Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nat Immunol ; 13(12): 1178-86, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23104095

RESUMEN

We report the clinical description and molecular dissection of a new fatal human inherited disorder characterized by chronic autoinflammation, invasive bacterial infections and muscular amylopectinosis. Patients from two kindreds carried biallelic loss-of-expression and loss-of-function mutations in HOIL1 (RBCK1), a component of the linear ubiquitination chain assembly complex (LUBAC). These mutations resulted in impairment of LUBAC stability. NF-κB activation in response to interleukin 1ß (IL-1ß) was compromised in the patients' fibroblasts. By contrast, the patients' mononuclear leukocytes, particularly monocytes, were hyper-responsive to IL-1ß. The consequences of human HOIL-1 and LUBAC deficiencies for IL-1ß responses thus differed between cell types, consistent with the unique association of autoinflammation and immunodeficiency in these patients. These data suggest that LUBAC regulates NF-κB-dependent IL-1ß responses differently in different cell types.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo IV/genética , Enfermedades Autoinflamatorias Hereditarias/genética , Síndromes de Inmunodeficiencia/genética , FN-kappa B/metabolismo , Ubiquitina-Proteína Ligasas/genética , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Proteínas de Ciclo Celular/genética , Línea Celular , Fibroblastos/inmunología , Fibroblastos/metabolismo , Humanos , Síndromes de Inmunodeficiencia/metabolismo , Interleucina-1beta/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/genética , Factores de Transcripción , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
2.
Mol Cell ; 45(4): 553-66, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22365832

RESUMEN

Plk1 activation is required for progression through mitotic entry to cytokinesis. Here we show that at mitotic entry, Plk1 phosphorylates Optineurin (Optn) at serine 177 and that this dissociates Optn from the Golgi-localized GTPase Rab8, inducing its translocation into the nucleus. Mass spectrometry analysis revealed that Optn is associated with a myosin phosphatase complex (MP), which antagonizes the mitotic function of Plk1. Our data also indicate that Optn functionally connects this complex to Plk1 by promoting phosphorylation of the myosin phosphatase targeting subunit 1 (MYPT1). Accordingly, silencing Optn expression increases Plk1 activity and induces abscission failure and multinucleation, which were rescued upon expression of wild-type (WT) Optn, but not a phospho-deficient mutant (S177A) that cannot translocate into the nucleus during mitosis. Overall, these results highlight an important role of Optn in the spatial and temporal coordination of Plk1 activity.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Factor de Transcripción TFIIIA/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Retroalimentación Fisiológica , Células HEK293 , Células HeLa , Humanos , Proteínas de Transporte de Membrana , Fosforilación , Factor de Transcripción TFIIIA/química , Factor de Transcripción TFIIIA/fisiología , Quinasa Tipo Polo 1
3.
Hum Mol Genet ; 25(7): 1281-93, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26769674

RESUMEN

Patients with cystic fibrosis (CF) display low bone mass and alterations in bone formation. Mice carrying the F508del genetic mutation in the cystic fibrosis conductance regulator (Cftr) gene display reduced bone formation and decreased bone mass. However, the underlying molecular mechanisms leading to these skeletal defects are unknown, which precludes the development of an efficient anti-osteoporotic therapeutic strategy. Here we report a key role for the intermediate filament protein keratin 8 (Krt8), in the osteoblast dysfunctions in F508del-Cftr mice. We found that murine and human osteoblasts express Cftr and Krt8 at low levels. Genetic studies showed that Krt8 deletion (Krt8(-/-)) in F508del-Cftr mice increased the levels of circulating markers of bone formation, corrected the expression of osteoblast phenotypic genes, promoted trabecular bone formation and improved bone mass and microarchitecture. Mechanistically, Krt8 deletion in F508del-Cftr mice corrected overactive NF-κB signaling and decreased Wnt-ß-catenin signaling induced by the F508del-Cftr mutation in osteoblasts. In vitro, treatment with compound 407, which specifically disrupts the Krt8-F508del-Cftr interaction in epithelial cells, corrected the abnormal NF-κB and Wnt-ß-catenin signaling and the altered phenotypic gene expression in F508del-Cftr osteoblasts. In vivo, short-term treatment with 407 corrected the altered Wnt-ß-catenin signaling and bone formation in F508del-Cftr mice. Collectively, the results show that genetic or pharmacologic targeting of Krt8 leads to correction of osteoblast dysfunctions, altered bone formation and osteopenia in F508del-Cftr mice, providing a therapeutic strategy targeting the Krt8-F508del-CFTR interaction to correct the abnormal bone formation and bone loss in cystic fibrosis.


Asunto(s)
Enfermedades Óseas Metabólicas/etiología , Fibrosis Quística/complicaciones , Eliminación de Gen , Queratina-8/genética , Osteogénesis , Animales , Enfermedades Óseas Metabólicas/metabolismo , Fibrosis Quística/metabolismo , Fibrosis Quística/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , FN-kappa B , Osteoblastos/metabolismo , Transducción de Señal , Adulto Joven , beta Catenina
4.
Methods ; 115: 80-90, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27713081

RESUMEN

We present TrackMate, an open source Fiji plugin for the automated, semi-automated, and manual tracking of single-particles. It offers a versatile and modular solution that works out of the box for end users, through a simple and intuitive user interface. It is also easily scriptable and adaptable, operating equally well on 1D over time, 2D over time, 3D over time, or other single and multi-channel image variants. TrackMate provides several visualization and analysis tools that aid in assessing the relevance of results. The utility of TrackMate is further enhanced through its ability to be readily customized to meet specific tracking problems. TrackMate is an extensible platform where developers can easily write their own detection, particle linking, visualization or analysis algorithms within the TrackMate environment. This evolving framework provides researchers with the opportunity to quickly develop and optimize new algorithms based on existing TrackMate modules without the need of having to write de novo user interfaces, including visualization, analysis and exporting tools. The current capabilities of TrackMate are presented in the context of three different biological problems. First, we perform Caenorhabditis-elegans lineage analysis to assess how light-induced damage during imaging impairs its early development. Our TrackMate-based lineage analysis indicates the lack of a cell-specific light-sensitive mechanism. Second, we investigate the recruitment of NEMO (NF-κB essential modulator) clusters in fibroblasts after stimulation by the cytokine IL-1 and show that photodamage can generate artifacts in the shape of TrackMate characterized movements that confuse motility analysis. Finally, we validate the use of TrackMate for quantitative lifetime analysis of clathrin-mediated endocytosis in plant cells.


Asunto(s)
Rastreo Celular/métodos , Embrión no Mamífero/ultraestructura , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Análisis de la Célula Individual/métodos , Programas Informáticos , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Algoritmos , Animales , Arabidopsis/metabolismo , Arabidopsis/ultraestructura , Caenorhabditis elegans , Rastreo Celular/estadística & datos numéricos , Clatrina/genética , Clatrina/metabolismo , Embrión no Mamífero/metabolismo , Endocitosis , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Regulación de la Expresión Génica de las Plantas , Fototransducción , Células Vegetales/metabolismo , Células Vegetales/ultraestructura , Análisis de la Célula Individual/estadística & datos numéricos
5.
J Allergy Clin Immunol ; 140(6): 1671-1682.e2, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28249776

RESUMEN

BACKGROUND: Incontinentia pigmenti (IP; MIM308300) is a severe, male-lethal, X-linked, dominant genodermatosis resulting from loss-of-function mutations in the IKBKG gene encoding nuclear factor κB (NF-κB) essential modulator (NEMO; the regulatory subunit of the IκB kinase [IKK] complex). In 80% of cases of IP, the deletion of exons 4 to 10 leads to the absence of NEMO and total inhibition of NF-κB signaling. Here we describe a new IKBKG mutation responsible for IP resulting in an inactive truncated form of NEMO. OBJECTIVES: We sought to identify the mechanism or mechanisms by which the truncated NEMO protein inhibits the NF-κB signaling pathway. METHODS: We sequenced the IKBKG gene in patients with IP and performed complementation and transactivation assays in NEMO-deficient cells. We also used immunoprecipitation assays, immunoblotting, and an in situ proximity ligation assay to characterize the truncated NEMO protein interactions with IKK-α, IKK-ß, TNF receptor-associated factor 6, TNF receptor-associated factor 2, receptor-interacting protein 1, Hemo-oxidized iron regulatory protein 2 ligase 1 (HOIL-1), HOIL-1-interacting protein, and SHANK-associated RH domain-interacting protein. Lastly, we assessed NEMO linear ubiquitination using immunoblotting and investigated the formation of NEMO-containing structures (using immunostaining and confocal microscopy) after cell stimulation with IL-1ß. RESULTS: We identified a novel splice mutation in IKBKG (c.518+2T>G, resulting in an in-frame deletion: p.DelQ134_R256). The mutant NEMO lacked part of the CC1 coiled-coil and HLX2 helical domain. The p.DelQ134_R256 mutation caused inhibition of NF-κB signaling, although the truncated NEMO protein interacted with proteins involved in activation of NF-κB signaling. The IL-1ß-induced formation of NEMO-containing structures was impaired in fibroblasts from patients with IP carrying the truncated NEMO form (as also observed in HOIL-1-/- cells). The truncated NEMO interaction with SHANK-associated RH domain-interacting protein was impaired in a male fetus with IP, leading to defective linear ubiquitination. CONCLUSION: We identified a hitherto unreported disease mechanism (defective linear ubiquitination) in patients with IP.


Asunto(s)
Fibroblastos/fisiología , Quinasa I-kappa B/metabolismo , Incontinencia Pigmentaria/metabolismo , Piel/patología , Ubiquitinas/metabolismo , Clonación Molecular , Femenino , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Incontinencia Pigmentaria/genética , Masculino , Mutación/genética , FN-kappa B/metabolismo , Linaje , Unión Proteica , Transducción de Señal , Activación Transcripcional , Ubiquitinación
6.
PLoS Pathog ; 11(4): e1004877, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25923723

RESUMEN

Viral invasion into a host is initially recognized by the innate immune system, mainly through activation of the intracellular cytosolic signaling pathway and coordinated activation of interferon regulatory factor 3 (IRF3) and nuclear factor kappa B (NF-κB) transcription factors that promote type I interferon gene induction. The TANK-binding Kinase 1 (TBK1) phosphorylates and activates IRF3. Here, we show that Optineurin (Optn) dampens the antiviral innate immune response by targeting the deubiquitinating enzyme CYLD to TBK1 in order to inhibit its enzymatic activity. Importantly, we found that this regulatory mechanism is abolished at the G2/M phase as a consequence of the nuclear translocation of CYLD and Optn. As a result, we observed, at this cell division stage, an increased activity and phosphorylation of TBK1 that lead to its relocalization to mitochondria and to enhanced interferon production, suggesting that this process, which relies on Optn function, might be of major importance to mount a preventive antiviral response during mitosis.


Asunto(s)
Inmunidad Innata , Interferón beta/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción TFIIIA/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba , Transporte Activo de Núcleo Celular , Sustitución de Aminoácidos , Proteínas de Ciclo Celular , Línea Celular , Enzima Desubiquitinante CYLD , Fase G2 , Genes Reporteros , Humanos , Interferón beta/genética , Proteínas de Transporte de Membrana , Mutación , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factor de Transcripción TFIIIA/antagonistas & inhibidores , Factor de Transcripción TFIIIA/genética , Proteínas Supresoras de Tumor/agonistas , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
7.
J Biol Chem ; 290(29): 18009-18017, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26060255

RESUMEN

The prevalent human ΔF508 mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) is associated with reduced bone formation and bone loss in mice. The molecular mechanisms by which the ΔF508-CFTR mutation causes alterations in bone formation are poorly known. In this study, we analyzed the osteoblast phenotype in ΔF508-CFTR mice and characterized the signaling mechanisms underlying this phenotype. Ex vivo studies showed that the ΔF508-CFTR mutation negatively impacted the differentiation of bone marrow stromal cells into osteoblasts and the activity of osteoblasts, demonstrating that the ΔF508-CFTR mutation alters both osteoblast differentiation and function. Treatment with a CFTR corrector rescued the abnormal collagen gene expression in ΔF508-CFTR osteoblasts. Mechanistic analysis revealed that NF-κB signaling and transcriptional activity were increased in mutant osteoblasts. Functional studies showed that the activation of NF-κB transcriptional activity in mutant osteoblasts resulted in increased ß-catenin phosphorylation, reduced osteoblast ß-catenin expression, and altered expression of Wnt/ß-catenin target genes. Pharmacological inhibition of NF-κB activity or activation of canonical Wnt signaling rescued Wnt target gene expression and corrected osteoblast differentiation and function in bone marrow stromal cells and osteoblasts from ΔF508-CFTR mice. Overall, the results show that the ΔF508-CFTR mutation impairs osteoblast differentiation and function as a result of overactive NF-κB and reduced Wnt/ß-catenin signaling. Moreover, the data indicate that pharmacological inhibition of NF-κB or activation of Wnt/ß-catenin signaling can rescue the abnormal osteoblast differentiation and function induced by the prevalent ΔF508-CFTR mutation, suggesting novel therapeutic strategies to correct the osteoblast dysfunctions in cystic fibrosis.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , FN-kappa B/inmunología , Osteoblastos/citología , Vía de Señalización Wnt , Animales , Diferenciación Celular , Células Cultivadas , Masculino , Ratones , Osteoblastos/inmunología , Osteoblastos/patología , beta Catenina/inmunología
10.
Cancers (Basel) ; 14(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35158811

RESUMEN

Human Herpesvirus 8 (HHV-8) is associated with three main severe orphan malignancies, Kaposi's sarcoma (KS), multicentric Castleman's disease (MCD), and primary effusion lymphoma (PEL), which present few therapeutic options. We identified the antimalarial primaquine diphosphate (PQ) as a promising therapeutic candidate for HHV-8-associated PEL and KS. Indeed, PQ strongly reduced cell viability through caspase-dependent apoptosis, specifically in HHV-8-infected PEL cells. Reactive oxygen species (ROS)- and endoplasmic reticulum (ER) stress-mediated apoptosis signaling pathways were found to be part of the in vitro cytotoxic effect of PQ. Moreover, PQ treatment had a clinically positive effect in a nonobese diabetic (NOD)/SCID xenograft PEL mouse model, showing a reduction in tumor growth and an improvement in survival. Finally, an exploratory proof-of-concept clinical trial in four patients harboring severe KS was conducted, with the main objectives to assess the efficacy, the safety, and the tolerability of PQ, and which demonstrated a positive efficacy on Kaposi's sarcoma-related lesions and lymphedema.

11.
iScience ; 25(10): 105066, 2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36093378

RESUMEN

Patients with severe COVID-19 show an altered immune response that fails to control the viral spread and suffer from exacerbated inflammatory response, which eventually can lead to death. A major challenge is to develop an effective treatment for COVID-19. NF-κB is a major player in innate immunity and inflammatory process. By a high-throughput screening approach, we identified FDA-approved compounds that inhibit the NF-κB pathway and thus dampen inflammation. Among these, we show that Auranofin prevents post-translational modifications of NF-κB effectors and their recruitment into activating complexes in response to SARS-CoV-2 infection or cytokine stimulation. In addition, we demonstrate that Auranofin counteracts several steps of SARS-CoV-2 infection. First, it inhibits a raft-dependent endocytic pathway involved in SARS-CoV-2 entry into host cells; Second, Auranofin alters the ACE2 mobility at the plasma membrane. Overall, Auranofin should prevent SARS-CoV-2 infection and inflammatory damages, offering new opportunities as a repurposable drug candidate to treat COVID-19.

12.
Science ; 376(6599): eabm6380, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35587511

RESUMEN

The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.


Asunto(s)
Toxinas Bacterianas , Síndrome del Maullido del Gato , Endopeptidasas , Haploinsuficiencia , Proteínas Hemolisinas , Infecciones Estafilocócicas , Staphylococcus aureus , Toxinas Bacterianas/inmunología , Síndrome del Maullido del Gato/genética , Síndrome del Maullido del Gato/inmunología , Endopeptidasas/genética , Haploinsuficiencia/genética , Haploinsuficiencia/inmunología , Proteínas Hemolisinas/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Celular/genética , Necrosis , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/patología
13.
Front Immunol ; 12: 716469, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34434197

RESUMEN

The transcription factor NF-κB regulates multiple aspects of innate and adaptive immune functions and serves as a pivotal mediator of inflammatory response. In the first part of this review, we discuss the NF-κB inducers, signaling pathways, and regulators involved in immune homeostasis as well as detail the importance of post-translational regulation by ubiquitination in NF-κB function. We also indicate the stages of central and peripheral tolerance where NF-κB plays a fundamental role. With respect to central tolerance, we detail how NF-κB regulates medullary thymic epithelial cell (mTEC) development, homeostasis, and function. Moreover, we elaborate on its role in the migration of double-positive (DP) thymocytes from the thymic cortex to the medulla. With respect to peripheral tolerance, we outline how NF-κB contributes to the inactivation and destruction of autoreactive T and B lymphocytes as well as the differentiation of CD4+-T cell subsets that are implicated in immune tolerance. In the latter half of the review, we describe the contribution of NF-κB to the pathogenesis of autoimmunity and autoinflammation. The recent discovery of mutations involving components of the pathway has both deepened our understanding of autoimmune disease and informed new therapeutic approaches to treat these illnesses.


Asunto(s)
Autoinmunidad , Inflamación/metabolismo , FN-kappa B/metabolismo , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad/genética , Proteínas Portadoras/metabolismo , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Humanos , Proteínas de Punto de Control Inmunitario/genética , Tolerancia Inmunológica , Inflamación/etiología , Inflamación/patología , Inflamación/terapia , Terapia Molecular Dirigida , FN-kappa B/genética , Unión Proteica , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
14.
J Cell Biol ; 161(6): 1053-66, 2003 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-12821644

RESUMEN

Activating mutations in FGF receptor 3 (FGFR3) cause several human dwarfism syndromes by affecting both chondrocyte proliferation and differentiation. Using microarray and biochemical analyses of FGF-treated rat chondrosarcoma chondrocytes, we show that FGF inhibits chondrocyte proliferation by initiating multiple pathways that result in the induction of antiproliferative functions and the down-regulation of growth-promoting molecules. The initiation of growth arrest is characterized by the rapid dephosphorylation of the retinoblastoma protein (pRb) p107 and repression of a subset of E2F target genes by a mechanism that is independent of cyclin E-Cdk inhibition. In contrast, hypophosphorylation of pRb and p130 occur after growth arrest is first detected, and may contribute to its maintenance. Importantly, we also find a number of gene expression changes indicating that FGF promotes many aspects of hypertrophic differentiation, a notion supported by in situ analysis of developing growth plates from mice expressing an activated form of FGFR3. Thus, FGF may coordinate the onset of differentiation with chondrocyte growth arrest in the developing growth plate.


Asunto(s)
Huesos/embriología , Cartílago/embriología , Proteínas de Ciclo Celular , Condrocitos/metabolismo , Proteínas de Unión al ADN , Factores de Crecimiento de Fibroblastos/metabolismo , Osteogénesis/genética , Proteínas Tirosina Quinasas , Animales , Huesos/citología , Huesos/metabolismo , Cartílago/crecimiento & desarrollo , Cartílago/metabolismo , Diferenciación Celular/genética , División Celular/genética , Condrocitos/citología , Ciclina E/genética , Ciclina E/metabolismo , Regulación hacia Abajo/genética , Factores de Transcripción E2F , Factores de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica/genética , Genes Reguladores/genética , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratas , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteína p107 Similar a la del Retinoblastoma , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
15.
J Cell Biol ; 158(4): 741-50, 2002 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12177046

RESUMEN

Unregulated FGF signaling affects endochondral ossification and long bone growth, causing several genetic forms of human dwarfism. One major mechanism by which FGFs regulate endochondral bone growth is through their inhibitory effect on chondrocyte proliferation. Because mice with targeted mutations of the retinoblastoma (Rb)-related proteins p107 and p130 present severe endochondral bone defects with excessive chondrocyte proliferation, we have investigated the role of the Rb family of cell cycle regulators in the FGF response. Using a chondrocyte cell line, we found that FGF induced a rapid dephosphorylation of all three proteins of the Rb family (pRb, p107, and p130) and a blockade of the cells in the G1 phase of the cell cycle. This cell cycle block was reversed by inactivation of Rb proteins with viral oncoproteins such as polyoma large T (PyLT) antigen and Adenovirus E1A. Expression of a PyLT mutant that efficiently binds pRb, but not p107 and p130, allowed the cells to be growth inhibited by FGF, suggesting that pRb itself is not involved in the FGF response. To investigate more precisely the role of the individual Rb family proteins in FGF-mediated growth inhibition, we used chondrocyte micromass culture of limb bud cells isolated from mice lacking Rb proteins individually or in combination. Although wild-type as well as Rb-/- chondrocytes were similarly growth inhibited by FGF, chondrocytes null for p107 and p130 did not respond to FGF. Furthermore, FGF treatment of metatarsal bone rudiments obtained from p107-/-;p130-/- embryos failed to inhibit proliferation of growth plate chondrocytes, whereas rudiments from p107-null or p130-null embryos showed only a slight inhibition of growth. Our findings indicate that p107 and p130, but not pRb, are critical effectors of FGF-mediated growth inhibition in chondrocytes.


Asunto(s)
Condrocitos/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Proteínas , Transducción de Señal/fisiología , Animales , Huesos/citología , Huesos/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Ratones , Mutación , Proteínas Nucleares/genética , Técnicas de Cultivo de Órganos , Fosfoproteínas/genética , Fosforilación , Proteína de Retinoblastoma/química , Proteína p107 Similar a la del Retinoblastoma , Proteína p130 Similar a la del Retinoblastoma
16.
iScience ; 20: 292-309, 2019 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-31605944

RESUMEN

CEP55 regulates the final critical step of cell division termed cytokinetic abscission. We report herein that CEP55 contains two NEMO-like ubiquitin-binding domains (UBDs), NOA and ZF, which regulate its function in a different manner. In vitro studies of isolated domains showed that NOA adopts a dimeric coiled-coil structure, whereas ZF is based on a UBZ scaffold. Strikingly, CEP55 knocked-down HeLa cells reconstituted with the full-length CEP55 ubiquitin-binding defective mutants, containing structure-guided mutations either in NOACEP55 or ZFCEP55 domains, display severe abscission defects. In addition, the ZFCEP55 can be functionally replaced by some ZF-based UBDs belonging to the UBZ family, indicating that the essential function of ZFCEP55 is to act as ubiquitin receptor. Our work reveals an unexpected role of CEP55 in non-degradative ubiquitin signaling during cytokinetic abscission and provides a molecular basis as to how CEP55 mutations can lead to neurological disorders such as the MARCH syndrome.

17.
Front Immunol ; 9: 1243, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29971063

RESUMEN

Optineurin (Optn) is a 577 aa protein encoded by the Optn gene. Mutations of Optn are associated with normal tension glaucoma and amyotrophic lateral sclerosis, and its gene has also been linked to the development of Paget's disease of bone and Crohn's disease. Optn is involved in diverse cellular functions, including NF-κB regulation, membrane trafficking, exocytosis, vesicle transport, reorganization of actin and microtubules, cell cycle control, and autophagy. Besides its role in xenophagy and autophagy of aggregates, Optn has been identified as a primary autophagy receptor, among the five adaptors that translocate to mitochondria during mitophagy. Mitophagy is a selective macroautophagy process during which irreparable mitochondria are degraded, preventing accumulation of defective mitochondria and limiting the release of reactive oxygen species and proapoptotic factors. Mitochondrial quality control via mitophagy is central to the health of cells. One of the important surveillance pathways of mitochondrial health is the recently defined signal transduction pathway involving the mitochondrial PTEN-induced putative kinase 1 (PINK1) protein and the cytosolic RING-between-RING ubiquitin ligase Parkin. Both of these proteins, when mutated, have been identified in certain forms of Parkinson's disease. By targeting ubiquitinated mitochondria to autophagosomes through its association with autophagy related proteins, Optn is responsible for a critical step in mitophagy. This review reports recent discoveries on the role of Optn in mitophagy and provides insight into its link with neurodegenerative diseases. We will also discuss the involvement of Optn in other pathologies in which mitophagy dysfunctions are involved including cancer.


Asunto(s)
Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Factor de Transcripción TFIIIA/genética , Factor de Transcripción TFIIIA/metabolismo , Animales , Biomarcadores , Proteínas de Ciclo Celular , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Humanos , Proteínas de Transporte de Membrana , Mitofagia/genética , Neoplasias/patología , Enfermedades Neurodegenerativas/patología , Transducción de Señal
18.
Cytokine Growth Factor Rev ; 29: 23-33, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26976762

RESUMEN

The innate immune system has evolved to detect and neutralize viral invasions. Triggering of this defense mechanism relies on the production and secretion of soluble factors that stimulate intracellular antiviral defense mechanisms. The Tank Binding Kinase 1 (TBK1) is a serine/threonine kinase in the innate immune signaling pathways including the antiviral response and the host defense against cytosolic infection by bacteries. Given the critical roles of TBK1, important regulatory mechanisms are required to regulate its activity. Among these, Optineurin (Optn) was shown to negatively regulate the interferon response, in addition to its important role in membrane trafficking, protein secretion, autophagy and cell division. As Optn does not carry any enzymatic activity, its functions depend on its precise subcellular localization and its interaction with other proteins, especially with components of the innate immune pathway. This review highlights advances in our understanding of Optn mechanisms of action with focus on the relationships between Optn and TBK1 and their implication in host defense against pathogens. Specifically, how the antiviral immune system is controlled during the cell cycle by the Optn/TBK1 axis and the physiological consequences of this regulatory mechanism are described. This review may serve to a better understanding of the relationships between the different functions of Optn, including those related to immune responses and its associated pathologies such as primary open-angle glaucoma, amyotrophic lateral sclerosis and Paget's disease of bone.


Asunto(s)
Ciclo Celular/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Interferones/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Factor de Transcripción TFIIIA/inmunología , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/patología , Proteínas de Ciclo Celular , Glaucoma de Ángulo Abierto/inmunología , Glaucoma de Ángulo Abierto/patología , Humanos , Proteínas de Transporte de Membrana , Osteítis Deformante/inmunología , Osteítis Deformante/patología , Virosis/inmunología , Virosis/patología
19.
Nat Commun ; 7: 12629, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27586688

RESUMEN

The NF-κB pathway has critical roles in cancer, immunity and inflammatory responses. Understanding the mechanism(s) by which mutations in genes involved in the pathway cause disease has provided valuable insight into its regulation, yet many aspects remain unexplained. Several lines of evidence have led to the hypothesis that the regulatory/sensor protein NEMO acts as a biological binary switch. This hypothesis depends on the formation of a higher-order structure, which has yet to be identified using traditional molecular techniques. Here we use super-resolution microscopy to reveal the existence of higher-order NEMO lattice structures dependent on the presence of polyubiquitin chains before NF-κB activation. Such structures may permit proximity-based trans-autophosphorylation, leading to cooperative activation of the signalling cascade. We further show that NF-κB activation results in modification of these structures. Finally, we demonstrate that these structures are abrogated in cells derived from incontinentia pigmenti patients.


Asunto(s)
Quinasa I-kappa B/ultraestructura , Incontinencia Pigmentaria/patología , Microscopía/métodos , FN-kappa B/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/fisiología , Unión Proteica , Estructura Secundaria de Proteína , Ubiquitina/metabolismo
20.
J Exp Med ; 212(6): 939-51, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26008899

RESUMEN

Inherited, complete deficiency of human HOIL-1, a component of the linear ubiquitination chain assembly complex (LUBAC), underlies autoinflammation, infections, and amylopectinosis. We report the clinical description and molecular analysis of a novel inherited disorder of the human LUBAC complex. A patient with multiorgan autoinflammation, combined immunodeficiency, subclinical amylopectinosis, and systemic lymphangiectasia, is homozygous for a mutation in HOIP, the gene encoding the catalytic component of LUBAC. The missense allele (L72P, in the PUB domain) is at least severely hypomorphic, as it impairs HOIP expression and destabilizes the whole LUBAC complex. Linear ubiquitination and NF-κB activation are impaired in the patient's fibroblasts stimulated by IL-1ß or TNF. In contrast, the patient's monocytes respond to IL-1ß more vigorously than control monocytes. However, the activation and differentiation of the patient's B cells are impaired in response to CD40 engagement. These cellular and clinical phenotypes largely overlap those of HOIL-1-deficient patients. Clinical differences between HOIL-1- and HOIP-mutated patients may result from differences between the mutations, the loci, or other factors. Our findings show that human HOIP is essential for the assembly and function of LUBAC and for various processes governing inflammation and immunity in both hematopoietic and nonhematopoietic cells.


Asunto(s)
Regulación de la Expresión Génica , Ubiquitina-Proteína Ligasas/deficiencia , Alelos , Secuencia de Aminoácidos , Ligando de CD40/metabolismo , Catálisis , Femenino , Fibroblastos/metabolismo , Prueba de Complementación Genética , Mutación de Línea Germinal , Enfermedad del Almacenamiento de Glucógeno Tipo IV/patología , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/patología , Inflamación , Linfangiectasia/patología , Monocitos/metabolismo , Mutación Missense , FN-kappa B/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción , Ubiquitina/química , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda