance of this cytotoxic mechanism. Collectively, these results establish TNF- and RIPK1 kinase activity-dependent NLRP3 inflammasome activation and macrophage apoptosis as key host-pathogen mechanisms in lethal anthrax.


Asunto(s)
Apoptosis , Bacillus anthracis/metabolismo , Caspasa 8/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Carbunco , Caspasa 8/genética , Interacciones Huésped-Patógeno/fisiología , Inflamasomas/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Piroptosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal
4.
Cell Res ; 32(3): 227-228, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34934194
5.
Nanoscale ; 13(13): 6592-6604, 2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33885539

RESUMEN

Inflammasomes are multi-protein complexes that guard against cellular stress and microbial infections. Inflammasome activation studies frequently require delivery of pathogen-derived virulence factors into the cytosol of macrophages and other innate immune cells. This is a challenging requirement since primary macrophages are difficult-to-transfect, especially when it comes to the intracellular delivery of proteins. Here, we report on the use of nanoparticle-sensitized photoporation as a promising upcoming intracellular delivery technology for delivering proteins of various molecular weights into the cytosol of primary macrophages. While 60-70 nm gold nanoparticles are the most commonly used sensitizing nanoparticles for photoporation, here we find that 0.5 µm iron oxide nanoparticles perform markedly better on primary macrophages. We demonstrate that LFn-FlaA or lipopolysaccharides can be delivered in primary macrophages resulting in activation of the NLRC4 or the non-canonical inflammasome, respectively. We furthermore show that photoporation can be used for targeted delivery of these toxins into selected cells, opening up the possibility to study the interaction between inflammasome activated cells and surrounding healthy cells. Taken together, these results show that nanoparticle-sensitized photoporation is very well suited to deliver pathogenic virulence factors in primary macrophages, thus constituting an effective new enabling technology for inflammasome activation studies.


Asunto(s)
Inflamasomas , Nanopartículas del Metal , Oro , Lipopolisacáridos , Macrófagos , Nanopartículas del Metal/toxicidad
7.
PLoS Biol ; 17(9): e3000354, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31525186

RESUMEN

The nucleotide-binding-domain (NBD)-and leucine-rich repeat (LRR)-containing (NLR) family, pyrin-domain-containing 3 (NLRP3) inflammasome drives pathological inflammation in a suite of autoimmune, metabolic, malignant, and neurodegenerative diseases. Additionally, NLRP3 gain-of-function point mutations cause systemic periodic fever syndromes that are collectively known as cryopyrin-associated periodic syndrome (CAPS). There is significant interest in the discovery and development of diarylsulfonylurea Cytokine Release Inhibitory Drugs (CRIDs) such as MCC950/CRID3, a potent and selective inhibitor of the NLRP3 inflammasome pathway, for the treatment of CAPS and other diseases. However, drug discovery efforts have been constrained by the lack of insight into the molecular target and mechanism by which these CRIDs inhibit the NLRP3 inflammasome pathway. Here, we show that the NAIP, CIITA, HET-E, and TP1 (NACHT) domain of NLRP3 is the molecular target of diarylsulfonylurea inhibitors. Interestingly, we find photoaffinity labeling (PAL) of the NACHT domain requires an intact (d)ATP-binding pocket and is substantially reduced for most CAPS-associated NLRP3 mutants. In concordance with this finding, MCC950/CRID3 failed to inhibit NLRP3-driven inflammatory pathology in two mouse models of CAPS. Moreover, it abolished circulating levels of interleukin (IL)-1ß and IL-18 in lipopolysaccharide (LPS)-challenged wild-type mice but not in Nlrp3L351P knock-in mice and ex vivo-stimulated mutant macrophages. These results identify wild-type NLRP3 as the molecular target of MCC950/CRID3 and show that CAPS-related NLRP3 mutants escape efficient MCC950/CRID3 inhibition. Collectively, this work suggests that MCC950/CRID3-based therapies may effectively treat inflammation driven by wild-type NLRP3 but not CAPS-associated mutants.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/genética , Furanos/farmacología , Inflamasomas/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Citocinas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos , Humanos , Indenos , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Dominios Proteicos , Sulfonas
8.
Nat Microbiol ; 4(11): 1805-1814, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31308522

RESUMEN

Anthrax is an ancient and deadly disease caused by the spore-forming bacterial pathogen Bacillus anthracis. At present, anthrax mostly affects wildlife and livestock, although it remains a concern for human public health-primarily for people who handle contaminated animal products and as a bioterrorism threat due to the high resilience of spores, a high fatality rate of cases and the lack of a civilian vaccination programme1,2. The cell surface of B. anthracis is covered by a protective paracrystalline monolayer-known as surface layer or S-layer-that is composed of the S-layer proteins Sap or EA1. Here, we generate nanobodies to inhibit the self-assembly of Sap, determine the structure of the Sap S-layer assembly domain (SapAD) and show that the disintegration of the S-layer attenuates the growth of B. anthracis and the pathology of anthrax in vivo. SapAD comprises six ß-sandwich domains that fold and support the formation of S-layers independently of calcium. Sap-inhibitory nanobodies prevented the assembly of Sap and depolymerized existing Sap S-layers in vitro. In vivo, nanobody-mediated disruption of the Sap S-layer resulted in severe morphological defects and attenuated bacterial growth. Subcutaneous delivery of Sap inhibitory nanobodies cleared B. anthracis infection and prevented lethality in a mouse model of anthrax disease. These findings highlight disruption of S-layer integrity as a mechanism that has therapeutic potential in S-layer-carrying pathogens.


Asunto(s)
Carbunco/tratamiento farmacológico , Bacillus anthracis/efectos de los fármacos , Glicoproteínas de Membrana/química , Anticuerpos de Dominio Único/administración & dosificación , Animales , Carbunco/metabolismo , Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidad , Modelos Animales de Enfermedad , Inyecciones Subcutáneas , Glicoproteínas de Membrana/metabolismo , Ratones , Viabilidad Microbiana/efectos de los fármacos , Modelos Moleculares , Conformación Proteica en Lámina beta/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Anticuerpos de Dominio Único/farmacología
9.
Cell Rep ; 21(12): 3427-3444, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262324

RESUMEN

The caspase activation and recruitment domain (CARD)-based inflammasome sensors NLRP1b and NLRC4 induce caspase-1-dependent pyroptosis independent of the inflammasome adaptor ASC. Here, we show that NLRP1b and NLRC4 trigger caspase-8-mediated apoptosis as an alternative cell death program in caspase-1-/- macrophages and intestinal epithelial organoids (IECs). The caspase-8 adaptor FADD was recruited to ASC specks, which served as cytosolic platforms for caspase-8 activation and NLRP1b/NLRC4-induced apoptosis. We further found that caspase-1 protease activity dominated over scaffolding functions in suppressing caspase-8 activation and induction of apoptosis of macrophages and IECs. Moreover, TLR-induced c-FLIP expression inhibited caspase-8-mediated apoptosis downstream of ASC speck assembly, but did not affect pyroptosis induction by NLRP1b and NLRC4. Moreover, unlike during pyroptosis, NLRP1b- and NLRC4-elicited apoptosis retained alarmins and the inflammasome-matured cytokines interleukin 1ß (IL-1ß) and IL-18 intracellularly. This work identifies critical mechanisms regulating apoptosis induction by the inflammasome sensors NLRP1b and NLRC4 and suggests converting pyroptosis into apoptosis as a paradigm for suppressing inflammation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Piroptosis , Animales , Caspasa 8/metabolismo , Enterocitos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Toll-Like/metabolismo
10.
Immunity ; 47(2): 339-348.e4, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28801232

RESUMEN

The gut microbiota regulate susceptibility to multiple human diseases. The Nlrp6-ASC inflammasome is widely regarded as a hallmark host innate immune axis that shapes the gut microbiota composition. This notion stems from studies reporting dysbiosis in mice lacking these inflammasome components when compared with non-littermate wild-type animals. Here, we describe microbial analyses in inflammasome-deficient mice while minimizing non-genetic confounders using littermate-controlled Nlrp6-deficient mice and ex-germ-free littermate-controlled ASC-deficient mice that were all allowed to shape their gut microbiota naturally after birth. Careful microbial phylogenetic analyses of these cohorts failed to reveal regulation of the gut microbiota composition by the Nlrp6- and ASC-dependent inflammasomes. Our results obtained in two geographically separated animal facilities dismiss a generalizable impact of Nlrp6- and ASC-dependent inflammasomes on the composition of the commensal gut microbiota and highlight the necessity for littermate-controlled experimental design in assessing the influence of host immunity on gut microbial ecology.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Bacterias/genética , Colitis/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inflamasomas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD , Células Cultivadas , Colitis/inducido químicamente , Colitis/microbiología , Disbiosis/microbiología , Femenino , Antecedentes Genéticos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota , ARN Ribosómico 16S/análisis , Receptores de Superficie Celular/genética , Dodecil Sulfato de Sodio
11.
Proc Natl Acad Sci U S A ; 113(50): 14384-14389, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911804

RESUMEN

Familial Mediterranean fever (FMF) is the most common monogenic autoinflammatory disease worldwide. It is caused by mutations in the inflammasome adaptor Pyrin, but how FMF mutations alter signaling in FMF patients is unknown. Herein, we establish Clostridium difficile and its enterotoxin A (TcdA) as Pyrin-activating agents and show that wild-type and FMF Pyrin are differentially controlled by microtubules. Diverse microtubule assembly inhibitors prevented Pyrin-mediated caspase-1 activation and secretion of IL-1ß and IL-18 from mouse macrophages and human peripheral blood mononuclear cells (PBMCs). Remarkably, Pyrin inflammasome activation persisted upon microtubule disassembly in PBMCs of FMF patients but not in cells of patients afflicted with other autoinflammatory diseases. We further demonstrate that microtubules control Pyrin activation downstream of Pyrin dephosphorylation and that FMF mutations enable microtubule-independent assembly of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) micrometer-sized perinuclear structures (specks). The discovery that Pyrin mutations remove the obligatory requirement for microtubules in inflammasome activation provides a conceptual framework for understanding FMF and enables immunological screening of FMF mutations.


Asunto(s)
Fiebre Mediterránea Familiar/genética , Fiebre Mediterránea Familiar/metabolismo , Inflamasomas/metabolismo , Mutación , Pirina/genética , Pirina/metabolismo , Animales , Toxinas Bacterianas/toxicidad , Proteínas Adaptadoras de Señalización CARD/metabolismo , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/metabolismo , Enterotoxinas/toxicidad , Fiebre Mediterránea Familiar/inmunología , Células HEK293 , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microtúbulos/efectos de los fármacos , Microtúbulos/inmunología , Microtúbulos/metabolismo , Pirina/inmunología , Tubulina (Proteína)/metabolismo
14.
EMBO Mol Med ; 7(8): 1004-17, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25995337

RESUMEN

TNF is a central actor during inflammation and a well-recognized drug target for inflammatory diseases. We found that the mouse strain SPRET/Ei, known for extreme and dominant resistance against TNF-induced shock, displays weak expression of TNF receptor 1 protein (TNFR1) but normal mRNA expression, a trait genetically linked to the major TNFR1 coding gene Tnfrsf1a and to a locus harbouring the predicted TNFR1-regulating miR-511. This miRNA is a genuine TNFR1 regulator in cells. In mice, overexpression of miR-511 down-regulates TNFR1 and protects against TNF, while anti-miR-511 up-regulates TNFR1 and sensitizes for TNF, breaking the resistance of SPRET/Ei. We found that miR-511 inhibits endotoxemia and experimental hepatitis and that this miR is strongly induced by glucocorticoids and is a true TNFR1 modulator and thus an anti-inflammatory miR. Since minimal reductions of TNFR1 have considerable effects on TNF sensitivity, we believe that at least part of the anti-inflammatory effects of glucocorti-coids are mediated by induction of this miR, resulting in reduced TNFR1 expression.


Asunto(s)
Glucocorticoides/metabolismo , MicroARNs/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Regulación hacia Abajo , Ratones
15.
J Biol Chem ; 290(7): 4022-37, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25538244

RESUMEN

The cytokine TNF is a well known drug target for several inflammatory diseases such as Crohn disease. Despite the great success of TNF blockers, therapy could be improved because of high costs and side effects. Selective inhibition of TNF receptor (TNFR) 1 signaling holds the potential to greatly reduce the pro-inflammatory activity of TNF, thereby preserving the advantageous immunomodulatory signals mediated by TNFR2. We generated a selective human TNFR1 inhibitor based on Nanobody (Nb) technology. Two anti-human TNFR1 Nbs were linked with an anti-albumin Nb to generate Nb Alb-70-96 named "TNF Receptor-One Silencer" (TROS). TROS selectively binds and inhibits TNF/TNFR1 and lymphotoxin-α/TNFR1 signaling with good affinity and IC50 values, both of which are in the nanomolar range. Surface plasmon resonance analysis reveals that TROS competes with TNF for binding to human TNFR1. In HEK293T cells, TROS strongly reduces TNF-induced gene expression, like IL8 and TNF, in a dose-dependent manner; and in ex vivo cultured colon biopsies of CD patients, TROS inhibits inflammation. Finally, in liver chimeric humanized mice, TROS antagonizes inflammation in a model of acute TNF-induced liver inflammation, reflected in reduced human IL8 expression in liver and reduced IL6 levels in serum. These results demonstrate the considerable potential of TROS and justify the evaluation of TROS in relevant disease animal models of both acute and chronic inflammation and eventually in patients.


Asunto(s)
Colon/efectos de los fármacos , Enfermedad de Crohn/prevención & control , Inflamación/prevención & control , Hígado/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Anticuerpos de Dominio Único/farmacología , Secuencia de Aminoácidos , Animales , Colon/inmunología , Colon/patología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Femenino , Humanos , Inflamación/inmunología , Inflamación/patología , Hígado/inmunología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Datos de Secuencia Molecular , Conformación Proteica , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Resonancia por Plasmón de Superficie , Factor de Necrosis Tumoral alfa/farmacología
16.
FEBS J ; 281(20): 4568-82, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25154302

RESUMEN

Nucleotide-binding oligomerization domain receptors (NOD-like receptors, NLRs) are intracellular proteins that are chiefly known for their critical functions in inflammatory responses and host defense against microbial pathogens. Several NLRs have been demonstrated to assemble inflammasomes or to engage transcriptional signaling cascades that result in the production of pro-inflammatory cytokines and bactericidal factors. In recent years, NLRs have also emerged as key regulators of early mammalian embryogenesis and reproduction. A subset of phylogenetically related NLRs represents a new class of maternal effect genes that are highly expressed in maturing oocytes and pre-implantation embryos. Mutations in several of these NLRs have been linked to hereditary reproductive defects and imprinting diseases. In this review, we discuss the expression profiles, the emerging functions and molecular mode of action of these NLRs with newly recognized roles at the interfaces of the immune and reproductive systems. In addition, we provide an overview of coding mutations in NLRs that have been associated with human reproductive diseases, and outline crucial outstanding questions in this emerging research field.


Asunto(s)
Inmunidad Innata/inmunología , Proteínas Adaptadoras de Señalización NOD/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Reproducción/fisiología , Animales , Humanos , Proteínas Adaptadoras de Señalización NOD/inmunología , Receptores Citoplasmáticos y Nucleares/inmunología , Transducción de Señal
17.
Cytokine Growth Factor Rev ; 25(3): 285-300, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24746195

RESUMEN

TNF is an essential regulator of the immune system. Dysregulation of TNF plays a role in the pathology of many auto-immune diseases. TNF-blocking agents have proven successful in the treatment of such diseases. Development of novel, safer or more effective drugs requires a deeper understanding of the regulation of the pro-inflammatory activities of TNF and its receptors. The ubiquitously expressed TNFR1 is responsible for most TNF effects, while TNFR2 has a limited expression pattern and performs immune-regulatory functions. Despite extensive knowledge of TNFR1 signaling, the regulation of TNFR1 expression, its modifications, localization and processing are less clear and the data are scattered. Here we review the current knowledge of TNFR1 regulation and discuss the impact this has on the host.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Regulación de la Expresión Génica , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
18.
Int J Cancer ; 135(3): 742-50, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24382818

RESUMEN

Tumor necrosis factor (TNF) has remarkable antitumor effects, but its systemic therapeutic use is prevented by its lethal inflammatory effects. TNFR1 (P55) is essential for both the antitumor and toxic effects because both of them are absent in P55-deficient mice. In previous work we demonstrated that P55+/- mice are completely resistant to TNF toxicity, while the antitumor effects induced by TNF combined with interferon gamma (IFNγ) remain fully functional in these mice. Hence, a high dose of TNF/IFNγ has an excellent therapeutic potential when P55 levels are reduced, because TNF induces tumor regression without systemic toxicity. Here, we provide proof of principle for therapeutic application of this approach by using antisense oligonucleotides (ASOs). Treatment of mice with ASOs targeting P55 resulted in a strong reduction in P55 protein levels in liver, small intestine and blood mononuclear cells. This P55 downregulation was associated with significant protection of mice against acute TNF toxicity as measured by hypothermia, systemic inflammation and lethality. This treatment also protected mice against toxicity of TNF/IFNγ treatment in several cancer models: B16Bl6, Lewis lung carcinoma and a lung colony model. Our results confirm the therapeutic value of this strategy, which could lead to the development of a safer and more effective TNF/IFNγ antitumor therapy.


Asunto(s)
Carcinoma Pulmonar de Lewis/prevención & control , Modelos Animales de Enfermedad , Interferón gamma/toxicidad , Melanoma Experimental/prevención & control , Oligonucleótidos Antisentido/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Carcinoma Pulmonar de Lewis/inducido químicamente , Carcinoma Pulmonar de Lewis/genética , Femenino , Intestino Delgado/metabolismo , Hígado/metabolismo , Dosis Máxima Tolerada , Melanoma Experimental/inducido químicamente , Melanoma Experimental/genética , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Transducción de Señal
19.
EMBO Mol Med ; 5(7): 1000-16, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23723167

RESUMEN

Several pathological processes, such as sepsis and inflammatory bowel disease (IBD), are associated with impairment of intestinal epithelial barrier. Here, we investigated the role of matrix metalloproteinase MMP13 in these diseases. We observed that MMP13(-/-) mice display a strong protection in LPS- and caecal ligation and puncture-induced sepsis. We could attribute this protection to reduced LPS-induced goblet cell depletion, endoplasmic reticulum stress, permeability and tight junction destabilization in the gut of MMP13(-/-) mice compared to MMP13(+/+) mice. Both in vitro and in vivo, we found that MMP13 is able to cleave pro-TNF into bioactive TNF. By LC-MS/MS, we identified three MMP13 cleavage sites, which proves that MMP13 is an alternative TNF sheddase next to the TNF converting enzyme TACE. Similarly, we found that the same mechanism was responsible for the observed protection of the MMP13(-/-) mice in a mouse model of DSS-induced colitis. We identified MMP13 as an important mediator in sepsis and IBD via the shedding of TNF. Hence, we propose MMP13 as a novel drug target for diseases in which damage to the gut is essential.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Metaloproteinasa 13 de la Matriz/inmunología , Sepsis/inmunología , Sepsis/patología , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/inmunología , Colitis/patología , Sulfato de Dextran , Estrés del Retículo Endoplásmico , Femenino , Eliminación de Gen , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Lipopolisacáridos , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Sepsis/inducido químicamente , Sepsis/genética , Uniones Estrechas/inmunología , Uniones Estrechas/patología , Factor de Necrosis Tumoral alfa/inmunología
20.
J Clin Invest ; 123(6): 2590-603, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23676465

RESUMEN

TNF has remarkable antitumor activities; however, therapeutic applications have not been possible because of the systemic and lethal proinflammatory effects induced by TNF. Both the antitumor and inflammatory effects of TNF are mediated by the TNF receptor p55 (p55TNFR) (encoded by the Tnfrsf1a gene). The antitumor effect stems from an induction of cell death in tumor endothelium, but the cell type that initiates the lethal inflammatory cascade has been unclear. Using conditional Tnfrsf1a knockout or reactivation mice, we found that the expression level of p55TNFR in intestinal epithelial cells (IECs) is a crucial determinant in TNF-induced lethal inflammation. Remarkably, tumor endothelium and IECs exhibited differential sensitivities to TNF when p55TNFR levels were reduced. Tumor-bearing Tnfrsf1a⁺⁺/⁻ or IEC-specific p55TNFR-deficient mice showed resistance to TNF-induced lethality, while the tumor endothelium remained fully responsive to TNF-induced apoptosis and tumors regressed. We demonstrate proof of principle for clinical application of this approach using neutralizing anti-human p55TNFR antibodies in human TNFRSF1A knockin mice. Our results uncover an important cellular basis of TNF toxicity and reveal that IEC-specific or systemic reduction of p55TNFR mitigates TNF toxicity without loss of antitumor efficacy.


Asunto(s)
Antineoplásicos/farmacología , Mucosa Intestinal/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis , Citocinas/genética , Citocinas/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Endotelio/patología , Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Inflamación/inducido químicamente , Interferón gamma/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/uso terapéutico , Factor de Necrosis Tumoral alfa/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA