Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Gut ; 66(10): 1748-1760, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27371534

RESUMEN

OBJECTIVE: Epidemiological and clinical data indicate that patients suffering from IBD with long-standing colitis display a higher risk to develop colorectal high-grade dysplasia. Whereas carcinoma invasion and metastasis rely on basement membrane (BM) disruption, experimental evidence is lacking regarding the potential contribution of epithelial cell/BM anchorage on inflammation onset and subsequent neoplastic transformation of inflammatory lesions. Herein, we analyse the role of the α6ß4 integrin receptor found in hemidesmosomes that attach intestinal epithelial cells (IECs) to the laminin-containing BM. DESIGN: We developed new mouse models inducing IEC-specific ablation of α6 integrin either during development (α6ΔIEC) or in adults (α6ΔIEC-TAM). RESULTS: Strikingly, all α6ΔIEC mutant mice spontaneously developed long-standing colitis, which degenerated overtime into infiltrating adenocarcinoma. The sequence of events leading to disease onset entails hemidesmosome disruption, BM detachment, IL-18 overproduction by IECs, hyperplasia and enhanced intestinal permeability. Likewise, IEC-specific ablation of α6 integrin induced in adult mice (α6ΔIEC-TAM) resulted in fully penetrant colitis and tumour progression. Whereas broad-spectrum antibiotic treatment lowered tissue pathology and IL-1ß secretion from infiltrating myeloid cells, it failed to reduce Th1 and Th17 response. Interestingly, while the initial intestinal inflammation occurred independently of the adaptive immune system, tumourigenesis required B and T lymphocyte activation. CONCLUSIONS: We provide for the first time evidence that loss of IECs/BM interactions triggered by hemidesmosome disruption initiates the development of inflammatory lesions that progress into high-grade dysplasia and carcinoma. Colorectal neoplasia in our mouse models resemble that seen in patients with IBD, making them highly attractive for discovering more efficient therapies.


Asunto(s)
Adenocarcinoma/fisiopatología , Colitis/fisiopatología , Neoplasias Colorrectales/fisiopatología , Citocinas/metabolismo , Hemidesmosomas/fisiología , Integrina alfa6/genética , Integrina alfa6beta4/metabolismo , Mucosa Intestinal/metabolismo , Inmunidad Adaptativa , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Linfocitos B , Membrana Basal/fisiopatología , Caspasa 1/metabolismo , Colitis/genética , Colitis/metabolismo , Colitis/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Citocinas/genética , Células Epiteliales/metabolismo , Hemidesmosomas/genética , Homeostasis/genética , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Queratina-18/metabolismo , Queratina-8/metabolismo , Activación de Linfocitos , Ratones , Moco/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Permeabilidad , Índice de Severidad de la Enfermedad , Transducción de Señal , Linfocitos T
2.
Am J Respir Crit Care Med ; 189(7): 799-811, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24555512

RESUMEN

RATIONALE: Pseudomonas aeruginosa, a major problem pathogen responsible for severe infections in critically ill patients, triggers, through a functional type-3 secretion system (T3SS), the activation of an intracellular cytosolic sensor of innate immunity, NLRC4. Although the NLRC4-inflammasome-dependent response contributes to increased clearance of intracellular pathogens, it seems that NLRC4 inflammasome activation decreases the clearance of P. aeruginosa, a mainly extracellular pathogen. OBJECTIVES: We sought to determine the underlying mechanisms of this effect of the activation of NLRC4 by P. aeruginosa. METHODS: We established acute lung injury in wild-type and Nlrc4(-/-) mice using sublethal intranasal inocula of P. aeruginosa strain CHA expressing or not a functional T3SS. We studied 96-hour survival, lung injury, bacterial clearance from the lungs, cytokine secretion in bronchoalveolar lavage, lung antimicrobial peptide expression by quantitative polymerase chain reaction, and flow cytometry analysis of lung cells. MEASUREMENTS AND MAIN RESULTS: Nlrc4(-/-) mice showed enhanced bacterial clearance and decreased lung injury contributing to increased survival against extracellular P. aeruginosa strain expressing a functional T3SS. The mechanism involved decreased NLRC4-inflammasome-driven IL-18 secretion attenuating lung injury caused by excessive neutrophil recruitment. Additionally, in the lungs of Nlrc4(-/-) mice secretion of IL-17 by innate immune cells was increased and responsible for increased expression of lung epithelial antimicrobial peptides. Furthermore, IL-18 secretion was found to repress IL-17 and IL-17-driven lung antimicrobial peptide expression. CONCLUSIONS: We report a new role of the T3SS apparatus itself, independently of exotoxin translocation. Through NLRC4 inflammasome activation, the T3SS promotes IL-18 secretion, which dampens a beneficial IL-17-mediated antimicrobial host response.


Asunto(s)
Lesión Pulmonar Aguda/microbiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Inflamasomas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Biomarcadores/metabolismo , Líquido del Lavado Bronquioalveolar/química , Caspasa 1/metabolismo , Células Cultivadas , Femenino , Citometría de Flujo , Inmunidad Innata , Interleucina-17/metabolismo , Interleucina-18/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Proc Natl Acad Sci U S A ; 108(23): 9601-6, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21593405

RESUMEN

The colonic epithelium self-renews every 3 to 5 d, but our understanding of the underlying processes preserving wound healing from carcinogenesis remains incomplete. Here, we demonstrate that Nod-like receptor pyrin domain-containing protein 6 (NLRP6) suppresses inflammation and carcinogenesis by regulating tissue repair. NLRP6 was primarily produced by myofibroblasts within the stem-cell niche in the colon. Although NLRP6 expression was lowered in diseased colon, NLRP6-deficient mice were highly susceptible to experimental colitis. Upon injury, NLRP6 deficiency deregulated regeneration of the colonic mucosa and processes of epithelial proliferation and migration. Consistently, absence of NLRP6 accelerated colitis-associated tumor growth in mice. A gene-ontology analysis on a whole-genome expression profiling revealed a link between NLRP6 and self-renewal of the epithelium. Collectively, the integrity of the epithelial barrier is preserved by NLRP6 that may be manipulated to develop drugs capable of preventing adenoma formation in inflammatory bowel diseases.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/genética , Células Epiteliales/metabolismo , Receptores de Superficie Celular/genética , Animales , Movimiento Celular/genética , Colitis/genética , Colitis/metabolismo , Colitis/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Epiteliales/patología , Femenino , Perfilación de la Expresión Génica , Inmunohistoquímica , Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Superficie Celular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cicatrización de Heridas/genética
4.
Gut ; 62(12): 1714-23, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23172891

RESUMEN

OBJECTIVE: Inflammatory bowel diseases (IBD) have been intrinsically linked to a deregulated cytokine network, but novel therapeutic principles are urgently needed. Here we identify the interleukin (IL)-33 and its receptor ST2 as key negative regulators of wound healing and permeability in the colon of mice. DESIGN: Expression of IL-33 and ST2 was determined by qRT-PCR, ELISA, immunohistochemistry and western-blot analysis. Wild-type and St2(-/-) mice were used in wound healing experiments and in two experimental models of IBD triggered by 2,4,6-trinitrobenzene sulphonic acid or dextran sodium sulphate (DSS). Neutralisation of ST2 was performed by using a specific blocking antibody. RESULTS: Nuclear localisation and enhanced expression of IL-33 in myofibroblasts and enterocytes was linked to disease involvement independently of inflammation, while the expression of ST2 was primarily restricted to the colonic epithelia. In two experimental models of IBD, genetic ablation of ST2 significantly improved signs of colitis, while a sustained epithelial expression of the cyto-protective factor connexin-43 was observed in DSS-treated St2-deficient mice. Unexpectedly, absence of ST2 in non-hematopoietic cells was sufficient to protect against colitis. Consistently, specific inhibition of endogenous ST2-mediated signalling by treatment with neutralising antibody improved DSS-induced colitis. In addition, IL-33 treatment impaired epithelial barrier permeability in vitro and in vivo, whereas absence of ST2 enhanced wound healing response upon acute mechanical injury in the colon. CONCLUSIONS: Our study unveiled a novel non-hematopoietic function of IL-33 in epithelial barrier function and wound healing. Therefore, blocking the IL-33/ST2 axis may represent an efficient therapy in IBD.


Asunto(s)
Colitis Ulcerosa/etiología , Interleucinas/fisiología , Receptores de Interleucina/fisiología , Animales , Western Blotting , Células CACO-2 , Colitis Ulcerosa/fisiopatología , Colitis Ulcerosa/terapia , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Reacción en Cadena en Tiempo Real de la Polimerasa , Cicatrización de Heridas/fisiología
5.
Arthritis Rheum ; 63(7): 2142-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21480187

RESUMEN

OBJECTIVE: A new class of autoinflammatory syndromes called NLRP12-associated disorders (NLRP12AD) has been associated with mutations in NLRP12. Conflicting data on the putative role of NLRP12 in interleukin-1ß (IL-1ß) signaling have been found in in vitro analyses. This prospective study was undertaken to assess the secretion of IL-1ß and 3 IL-1ß-induced cytokines (IL-1 receptor antagonist [IL-1Ra], IL-6, and tumor necrosis factor α [TNFα]) in patients' peripheral blood mononuclear cells (PBMCs) cultured ex vivo and to evaluate the patients' response to IL-1Ra (anakinra), a major drug used in the treatment of autoinflammatory disorders. METHODS: Patients' disease manifestations and cytokine measurements were recorded before anakinra treatment was started, during 14 months of therapy, and after discontinuation of anakinra treatment. RESULTS: Spontaneous secretion of IL-1ß by patients' PBMCs was found to be dramatically increased (80-175 fold) compared to healthy controls. Consistent with these findings, anakinra initially led to a marked clinical improvement and to a rapid near-normalization of IL-1ß secretion. However, a progressive clinical relapse occurred secondarily, associated with an increase in TNFα secretion, persistent elevated levels of IL-1Ra and IL-6, and a reactivation of IL-1ß secretion. Anakinra was discontinued after 14 months of therapy. CONCLUSION: Our findings provide in vivo evidence of the crucial role of IL-1ß in the pathophysiology of NLRP12AD. This is the first time anakinra has been used to treat this disorder. This study provides new insights into the mechanisms underlying resistance to anti-IL-1 therapy observed in a few patients with autoinflammatory syndromes. Our data also point to the potential of ex vivo cytokine measurements as predictors of response to treatment.


Asunto(s)
Enfermedades Autoinflamatorias Hereditarias/inmunología , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ensayo de Inmunoadsorción Enzimática , Enfermedades Autoinflamatorias Hereditarias/tratamiento farmacológico , Enfermedades Autoinflamatorias Hereditarias/genética , Humanos , Masculino , Estudios Prospectivos , Transducción de Señal
6.
Bioorg Med Chem Lett ; 21(3): 1019-22, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21215621

RESUMEN

Several thalidomide analogues were synthesized and compared to thalidomide and its more active analogue, lenalidomide, for their ability to inhibit the production of the pro-inflammatory cytokine tumour necrosis factor (TNF)-α and interleukin (IL)-6 by LPS-activated peripheral blood mononuclear cells (PBMCs). Among these compounds, two analogues containing sulfonyl group displayed interesting downregulation of TNF-α and IL-6 production.


Asunto(s)
Interleucina-6/metabolismo , Talidomida/análogos & derivados , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Abajo , Diseño de Fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Talidomida/síntesis química , Talidomida/farmacología
7.
Arthritis Rheum ; 62(4): 1176-85, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20131254

RESUMEN

OBJECTIVE: To gain insight into the pathophysiology of an atypical familial form of an autoinflammatory disorder, characterized by autosomal-dominant sensorineural hearing loss, systemic inflammation, increased secretion of interleukin-1beta (IL-1beta), and the absence of any cutaneous manifestations, and to assess the functional consequences of a missense mutation identified in the leucine-rich repeat (LRR) domain of NLRP3. METHODS: Microsatellite markers were used to test the familial segregation of the NLRP3 locus with the disease phenotype. All NLRP3 exons were screened for mutations by sequencing. Functional assays were performed in HEK 293T cells to determine the effects of mutated (versus normal) NLRP3 proteins on NF-kappaB activation, caspase 1 signaling, and speck formation. RESULTS: A heterozygous NLRP3 missense mutation (p.Tyr859Cys) was identified in exon 6, which encodes the LRR domain of the protein. This mutation was found to segregate with the disease phenotype within the family, and had a moderate activating effect on speck formation and procaspase 1 processing and did not alter the inhibitory properties of NLRP3 on NF-kappaB signaling. CONCLUSION: This report is the first to describe a familial form of a cryopyrinopathy associated with a mutation outside of exon 3 of NLRP3. This finding, together with the known efficacy of anti-IL-1 treatments in these disorders, underlines the importance of screening all exons of NLRP3 in patients who present with atypical manifestations. In addition, the gain of function associated with this mutation in terms of activation of caspase 1 signaling was consistent with the observed inflammatory phenotype. Therefore, this study of the functional consequences of an LRR mutation sheds new light on the clinical relevance of in vitro assays.


Asunto(s)
Proteínas Portadoras/genética , Mutación de Línea Germinal , Enfermedades Autoinflamatorias Hereditarias/genética , Secuencia de Aminoácidos , Línea Celular , Citocinas/sangre , ADN/genética , Cartilla de ADN , Femenino , Variación Genética , Enfermedades Autoinflamatorias Hereditarias/fisiopatología , Humanos , Interleucina-1beta/sangre , Riñón/embriología , Masculino , Monocitos/patología , Madres , Mutación Missense , Proteína con Dominio Pirina 3 de la Familia NLR , Plásmidos , Hermanos
9.
Nat Commun ; 9(1): 5338, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30559449

RESUMEN

Mutations in the nucleotide-binding oligomerization domain protein 12 (NLRP12) cause recurrent episodes of serosal inflammation. Here we show that NLRP12 efficiently sequesters HSP90 and promotes K48-linked ubiquitination and degradation of NOD2 in response to bacterial muramyl dipeptide (MDP). This interaction is mediated by the linker-region proximal to the nucleotide-binding domain of NLRP12. Consequently, the disease-causing NLRP12 R284X mutation fails to repress MDP-induced NF-κB and subsequent activity of the JAK/STAT signaling pathway. While NLRP12 deficiency renders septic mice highly susceptible towards MDP, a sustained sensing of MDP through NOD2 is observed among monocytes lacking NLRP12. This loss of tolerance in monocytes results in greater colonization resistance towards Citrobacter rodentium. Our data show that this is a consequence of NOD2-dependent accumulation of inflammatory mononuclear cells that correlates with induction of interferon-stimulated genes. Our study unveils a relevant process of tolerance towards the gut microbiota that is exploited by an attaching/effacing enteric pathogen.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/metabolismo , Cápsulas Bacterianas/metabolismo , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Proteínas HSP90 de Choque Térmico/metabolismo , Tolerancia Inmunológica/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Línea Celular , Infecciones por Enterobacteriaceae/microbiología , Microbioma Gastrointestinal/inmunología , Células HEK293 , Humanos , Inflamación/inmunología , Inflamación/microbiología , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Ubiquitinación
10.
Gut Microbes ; 4(4): 353-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23778641

RESUMEN

The gut flora is composed of a huge number of diverse, well-adapted symbionts that interact with epithelial lining throughout the host's entire life. Not all commensals have the same ability to maintain quiescent, protective inflammation. Importantly, instability in the composition of gut microbial communities (referred to as dysbiosis) has been linked to loss of gut barrier in the context of common human illnesses with increasing socio-economic impacts, such as Crohn disease and colorectal cancer. Our recent findings suggest that disease-predisposing dysbiosis can now be intentionally manipulated by targeting the major Crohn disease-predisposing NOD2 gene. That knowledge will not only add a new dimension to the often overlooked microbiology of Crohn disease and colorectal cancer, but will also have a broad impact on biomedical sciences worldwide.


Asunto(s)
Colitis/etiología , Neoplasias Colorrectales/etiología , Proteína Adaptadora de Señalización NOD2/deficiencia , Animales , Femenino , Humanos , Masculino , Embarazo
11.
J Clin Invest ; 123(2): 700-11, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23281400

RESUMEN

Instability in the composition of gut bacterial communities (dysbiosis) has been linked to common human intestinal disorders, such as Crohn's disease and colorectal cancer. Here, we show that dysbiosis caused by Nod2 deficiency gives rise to a reversible, communicable risk of colitis and colitis-associated carcinogenesis in mice. Loss of either Nod2 or RIP2 resulted in a proinflammatory microenvironment that enhanced epithelial dysplasia following chemically induced injury. The condition could be improved by treatment with antibiotics or an anti-interleukin-6 receptor-neutralizing antibody. Genotype-dependent disease risk was communicable via maternally transmitted microbiota in both Nod2-deficient and WT hosts. Furthermore, reciprocal microbiota transplantation reduced disease risk in Nod2-deficient mice and led to long-term changes in intestinal microbial communities. Conversely, disease risk was enhanced in WT hosts that were recolonized with dysbiotic fecal microbiota from Nod2-deficient mice. Thus, we demonstrated that licensing of dysbiotic microbiota is a critical component of disease risk. Our results demonstrate that NOD2 has an unexpected role in shaping a protective assembly of gut bacterial communities and suggest that manipulation of dysbiosis is a potential therapeutic approach in the treatment of human intestinal disorders.


Asunto(s)
Colitis/etiología , Neoplasias Colorrectales/etiología , Proteína Adaptadora de Señalización NOD2/deficiencia , Animales , Colitis/metabolismo , Colitis/microbiología , Colitis/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Sistema Digestivo/metabolismo , Sistema Digestivo/microbiología , Sistema Digestivo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Metagenoma , Ratones , Ratones Noqueados , Proteína Adaptadora de Señalización NOD2/genética , Embarazo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factores de Riesgo
12.
Eur Cytokine Netw ; 20(3): 101-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19825518

RESUMEN

The mevalonate kinase deficiency (MKD), including hyperimmunoglobulinemia D periodic fever syndrome (HIDS) and the more severe mevalonic aciduria are rare, autosomal recessive, autoinflammatory diseases belonging to the hereditary periodic fever (HPF) family. Other members include: familial mediterranean fever (FMF), the cryopyrin-associated periodic syndromes (CAPS) and TNFR-associated periodic syndromes (TRAPS). MKD is caused by mutations in the gene encoding mevalonate kinase (MK), an enzyme of the cholesterol pathway, leading to its inactivation. The molecular mechanisms linking MKD and abnormalities of isoprenoid biosynthesis to cytokine production and inflammation have yet to be fully elucidated. Statins, which are extensively prescribed for lowering cholesterol, are potent inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase, the enzyme directly upstream of MK. In this review, we discuss recent reports demonstrating that in vitro inhibition of the mevalonate pathway by statins specifically increases the production, by activated monocytes, of cytokines of the IL-1 family, by enhancing caspase-1 activity, the enzyme responsible for IL-1beta and IL-18 maturation. The molecular mechanisms involve geranylgeranylation and the enhancement of the activity of G proteins such as Rac-1. Interestingly, activated fibroblasts from MKD patients secrete more IL-1beta than fibroblasts from healthy donors. Taken together, these data highlight the specific enhancement of the IL-1 family of cytokines, the maturation of which is caspase-1-dependent in MKD. Finally, the spectacular decrease in febrile attacks in patients with severe HIDS under IL-1 receptor antagonist (anakinra) treatment, reinforces this hypothesis. Deregulated caspase-1 activation could be responsible for the inflammatory component of MKD, thereby mechanistically linking MKD to FMF and CAPS through cytokines of the IL-1 family.


Asunto(s)
Caspasa 1/metabolismo , Inflamación/enzimología , Inflamación/inmunología , Interleucina-1/metabolismo , Deficiencia de Mevalonato Quinasa/enzimología , Deficiencia de Mevalonato Quinasa/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inflamación/complicaciones , Deficiencia de Mevalonato Quinasa/complicaciones , Deficiencia de Mevalonato Quinasa/tratamiento farmacológico
13.
Eur Cytokine Netw ; 20(3): 112-20, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19825520

RESUMEN

OBJECTIVE: The effects of statins (3-hydroxy-3-methylglutaryl coenzyme A reductase-HMGR-inhibitors) on the inflammatory response remain unclear. HMGR is implicated in the mevalonate pathway, directly upstream of cholesterol biosynthesis. We studied the impairment by this pathway of cytokine production by peripheral blood mononuclear cells (PBMCs) and THP-1 cells. The aim was to identify a specific cytokine "signature" of cells under simvastatin treatment in order to link pharmacological inhibition of the mevalonate pathway and inflammation. METHODS: Normal human PBMCs and THP-1 cells were cultured with inhibitors of HMGR (simvastatin), geranylgeranyltransferase (GGTI-298), farnesyltransferase (FTI-277), and/or caspase-1 (Z-VAD(Ome)-FMK). Following culture, cytokine production, caspase-1 activity, IL-1beta mRNA and Rac-1 activity were determined. RESULTS: Pharmacological inhibition of the mevalonate pathway specifically enhanced the release of IL-1alpha, IL-1beta and IL-18 and inhibited IL-1ra production by LPS-activated PBMCs and THP-1 cells. Simvastatin did not modify pro-IL-1beta expression, but enhanced caspase-1 activity, the enzyme responsible for IL-1beta and IL-18 maturation. GGTI-298 also enhanced IL-1-family cytokine production, showing that geranylgeranylation is involved in caspase-1 activation. Additionally, simvastatin enhanced Rac-1 activity. CONCLUSION: Pharmacological inhibition of the mevalonate pathway by statins highlighted the specific induction of the proinflammatory cytokines of the IL-1 family whose maturation is either directly (i.e. IL-1beta and IL-18), or indirectly (i.e. IL-1alpha) dependant on caspase-1.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Interleucina-1/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Ácido Mevalónico/metabolismo , Monocitos/metabolismo , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Caspasa 1/metabolismo , Línea Celular , Activación Enzimática/efectos de los fármacos , Guanosina Trifosfato/metabolismo , Humanos , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Monocitos/efectos de los fármacos , Monocitos/enzimología , Fosforilación/efectos de los fármacos , Prenilación de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Simvastatina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA