Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35163099

RESUMEN

De novo somatic mutations are well documented in diseases such as neoplasia but are rarely reported in rare diseases. Hovewer, severe genetic diseases that are not compatible with embryonic development are caused exclusively by deleterious mutations that could only be found as mosaic and not as inherited mutations. We will review here the paradigmatic case of Incontinentia Pigmenti, a rare X-linked dominant disease caused by deficiency of the NEMO (also called IKKgamma) protein, which plays a pivotal role in tissue homeostasis. The loss-of-function mutations of NEMO are embryonically lethal in males while females survive because of unbalanced X-inactivation due to NEMO wild type (WT) expressing cells survival despite of NEMO mutant expressing cells. The few surviving IP males are obligatory mosaic mutants with the typical clinical presentation of IP in female. Indeed, the IP pathogenesis in the female and most likely also in the male somatic mosaics is based on the cellular effects of an impaired NEMO activity, but in the context of the interaction of genetically different cells in the affected tissue, which might underline the inflammatory status.


Asunto(s)
Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Incontinencia Pigmentaria/patología , Mutación con Pérdida de Función , Mosaicismo , Humanos , Incontinencia Pigmentaria/etiología , Incontinencia Pigmentaria/metabolismo , Masculino
2.
Biochem Biophys Res Commun ; 589: 267-274, 2022 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-34933200

RESUMEN

The deprivation of myocardial nutrition causes cardiomyocyte death and disturbance of energy metabolism. IKKε plays an important regulatory role in many biological events such as inflammation, redox reaction, cell death, etc. However, the more in-depth mechanism by which IKKε contributes to cardiomyocytes death in nutrition deprivation remains poorly understood. IKKε expression was knocked down by siRNA in H9c2 cells, and cells were cultured under starvation conditions to simulate ischemic conditions. Starvation triggered greater NLRP3 activation, accompanied by more IL-1ß, IL-18 and caspase-1 release in the siIKKε H9c2 cells compared with the control H9c2 cells. Western blot and immunofluorescence showed that the IKKε konckdown promoted NLRP3 expressions and ROS release under starvation conditions. Furthermore, electron micrography and JC-1 analysis revealed that IKKε konckdown resulted in aggravated mitochondrial damage and more mitochondrial ROS (mtROS) released in vitro. Notably, Western blot analysis showed that IKKε deficiency activated the TBK1 and IRF3 signaling pathways to promote pyroptosis in vitro. Collectively, our results indicate that IKKε protects against cardiomyocyte injury by reducing mitochondrial damage and NLRP3 expression following nutrition deprivation via regulation of the TBK1/IRF3 signaling pathway. This study further revealed the mechanism of IKKε in inflammation and myocardial nutrition deprivation.


Asunto(s)
Citoprotección , Quinasa I-kappa B/metabolismo , Inflamasomas/metabolismo , Mitocondrias/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Quinasa I-kappa B/deficiencia , Factor 3 Regulador del Interferón/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas
3.
J Immunol Res ; 2021: 5521051, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34917688

RESUMEN

Aims. Acute kidney injury (AKI) can lead to chronic kidney disease (CKD), and macrophages play a key role in this process. The aim of this study was to discover the role of IκB kinase α (IKKα) in macrophages in the process of AKI-to-CKD transition. Main Methods. We crossed lyz2-Cre mice with IKKα-floxed mice to generate mice with IKKα ablation in macrophages (Mac IKKα-/-). A mouse renal ischemia/reperfusion injury (IRI) model was induced by clamping the renal artery for 45 minutes. Treated mice were evaluated for blood biochemistry, tissue histopathology, and fibrosis markers. Macrophages were isolated from the peritoneal cavity for coculturing with tubular epithelial cells (TECs) and flow cytometry analysis. Key Findings. We found that fibrosis and kidney function loss after IRI were significantly alleviated in Mac IKKα-/- mice compared with wild-type (WT) mice. The expression of fibrosis markers and the infiltration of M2 macrophages were decreased in the kidneys of Mac IKKα-/- mice after IRI. The in vitro experiment showed that the IRI TECs cocultured with IKKα-/- macrophages (KO MΦs) downregulated the fibrosis markers accompanied by a downregulation of Wnt/ß-catenin signaling. Significance. These data support the hypothesis that IKKα is involved in mediating macrophage polarization and increasing the expression of fibrosis-promoting inflammatory factors in macrophages. Therefore, knockdown of IKKα in macrophages may be a potential method that can be used to alleviate the AKI-to-CKD transition after IRI.


Asunto(s)
Quinasa I-kappa B/deficiencia , Túbulos Renales/patología , Macrófagos/inmunología , Daño por Reperfusión/inmunología , Animales , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales , Fibrosis , Técnicas de Silenciamiento del Gen , Humanos , Quinasa I-kappa B/genética , Túbulos Renales/irrigación sanguínea , Activación de Macrófagos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Cultivo Primario de Células , Daño por Reperfusión/complicaciones , Daño por Reperfusión/patología , Vía de Señalización Wnt/inmunología
4.
Biochem Biophys Res Commun ; 550: 184-190, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33706102

RESUMEN

Linear ubiquitination is an atypic ubiquitination process that directly connects the N- and C-termini of ubiquitin and is catalyzed by HOIL-1-interacting protein (HOIP). It is involved in the immune response or apoptosis by activating the nuclear factor-κB pathway and is associated with polyglucosan body myopathy 1, an autosomal recessive disorder with progressive muscle weakness and cardiomyopathy. However, little is currently known regarding the function of linear ubiquitination in muscles. Here, we investigated the role of linear ubiquitin E3 ligase (LUBEL), a DrosophilaHOIP ortholog, in the development and aging of muscles. The muscles of the flies with down-regulation of LUBEL or its downstream factors, kenny and Relish, developed normally, and there were no obvious abnormalities in function in young flies. However, the locomotor activity of the LUBEL RNAi flies was reduced compared to age-matched control, while LUBEL RNAi did not affect the increased mitochondrial fusion or myofiber disorganization during aging. Interestingly, the accumulation of polyubiquitinated protein aggregation during aging decreased in muscles by silencing LUBEL, kenny, or Relish. Meanwhile, the levels of autophagy and global translation, which are implicated in the maintenance of proteostasis, did not change due to LUBEL down-regulation. In conclusion, we propose a new role of linear ubiquitination in proteostasis in the muscle aging.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Músculos/metabolismo , Proteostasis , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Autofagia , Regulación hacia Abajo , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimología , Silenciador del Gen , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/metabolismo , Locomoción , Masculino , Fuerza Muscular , Músculos/enzimología , FN-kappa B/metabolismo , Poliubiquitina/metabolismo , Agregado de Proteínas , Biosíntesis de Proteínas , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
5.
Oxid Med Cell Longev ; 2021: 6429197, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33628362

RESUMEN

I-κB kinase-ε (IKKε) is a member of the IKK complex and a proinflammatory regulator that is active in many diseases. Angiotensin II (Ang II) is a vasoconstricting peptide hormone, and Ang II-induced myocardial hypertrophy is a common cardiovascular disease that can result in heart failure. In this study, we sought to determine the role of IKKε in the development of Ang II-induced myocardial hypertrophy in mice. Wild-type (WT) and IKKε-knockout (IKKε-KO) mice were generated and infused with saline or Ang II for 8 weeks. We found that WT mouse hearts have increased IKKε expression after 8 weeks of Ang II infusion. Our results further indicated that IKKε-KO mice have attenuated myocardial hypertrophy and alleviated heart failure compared with WT mice. Additionally, Ang II-induced expression of proinflammatory and collagen factors was much lower in the IKKε-KO mice than in the WT mice. Apoptosis and pyroptosis were also ameliorated in IKKε-KO mice. Mechanistically, IKKε bound to extracellular signal-regulated kinase (ERK) and the mitogen-activated protein kinase p38, resulting in MAPK/ERK kinase (MEK) phosphorylation, and IKKε deficiency inhibited the phosphorylation of MEK-ERK1/2 and p38 in mouse heart tissues after 8 weeks of Ang II infusion. The findings of our study reveal that IKKε plays an important role in the development of Ang II-induced myocardial hypertrophy and may represent a potential therapeutic target for the management of myocardial hypertrophy.


Asunto(s)
Cardiomegalia/enzimología , Cardiomegalia/patología , Quinasa I-kappa B/deficiencia , Miocardio/patología , Angiotensina II , Animales , Colágeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Insuficiencia Cardíaca/complicaciones , Quinasa I-kappa B/metabolismo , Inflamación/complicaciones , Inflamación/patología , Sistema de Señalización de MAP Quinasas , Ratones Noqueados , Piroptosis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J Biol Chem ; 295(49): 16691-16699, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-32978258

RESUMEN

Autophagy plays critical roles in the maintenance of endothelial cells in response to cellular stress caused by blood flow. There is growing evidence that both cell adhesion and cell detachment can modulate autophagy, but the mechanisms responsible for this regulation remain unclear. Immunoglobulin and proline-rich receptor-1 (IGPR-1) is a cell adhesion molecule that regulates angiogenesis and endothelial barrier function. In this study, using various biochemical and cellular assays, we demonstrate that IGPR-1 is activated by autophagy-inducing stimuli, such as amino acid starvation, nutrient deprivation, rapamycin, and lipopolysaccharide. Manipulating the IκB kinase ß activity coupled with in vivo and in vitro kinase assays demonstrated that IκB kinase ß is a key serine/threonine kinase activated by autophagy stimuli and that it catalyzes phosphorylation of IGPR-1 at Ser220 The subsequent activation of IGPR-1, in turn, stimulates phosphorylation of AMP-activated protein kinase, which leads to phosphorylation of the major pro-autophagy proteins ULK1 and Beclin-1 (BECN1), increased LC3-II levels, and accumulation of LC3 punctum. Thus, our data demonstrate that IGPR-1 is activated by autophagy-inducing stimuli and in response regulates autophagy, connecting cell adhesion to autophagy. These findings may have important significance for autophagy-driven pathologies such cardiovascular diseases and cancer and suggest that IGPR-1 may serve as a promising therapeutic target.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Antígenos CD28/metabolismo , Adhesión Celular , Secuencias de Aminoácidos , Animales , Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Beclina-1/metabolismo , Antígenos CD28/química , Antígenos CD28/genética , Adhesión Celular/efectos de los fármacos , Células HEK293 , Humanos , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/farmacología , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación/efectos de los fármacos , Primates , ARN Guía de Kinetoplastida/metabolismo , Sirolimus/farmacología , Especificidad por Sustrato
7.
Artículo en Inglés | MEDLINE | ID: mdl-32117051

RESUMEN

Extracellular matrix mineralization is critical for osteogenesis, and its dysregulation could result in osteoporosis and vascular calcification. IKK/NF-κB activation inhibits differentiation of osteoblasts, and reduces extracellular matrix mineralization, however the underlying mechanisms are poorly understood. In this study, we used CRISPR/Cas9 system to permanently inactivate IKKß in preosteoblast cells and confirmed that such cells displayed dramatic increase in extracellular matrix mineralization associated with JNK phosphorylation. Such observation was also found in our study using IKKß-deficient primary murine osteoblasts. Interestingly, we found that in Ikbkb-/-Mapk8-/- or Ikbkb-/-Mapk9-/- double knockout cells, the enhanced mineralization caused by IKKß deficiency was completely abolished, and deletion of either Mapk8 or Mapk9 was sufficient to dampen c-Jun phosphorylation. In further experiments, we discovered that absence of JNK1 or JNK2 on IKKß-deficient background resulted in highly conserved transcriptomic alteration in response to osteogenic induction. Therefore, identification of the indispensable roles of JNK1 and JNK2 in activating c-Jun and promoting osteoblast differentiation on IKKß-deficient background provided novel insights into restoring homeostasis in extracellular matrix mineralization.


Asunto(s)
Calcificación Fisiológica/genética , Matriz Extracelular/metabolismo , Quinasa I-kappa B/genética , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Proteína Quinasa 9 Activada por Mitógenos/fisiología , Osteoblastos/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Células Cultivadas , Matriz Extracelular/genética , Técnicas de Inactivación de Genes , Quinasa I-kappa B/deficiencia , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/genética , Osteoblastos/fisiología , Osteogénesis/genética , Fosforilación/genética , Transducción de Señal/genética
8.
J Ethnopharmacol ; 255: 112760, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32173427

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The Chinese herbal medicine Qing-Dai (also known as Indigo naturalis) extracted from indigo-bearing plants including Baphicacanthus cusia (Ness) Bremek was previously reported to exhibit anti-psoriatic effects in topical treatment. TH17 was later established as a key player in the pathogenesis of psoriasis. We investigated the anti-TH17 effect of Indigo naturalis and its active compounds. The aim of this study is to evaluate the toxicity of Indigo naturalis (IN) and its derivatives on five cell types involved in psoriasis, and to study the anti-inflammatory mechanism for the toxicity. MATERIALS AND METHODS: Following the fingerprint and quantity analysis of indirubin, indigo, and tryptanthrin in IN extract, we used MTS kits to measure the anti-proliferative effect of IN and three active compounds on five different cell types identified in psoriatic lesions. Quantitative RT-PCR analysis was used to measure the expression of various genes identified in the activated keratinocytes and TH17 polarized gene expression in RORγt-expressing T cells. RESULTS: We showed that IN differentially inhibited the proliferation of keratinocytes and endothelial cells but not monocytes, fibroblasts nor Jurkat T cells. Among three active compounds identified in IN, tryptanthrin was the most potent compound to reduce their proliferation. In addition to differentially reducing IL6 and IL8 expression, both IN and tryptanthrin also potently decreased the expression of anti-microbial S100A9 peptide, CCL20 chemokine, IL1B and TNFA cytokines, independent of NF-κB-p65-activation. Their attenuating effect was also detected on the expression of signature cytokines or chemokines induced during RORγT-induced TH17 polarization. CONCLUSIONS: We were the first to confirm a direct anti-TH17 effect of both IN herbal extract and tryptanthrin.


Asunto(s)
Citocinas/metabolismo , Medicamentos Herbarios Chinos/farmacología , Inmunosupresores/farmacología , Mediadores de Inflamación/metabolismo , Psoriasis/prevención & control , Quinazolinas/farmacología , Piel/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Células Jurkat , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/metabolismo , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fenotipo , Psoriasis/genética , Psoriasis/inmunología , Psoriasis/metabolismo , Piel/inmunología , Piel/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Células U937
9.
Oxid Med Cell Longev ; 2020: 3602824, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32064021

RESUMEN

Abdominal aortic aneurysm (AAA) is a vascular disorder that is considered a chronic inflammatory disease. However, the precise molecular mechanisms involved in AAA have not been fully elucidated. Recently, significant progress has been made in understanding the function and mechanism of action of inhibitor of kappa B kinase epsilon (IKKε) in inflammatory and metabolic diseases. The angiotensin II- (Ang II-) induced or pharmacological inhibitors were established to test the effects of IKKε on AAA in vivo. After mice were continuously stimulated with Ang II for 28 days, morphologically, we found that knockout of IKKε reduced AAA formation and drastically reduced maximal diameter and severity. We also observed a decrease in elastin degradation and medial destruction, which were independent of systolic blood pressure or plasma cholesterol concentrations. Western blot analyses and immunohistochemical staining were carried out to measure IKKε expression in AAA tissues and cell lines. AAA phenotype of mice was measured by ultrasound and biochemical indexes. In zymography, immunohistology staining, immunofluorescence staining, and reactive oxygen species (ROS) analysis, TUNEL assay was used to examine the effects of IKKε on AAA progression in AAA mice. IKKε deficiency significantly inhibited inflammatory macrophage infiltration, matrix metalloproteinase (MMP) activity, ROS production, and vascular smooth muscle cell (VSMC) apoptosis. We used primary mouse aortic VSMC isolated from apolipoprotein E (Apoe) -/- and Apoe-/-IKKε -/- mice. Mechanistically, IKKε deficiency blunted the activation of the ERK1/2 pathway. The IKKε inhibitor, amlexanox, has the same impact in AAA. Our results demonstrate a critical role of IKKε in AAA formation induced by Ang II in Apoe-/- mice. Targeting IKKε may constitute a novel therapeutic strategy to prevent AAA progression.


Asunto(s)
Angiotensina II/toxicidad , Aneurisma de la Aorta Abdominal/metabolismo , Apoptosis/genética , Quinasa I-kappa B/deficiencia , Inflamación/metabolismo , Estrés Oxidativo/genética , Anciano , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/enzimología , Aneurisma de la Aorta Abdominal/fisiopatología , Apolipoproteínas E/deficiencia , Apoptosis/efectos de los fármacos , Elastina/metabolismo , Femenino , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Inflamación/genética , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Macrófagos/metabolismo , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Persona de Mediana Edad , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Especies Reactivas de Oxígeno/metabolismo
10.
Transplant Proc ; 52(2): 647-652, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32035679

RESUMEN

BACKGROUND: X-linked EDA-ID1 (ectodermal dysplasia, anhidrotic, with immunodeficiency 1, Online Mendelian Inheritance in Man [OMIM] 300291), or NEMO (nuclear factor kappa B essential modulator) deficiency syndrome, is caused by mutations in the IKBKG/NEMO gene. We report the case of a boy with EDA-ID1 who underwent allogeneic stem cell transplantation. METHODS: In early infancy, the patient developed an atypical, severe, initial manifestation resembling Omenn syndrome with infections, and he underwent allogeneic stem cell transplantation from an unrelated 9 of 10 HLA matched donor with a mismatch in the DQB1 allele after conditioning with treosulfan, fludarabine, thiotepa, and antithymocyte globulin (Grafalon). The post-transplant period was complicated by cytomegalovirus replication and mild, grade 2 graft vs host disease. Because of NEMO deficiency syndrome-associated enteropathy and continuous weight loss, parenteral nutrition was started and the patient was fed an elemental formula and a gluten-free diet. Over a period of 3 years, the patient had 7 incidents of blood stream infections caused by Staphylococci or gut-derived Gram-negative flora, with 1 incident of septic shock caused by Escherichia coli. The blood stream infection stopped after gastrointestinal tract decontamination was done once per month for 7-day courses alternately with rifaximin, vancomycin, and gentamicin sulfate. CONCLUSIONS: Patients with NEMO deficiency syndrome require very complex, multidisciplinary care, and immunodeficiency correction can only be observed as one of the critical points in patient care. Developmental problems, enteropathy with the need for intravenous hyperalimentation, and specific interventions for other clinical manifestations of multifaceted syndrome are needed for proper care.


Asunto(s)
Busulfano/análogos & derivados , Displasia Ectodérmica/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Acondicionamiento Pretrasplante/métodos , Suero Antilinfocítico/uso terapéutico , Busulfano/uso terapéutico , Humanos , Quinasa I-kappa B/deficiencia , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Lactante , Masculino , Tiotepa/uso terapéutico , Trasplante Homólogo , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico
11.
J Hepatol ; 72(5): 960-975, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31954207

RESUMEN

BACKGROUND & AIMS: Hepatic innate immune control of viral infections has largely been attributed to Kupffer cells, the liver-resident macrophages. However, hepatocytes, the parenchymal cells of the liver, also possess potent immunological functions in addition to their known metabolic functions. Owing to their abundance in the liver and known immunological functions, we aimed to investigate the direct antiviral mechanisms employed by hepatocytes. METHODS: Using lymphocytic choriomeningitis virus (LCMV) as a model of liver infection, we first assessed the role of myeloid cells by depletion prior to infection. We investigated the role of hepatocyte-intrinsic innate immune signaling by infecting mice lacking canonical NF-κB signaling (IkkßΔHep) specifically in hepatocytes. In addition, mice lacking hepatocyte-specific interferon-α/ß signaling-(IfnarΔHep), or interferon-α/ß signaling in myeloid cells-(IfnarΔMyel) were infected. RESULTS: Here, we demonstrate that LCMV activates NF-κB signaling in hepatocytes. LCMV-triggered NF-κB activation in hepatocytes did not depend on Kupffer cells or TNFR1 signaling but rather on Toll-like receptor signaling. LCMV-infected IkkßΔHep livers displayed strongly elevated viral titers due to LCMV accumulation within hepatocytes, reduced interferon-stimulated gene (ISG) expression, delayed intrahepatic immune cell influx and delayed intrahepatic LCMV-specific CD8+ T cell responses. Notably, viral clearance and ISG expression were also reduced in LCMV-infected primary hepatocytes lacking IKKß, demonstrating a hepatocyte-intrinsic effect. Similar to livers of IkkßΔHep mice, enhanced hepatocytic LCMV accumulation was observed in livers of IfnarΔHep mice, whereas IfnarΔMyel mice were able to control LCMV infection. Hepatocytic NF-κB signaling was also required for efficient ISG induction in HDV-infected dHepaRG cells and interferon-α/ß-mediated inhibition of HBV replication in vitro. CONCLUSIONS: Together, these data show that hepatocyte-intrinsic NF-κB is a vital amplifier of interferon-α/ß signaling, which is pivotal for strong early ISG responses, immune cell infiltration and hepatic viral clearance. LAY SUMMARY: Innate immune cells have been ascribed a primary role in controlling viral clearance upon hepatic infections. We identified a novel dual role for NF-κB signaling in infected hepatocytes which was crucial for maximizing interferon responses and initiating adaptive immunity, thereby efficiently controlling hepatic virus replication.


Asunto(s)
Hepacivirus/genética , Hepatitis C Crónica/genética , Hepatitis C Crónica/inmunología , Hepatocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Subunidad p50 de NF-kappa B/genética , Polimorfismo de Nucleótido Simple , Factor de Transcripción ReIA/metabolismo , Replicación Viral/genética , Adulto , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Genotipo , Hepatitis C Crónica/virología , Humanos , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Coriomeningitis Linfocítica/virología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Adulto Joven
12.
Life Sci Alliance ; 2(6)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31792060

RESUMEN

Through the progressive accumulation of genetic and epigenetic alterations in cellular physiology, non-small-cell lung cancer (NSCLC) evolves in distinct steps involving mutually exclusive oncogenic mutations in K-Ras or EGFR along with inactivating mutations in the p53 tumor suppressor. Herein, we show two independent in vivo lung cancer models in which CHUK/IKK-α acts as a major NSCLC tumor suppressor. In a novel transgenic mouse strain, wherein IKKα ablation is induced by tamoxifen (Tmx) solely in alveolar type II (AT-II) lung epithelial cells, IKKα loss increases the number and size of lung adenomas in response to the chemical carcinogen urethane, whereas IKK-ß instead acts as a tumor promoter in this same context. IKKα knockdown in three independent human NSCLC lines (independent of K-Ras or p53 status) enhances their growth as tumor xenografts in immune-compromised mice. Bioinformatics analysis of whole transcriptome profiling followed by quantitative protein and targeted gene expression validation experiments reveals that IKKα loss can result in the up-regulation of activated HIF-1-α protein to enhance NSCLC tumor growth under hypoxic conditions in vivo.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quinasa I-kappa B/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Receptores ErbB/genética , Femenino , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Quinasa I-kappa B/deficiencia , Neoplasias Pulmonares/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regulación hacia Arriba , Proteínas ras/genética
14.
Front Immunol ; 10: 2062, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552024

RESUMEN

The colonic microenvironment, stemming from microbial, immunologic, stromal, and epithelial factors, serves as an important determinant of the host response to enteric pathogenic colonization. Infection with the enteric bacterial pathogen Citrobacter rodentium elicits a strong mucosal Th1-mediated colitis and monocyte-driven inflammation activated via the classical NF-κB pathway. Research has focused on leukocyte-mediated signaling as the main driver for C. rodentium-induced colitis, however we hypothesize that epithelial cell NF-κB also contributes to the exacerbation of infectious colitis. To test this hypothesis, compartmentalized classical NF-κB defective mice, via the deletion of IKKß in either intestinal epithelial cells (IKKßΔIEC) or myeloid-derived cells (IKKßΔMY), and wild type (WT) mice were challenged with C. rodentium. Both pathogen colonization and colonic histopathology were significantly reduced in IKKß-deficient mice compared to WT mice. Interestingly, colonic IL-10, RegIIIγ, TNF-α, and iNOS gene expression were increased in IKKß-deficient mice in the absence of bacterial challenge. This was associated with increased p52, which is involved with activation of NF-κß through the alternative pathway. IKKß-deficient mice also had distinct differences in colonic tissue-associated and luminal microbiome that may confer protection against C. rodentium. Taken together, these data demonstrate that classical NF-κB signaling can lead to enhanced enteric pathogen colonization and resulting colonic histopathology.


Asunto(s)
Citrobacter rodentium/inmunología , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Infecciones por Enterobacteriaceae/etiología , Infecciones por Enterobacteriaceae/metabolismo , Microbioma Gastrointestinal , Quinasa I-kappa B/deficiencia , Animales , Colitis/etiología , Colitis/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados
16.
J Am Heart Assoc ; 8(12): e012009, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31203708

RESUMEN

Background Obesity-associated chronic inflammation has been known to contribute to atherosclerosis development, but the underlying mechanisms remain elusive. Recent studies have revealed novel functions of IKK ß (inhibitor of NF -κB [nuclear factor κB] kinase ß), a key coordinator of inflammation through activation of NF -κB, in atherosclerosis and adipose tissue development. However, it is not clear whether IKK ß signaling in adipocytes can also affect atherogenesis. This study aims to investigate the impact of adipocyte IKK ß expression on atherosclerosis development in lean and obese LDLR (low-density lipoprotein receptor)-deficient ( LDLR -/-) mice. Methods and Results To define the role of adipocyte IKK ß in atherogenesis, we generated adipocyte-specific IKK ß-deficient LDLR -/- ( IKK ßΔAd LDLR -/-) mice. Targeted deletion of IKK ß in adipocytes did not affect adiposity and atherosclerosis in lean LDLR -/- mice when fed a low-fat diet. In response to high-fat feeding, however, IKK ßΔAd LDLR -/- mice had defective adipose remodeling and increased adipose tissue and systemic inflammation. Deficiency of adipocyte IKK ß did not affect atherosclerotic lesion sizes but resulted in enhanced lesional inflammation and increased plaque vulnerability in obese IKK ßΔAd LDLR -/- mice. Conclusions These data demonstrate that adipocyte IKK ß signaling affects the evolution of atherosclerosis plaque vulnerability in obese LDLR -/- mice. This study suggests that the functions of IKK ß signaling in atherogenesis are complex, and IKK ß in different cell types or tissues may have different effects on atherosclerosis development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adipocitos/enzimología , Quinasa I-kappa B/deficiencia , Placa Aterosclerótica/etiología , Animales , Masculino , Ratones Obesos
17.
Am J Med Genet A ; 179(8): 1415-1419, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31119873

RESUMEN

Incontinentia Pigmenti (IP; MIM 308300) is an X-linked dominant genodermatosis caused by pathogenic variant in IKBKG. The phenotype in adults is poorly described compared to that in children. Questionnaire survey of 99 affected women showed an age at diagnosis from newborn to 41 years, with 53 diagnosed by 6 months of age and 30 as adults. Stage I, II, and III lesions persisted in 16%, 17%, and 71%, respectively, of those who had ever had them. IP is allelic to two forms of ectodermal dysplasia. Many survey respondents reported hypohidrosis and/or heat intolerance and most had Stage IV findings. This suggests that "Stage IV" may be congenitally dysplastic skin that becomes more noticeable with maturity. Fifty-one had dentures or implants with 26 having more invasive jaw or dental surgery. Half had wiry or uncombable hair. Seventy-three reported abnormal nails with 27 having long-term problems. Cataracts and retinal detachment were the reported causes of vision loss. Four had microphthalmia. Respondents without genetic confirmation of IP volunteered information suggesting more involved phenotype or possibly misassigned diagnosis. Ascertainment bias likely accounts for the low prevalence of neurocognitive problems in the respondents.


Asunto(s)
Catarata/genética , Displasia Ectodérmica/genética , Quinasa I-kappa B/genética , Incontinencia Pigmentaria/genética , Mutación , Desprendimiento de Retina/genética , Adolescente , Adulto , Anciano , Catarata/diagnóstico , Catarata/metabolismo , Catarata/patología , Implantes Dentales , Dentaduras , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/metabolismo , Displasia Ectodérmica/patología , Femenino , Expresión Génica , Cabello/metabolismo , Cabello/patología , Humanos , Quinasa I-kappa B/deficiencia , Incontinencia Pigmentaria/diagnóstico , Incontinencia Pigmentaria/metabolismo , Incontinencia Pigmentaria/patología , Persona de Mediana Edad , Uñas/metabolismo , Uñas/patología , Fenotipo , Desprendimiento de Retina/diagnóstico , Desprendimiento de Retina/metabolismo , Desprendimiento de Retina/patología , Índice de Severidad de la Enfermedad , Piel/metabolismo , Piel/patología , Encuestas y Cuestionarios , Diente/metabolismo , Diente/patología
18.
Environ Health Perspect ; 127(5): 57009, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31095431

RESUMEN

BACKGROUND: Pulmonary inflammation is believed to be central to the pathogenesis due to exposure to fine particulate matter with aerodynamic diameter [Formula: see text] ([Formula: see text]). This central role, however, has not yet been systemically examined. OBJECTIVE: In the present study, we exploited a lung epithelial cell-specific inhibitor [Formula: see text] kinase 2 (IKK2) knockout mouse model to determine the role of pulmonary inflammation in the pathophysiology due to exposure to diesel exhaust particulate matter (DEP). METHODS: [Formula: see text] (lung epithelial cell-specific IKK2 knockout, KO) and [Formula: see text] (wild-type, tgWT) mice were intratracheally instilled with either vehicle or DEP for 4 months, and their inflammatory response and glucose homeostasis were then assessed. RESULTS: In comparison with tgWT mice, lung epithelial cell-specific IKK2-deficient mice had fewer DEP exposure-induced bronchoalveolar lavage fluid immune cells and proinflammatory cytokines as well as fewer DEP exposure-induced circulating proinflammatory cytokines. Glucose and insulin tolerance tests revealed that lung epithelial cell-specific IKK2 deficiency resulted in markedly less DEP exposure-induced insulin resistance and greater glucose tolerance. Akt phosphorylation analyses of insulin-responsive tissues showed that DEP exposure primarily targeted hepatic insulin sensitivity. Lung epithelial cell-specific IKK2-deficient mice had significantly lower hepatic insulin resistance than tgWT mice had. Furthermore, this difference in insulin resistance was accompanied by consistent differences in hepatic insulin receptor substrate 1 serine phosphorylation and inflammatory marker expression. DISCUSSION: Our findings suggest that in a tissue-specific knockout mouse model, an IKK2-dependent pulmonary inflammatory response was essential for the development of abnormal glucose homeostasis due to exposure to DEP. https://doi.org/10.1289/EHP4591.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Glucosa/fisiología , Homeostasis , Quinasa I-kappa B/deficiencia , Material Particulado/toxicidad , Emisiones de Vehículos/toxicidad , Animales , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos
19.
Actas Dermosifiliogr (Engl Ed) ; 110(4): 273-278, 2019 May.
Artículo en Inglés, Español | MEDLINE | ID: mdl-30660327

RESUMEN

Incontinentia pigmenti (Bloch-Sulzberger syndrome) is a rare neuroectodermal dysplasia. It is an X-linked dominant disorder caused by mutations in the IKBKG/NEMO gene on Xq28. Approximately 80% of patients have a deletion of exons 4 to 10. Incontinentia pigmenti has an estimated incidence of 0.7 cases per 100,000 births. In hemizygous males, it is usually lethal, while in females, it has a wide spectrum of clinical manifestations. Incontinentia pigmenti is a multisystemic disease that invariably features skin changes. These changes are the main diagnostic criteria and they evolve in 4 stages, in association with other abnormalities affecting the central nervous system, eyes, teeth, mammary glands, hair, nails, skin, and other parts of the body. The aim of this brief review is to highlight the clinical features of this genodermatosis and underline the importance of case-by-case interdisciplinary management, including genetic counseling.


Asunto(s)
Incontinencia Pigmentaria , Diagnóstico Diferencial , Manejo de la Enfermedad , Femenino , Genes Ligados a X , Genotipo , Humanos , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/fisiología , Incontinencia Pigmentaria/epidemiología , Incontinencia Pigmentaria/genética , Incontinencia Pigmentaria/patología , Incontinencia Pigmentaria/terapia , Masculino , Especificidad de Órganos , Fenotipo , Medicina de Precisión , Eliminación de Secuencia , Piel/patología
20.
Neuropharmacology ; 146: 198-211, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30528326

RESUMEN

Inhibitor-kappaB kinase epsilon (IKKε, Ikbke) constitutes an NF-κB activating kinase with high homology to the classical I-κB kinase subunits, IKKα and IKKß. It is expressed in nociceptive neurons in the spinal cord and in dorsal root ganglia (DRG) and involved in inflammatory nociception. Under inflammatory conditions, IKKε deficient mice show significantly less nociceptive behavior in comparison to wild type mice associated with reduced activation of NF-κB and attenuated NF-κB-dependent gene expression. The role of IKKε in neuropathic pain has not been investigated so far. We applied the spared nerve injury (SNI) model of neuropathic pain in mice and found an increased expression of IKKε in the spinal cord, the DRGs and the sciatic nerve after induction of neuropathy. Genetic depletion of IKKε or pharmacological inhibition by amlexanox led to a significant reduction of mechanical hyperalgesia and cold allodynia in comparison to control mice. Transcription factor ELISA indicated that the effects are mediated by reduced activation of NF-κB. Furthermore, immunofluorescence staining, qPCR and Western Blot analyses revealed that the decreased pain-like behavior was associated with a reduced activation of microglia, diminished expression of c-fos as well as a decreased activation of MAP-Kinases. In summary, we conclude that IKKε modulates mechanisms of neuropathic pain by activating NF-κB. The administration of IKKε inhibitors might therefore constitute a new and promising approach for the therapy of neuropathic pain.


Asunto(s)
Aminopiridinas/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/deficiencia , Neuralgia/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/genética , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía , FN-kappa B/metabolismo , Neuralgia/inducido químicamente , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/metabolismo , Nervio Ciático/metabolismo , Transducción de Señal , Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA