Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Immunol ; 14(5): 470-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23542741

RESUMEN

Uncontrolled activation of tumor necrosis factor receptor-associated factor (TRAF) proteins may result in profound tissue injury by linking surface signals to cytokine release. Here we show that a ubiquitin E3 ligase component, Fbxo3, potently stimulates cytokine secretion from human inflammatory cells by destabilizing a sentinel TRAF inhibitor, Fbxl2. Fbxo3 and TRAF protein in circulation positively correlated with cytokine responses in subjects with sepsis, and we identified a polymorphism in human Fbxo3, with one variant being hypofunctional. A small-molecule inhibitor targeting Fbxo3 was sufficient to lessen severity of cytokine-driven inflammation in several mouse disease models. These studies identified a pathway of innate immunity that may be useful to detect subjects with altered immune responses during critical illness or provide a basis for therapeutic intervention targeting TRAF protein abundance.


Asunto(s)
Proteínas F-Box/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Sepsis/inmunología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Ciego/inmunología , Ciego/cirugía , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Secuencias F-Box/genética , Proteínas F-Box/genética , Humanos , Inmunomodulación , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Polimorfismo Genético , Estabilidad Proteica , Infecciones por Pseudomonas/genética , Pseudomonas aeruginosa/genética , ARN Interferente Pequeño/genética , Sepsis/genética , Transgenes/genética
2.
J Immunol ; 209(9): 1788-1795, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36113884

RESUMEN

Chronic lung allograft dysfunction is the major barrier to long-term survival in lung transplant recipients. Evidence supports type 1 alloimmunity as the predominant response in acute/chronic lung rejection, but the immunoregulatory mechanisms remain incompletely understood. We studied the combinatorial F-box E3 ligase system: F-box protein 3 (FBXO3; proinflammatory) and F-box and leucine-rich repeat protein 2 (FBXL2; anti-inflammatory and regulates TNFR-associated factor [TRAF] protein). Using the mouse orthotopic lung transplant model, we evaluated allografts from BALB/c → C57BL/6 (acute rejection; day 10) and found significant induction of FBXO3 and diminished FBXL2 protein along with elevated T-bet, IFN-γ, and TRAF proteins 1-5 compared with isografts. In the acute model, treatment with costimulation blockade (MR1/CTLA4-Ig) resulted in attenuated FBXO3, preserved FBXL2, and substantially reduced T-bet, IFN-γ, and TRAFs 1-5, consistent with a key role for type 1 alloimmunity. Immunohistochemistry revealed significant changes in the FBXO3/FBXL2 balance in airway epithelia and infiltrating mononuclear cells during rejection compared with isografts or costimulation blockade-treated allografts. In the chronic lung rejection model, DBA/2J/C57BL/6F1 > DBA/2J (day 28), we observed persistently elevated FBXO3/FBXL2 balance and T-bet/IFN-γ protein and similar findings from lung transplant recipient lungs with chronic lung allograft dysfunction versus controls. We hypothesized that FBXL2 regulated T-bet and found FBXL2 was sufficient to polyubiquitinate T-bet and coimmunoprecipitated with T-bet on pulldown experiments and vice versa in Jurkat cells. Transfection with FBXL2 diminished T-bet protein in a dose-dependent manner in mouse lung epithelial cells. In testing type 1 cytokines, TNF-α was found to negatively regulate FBXL2 protein and mRNA levels. Together, our findings show the combinatorial E3 ligase FBXO3/FBXL2 system plays a role in the regulation of T-bet through FBXL2, with negative cross-regulation of TNF-α on FBXL2 during lung allograft rejection.


Asunto(s)
Proteínas F-Box , Animales , Ratones , Abatacept , Aloinjertos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Rechazo de Injerto , Pulmón/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , ARN Mensajero , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
3.
Respir Res ; 18(1): 131, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659178

RESUMEN

BACKGROUND: Azithromycin, an antibiotic used for multiple infectious disorders, exhibits anti-inflammatory effects, but the molecular basis for this activity is not well characterized. Azithromycin inhibits IL-1ß-mediated inflammation that is dependent, in part, on inflammasome activity. Here, we investigated the effects of azithromycin on the NACHT, LRR, and PYD domains-containing protein 3 (NALP3) protein, which is the sensing component of the NALP3 inflammasome, in human monocytes. METHODS: THP-1 cells were treated with azithromycin alone, LPS alone, or both. NALP3 and IL-1ß protein levels were determined by immunoblotting. NLRP3 gene (encoding NALP3) transcript levels were determined by quantitative qPCR. In order to measure NLRP3 transcript decay, actinomycin D was used to impair gene transcription. THP-1 Lucia cells which contain an NF-κB responsive luciferase element were used to assess NF-κB activity in response to azithromycin, LPS, and azithromycin/LPS by measuring luminescence. To confirm azithromycin's effects on NLRP3 mRNA and promoter activity conclusively, HEK cells were lipofected with luciferase reporter constructs harboring either the 5' untranslated region (UTR) of the NLRP3 gene which included the promoter, the 3' UTR of the gene, or an empty plasmid prior to treatment with azithromycin and/or LPS, and luminescence was measured. RESULTS: Azithromycin decreased IL-1ß levels and reduced NALP3 protein levels in LPS-stimulated THP-1 monocytes through a mechanism involving decreased mRNA stability of the NALP3 - coding NLRP3 gene transcript as well as by decreasing NF-κB activity. Azithromycin accelerated NLRP3 transcript decay confirmed by mRNA stability and 3'UTR luciferase reporter assays, and yet the antibiotic had no effect on NLRP3 promoter activity in cells containing a 5' UTR reporter. CONCLUSIONS: These studies provide a unique mechanism whereby azithromycin exerts immunomodulatory actions in monocytes by destabilizing mRNA levels for a key inflammasome component, NALP3, leading to decreased IL-1ß-mediated inflammation.


Asunto(s)
Azitromicina/farmacología , Inflamasomas/metabolismo , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN Mensajero/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Azitromicina/uso terapéutico , Línea Celular , Células HEK293 , Humanos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Monocitos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Estabilidad Proteica/efectos de los fármacos , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética
4.
Am J Respir Cell Mol Biol ; 52(5): 622-33, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25286244

RESUMEN

Acute cellular rejection is a known risk factor for the development of obliterative bronchiolitis, which limits the long-term survival of lung transplant recipients. However, the T cell effector mechanisms in both of these processes remain incompletely understood. Using the mouse orthotopic lung transplant model, we investigated whether C57BL/6 T-bet(-/-) recipients of major histocompatibility complex (MHC)-mismatched BALB/c lung grafts develop rejection pathology and allospecific cytokine responses that differ from wild-type mice. T-bet(-/-) recipients demonstrated vigorous allograft rejection at 10 days, characterized by neutrophilic inflammation and predominantly CD8(+) T cells producing allospecific IL-17 and/or IFN-γ, in contrast to IFN-γ-dominant responses in WT mice. CD4(+) T cells produced IL-17 but not IFN-γ responses in T-bet(-/-) recipients, in contrast to WT controls. Costimulation blockade using anti-CD154 Ab significantly reduced allospecific CD8(+)IFN-γ(+) responses in both T-bet(-/-) and WT mice but had no attenuating effect on lung rejection pathology in T-bet(-/-) recipients or on the development of obliterative airway inflammation that occurred only in T-bet(-/-) recipients. However, neutralization of IL-17A significantly attenuated costimulation blockade-resistant rejection pathology and airway inflammation in T-bet(-/-) recipients. In addition, CXCL1 (neutrophil chemokine) was increased in T-bet(-/-) allografts, and IL-17 induced CXCL1 from mouse lung epithelial cells in vitro. Taken together, our data show that T-bet-deficient recipients of complete MHC-mismatched lung allografts develop costimulation blockade-resistant rejection characterized by neutrophilia and obliterative airway inflammation that is predominantly mediated by CD8(+)IL-17(+) T cells. Our data support T-bet-deficient mouse recipients of lung allografts as a viable animal model to study the immunopathogenesis of small airway injury in lung transplantation.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Rechazo de Injerto/etiología , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Trasplante de Pulmón/efectos adversos , Pulmón/metabolismo , Neutrófilos/metabolismo , Neumonía/etiología , Proteínas de Dominio T Box/metabolismo , Enfermedad Aguda , Aloinjertos , Animales , Anticuerpos/farmacología , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Rechazo de Injerto/prevención & control , Histocompatibilidad , Mediadores de Inflamación/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutrófilos/inmunología , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/patología , Neumonía/prevención & control , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
5.
J Immunol ; 191(10): 5247-55, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24123678

RESUMEN

Cytokine-driven inflammation underlies the pathobiology of a wide array of infectious and immune-related disorders. The TNFR-associated factor (TRAF) proteins have a vital role in innate immunity by conveying signals from cell surface receptors to elicit transcriptional activation of genes encoding proinflammatory cytokines. We discovered that a ubiquitin E3 ligase F box component, termed Fbxo3, potently stimulates cytokine secretion from human inflammatory cells by mediating the degradation of the TRAF inhibitory protein, Fbxl2. Analysis of the Fbxo3 C-terminal structure revealed that the bacterial-like ApaG molecular signature was indispensible for mediating Fbxl2 disposal and stimulating cytokine secretion. By targeting this ApaG motif, we developed a highly unique, selective genus of small-molecule Fbxo3 inhibitors that by reducing TRAF protein levels, potently inhibited cytokine release from human blood mononuclear cells. The Fbxo3 inhibitors effectively lessened the severity of viral pneumonia, septic shock, colitis, and cytokine-driven inflammation systemically in murine models. Thus, pharmacological targeting of Fbxo3 might be a promising strategy for immune-related disorders characterized by a heightened host inflammatory response.


Asunto(s)
Bencilaminas/uso terapéutico , Proteínas F-Box/metabolismo , Inflamación/tratamiento farmacológico , Piridinas/uso terapéutico , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Bencilaminas/farmacología , Línea Celular , Colitis/tratamiento farmacológico , Citocinas/biosíntesis , Citocinas/metabolismo , Proteínas F-Box/antagonistas & inhibidores , Inflamación/inmunología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Piridinas/farmacología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Biol Chem ; 288(9): 6306-16, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23319590

RESUMEN

Histone acetyltransferase binding to origin recognition complex (HBO1) plays a crucial role in DNA replication licensing and cell proliferation, yet its molecular regulation in cells is relatively unknown. Here an uncharacterized protein, Fbxw15, directly interacts with HBO1, a labile protein (t½ = ∼3 h), to mediate its ubiquitination (Lys(338)) and degradation in the cytoplasm. Fbxw15-mediated HBO1 depletion required mitogen-activated protein kinase 1 (Mek1), which was sufficient to trigger HBO1 phosphorylation and degradation in cells. Mek1 ability to produce HBO1 degradation was blocked by Fbxw15 silencing. Lipopolysaccharide induced HBO1 degradation, an effect abrogated by Fbxw15 or Mek1 cellular depletion. Modulation of Fbxw15 levels was able to differentially regulate histone H3K14 acetylation and cellular proliferation by altering HBO1 levels. These studies authenticate Fbxw15 as a ubiquitin E3 ligase subunit that mediates endotoxin-induced HBO1 depletion in cells, thereby controlling cell replicative capacity.


Asunto(s)
Proliferación Celular , Proteínas F-Box/metabolismo , Histona Acetiltransferasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitina/metabolismo , Acetilación , Animales , Línea Celular , Proteínas F-Box/genética , Silenciador del Gen , Histona Acetiltransferasas/genética , Histonas/genética , Histonas/metabolismo , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Ratones , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L534-42, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24441869

RESUMEN

MicroRNAs are small noncoding RNAs that inhibit protein expression. We have previously shown that the inhibition of the microRNA let-7d in epithelial cells caused changes consistent with epithelial-to-mesenchymal transition (EMT) both in vitro and in vivo. The aim of this study was to determine whether the introduction of let-7d into fibroblasts alters their mesenchymal properties. Transfection of primary fibroblasts with let-7d caused a decrease in expression of the mesenchymal markers α-smooth muscle actin, N-cadherin, fibroblast-specific protein-1, and fibronectin, as well as an increase in the epithelial markers tight junction protein-1 and keratin 19. Phenotypic changes were also present, including a delay in wound healing, reduced motility, and proliferation of fibroblasts following transfection. In addition, we examined the effects of transfection on fibroblast responsiveness to TGF-ß, an important factor in many fibrotic processes such as lung fibrosis and found that let-7d transfection significantly attenuated high-mobility group-A2 protein induction by TGF-ß. Our results indicate that administration of the epithelial microRNA let-7d can significantly alter the phenotype of primary fibroblasts.


Asunto(s)
Transición Epitelial-Mesenquimal , Fibroblastos/citología , Pulmón/metabolismo , MicroARNs/genética , Miofibroblastos/metabolismo , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Actinas/metabolismo , Cadherinas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Proteína HMGA2/metabolismo , Proteína HMGB2/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Queratina-19/metabolismo , Pulmón/citología , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/genética , Proteína de Unión al Calcio S100A4 , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Cicatrización de Heridas/genética , Proteína de la Zonula Occludens-1/metabolismo
8.
Blood ; 119(13): 3132-41, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22323446

RESUMEN

Hematologic maligancies exhibit a growth advantage by up-regulation of components within the molecular apparatus involved in cell-cycle progression. The SCF (Skip-Cullin1-F-box protein) E3 ligase family provides homeostatic feedback control of cell division by mediating ubiquitination and degradation of cell-cycle proteins. By screening several previously undescribed E3 ligase components, we describe the behavior of a relatively new SCF subunit, termed FBXL2, that ubiquitinates and destabilizes cyclin D2 protein leading to G(0) phase arrest and apoptosis in leukemic and B-lymphoblastoid cell lines. FBXL2 expression was strongly suppressed, and yet cyclin D2 protein levels were robustly expressed in acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL) patient samples. Depletion of endogenous FBXL2 stabilized cyclin D2 levels, whereas ectopically expressed FBXL2 decreased cyclin D2 lifespan. FBXL2 did not bind a phosphodegron within its substrate, which is typical of other F-box proteins, but uniquely targeted a calmodulin-binding signature within cyclin D2 to facilitate its polyubiquitination. Calmodulin competes with the F-box protein for access to this motif where it bound and protected cyclin D2 from FBXL2. Calmodulin reversed FBXL2-induced G(0) phase arrest and attenuated FBXL2-induced apoptosis of lymphoblastoid cells. These results suggest an antiproliferative effect of SCF(FBXL2) in lymphoproliferative malignancies.


Asunto(s)
Proliferación Celular , Ciclina D2/metabolismo , Proteínas F-Box/fisiología , Leucemia/patología , Animales , Apoptosis/genética , Apoptosis/fisiología , Calmodulina/metabolismo , Calmodulina/fisiología , Células Cultivadas , Regulación hacia Abajo/fisiología , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Regulación Leucémica de la Expresión Génica , Humanos , Células K562 , Leucemia/genética , Leucemia/metabolismo , Ratones , Proteolisis , Transfección , Células U937 , Ubiquitinación/genética
9.
FASEB J ; 27(7): 2611-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23512198

RESUMEN

Rac1, a member of the Rho family of GTPases, regulates diverse cellular functions, including cytoskeleton reorganization and cell migration. F-box proteins are major subunits within the Skp1-Cul1-F-box (SCF) E3 ubiquitin ligases that recognize specific substrates for ubiquitination. The role of F-box proteins in regulating Rac1 stability has not been studied. Mouse lung epithelial (MLE12) cells were used to investigate Rac1 stability and cell migration. Screening of an F-box protein library and in vitro ubiquitination assays identified FBXL19, a relatively new member of the F-box protein family that targets Rac1 for its polyubiquitination and proteasomal degradation. Overexpression of FBXL19 decreased both Rac1 active and inactive forms and significantly reduced cellular migration. Protein kinase AKT-mediated phosphorylation of Rac1 at serine(71) was essential for FBXL19-mediated Rac1 ubiquitination and depletion. Lysine(166) within Rac1 was identified as a polyubiquitination acceptor site. Rac1(S71A) and Rac1(K166R) mutant proteins were resistant to FBXL19-mediated ubiquitination and degradation. Further, ectopically expressed FBXL19 reduced cell migration in Rac1-overexpressing cells (P<0.01, Rac1 cells vs. FBXL19+Rac1 cells), but not in Rac1 lysine(166) mutant-overexpressing cells. FBXL19 diminished formation of the migratory leading edge. Thus, SCF(FBXL19) targets Rac1 for its disposal, a process regulated by AKT. These findings provide the first evidence of an F-box protein targeting a small G protein for ubiquitination and degradation to modulate cell migration.


Asunto(s)
Movimiento Celular , Proteínas de Unión al ADN/metabolismo , Proteínas F-Box/metabolismo , Neuropéptidos/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas F-Box/genética , Humanos , Immunoblotting , Ratones , Mutación , Neuropéptidos/genética , Fosforilación , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Serina/genética , Serina/metabolismo , Ubiquitinación , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1
10.
J Biol Chem ; 286(4): 2719-27, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21068446

RESUMEN

Acyl-CoA:lysophosphatidylcholine acyltransferase 1 (LPCAT1) is a relatively newly described and yet indispensable enzyme needed for generation of the bioactive surfactant phospholipid, dipalmitoylphosphatidylcholine (DPPtdCho). Here, we show that lipopolysaccharide (LPS) causes LPCAT1 degradation using the Skp1-Cullin-F-box ubiquitin E3 ligase component, ß-transducin repeat-containing protein (ß-TrCP), that polyubiquitinates LPCAT1, thereby targeting the enzyme for proteasomal degradation. LPCAT1 was identified as a phosphoenzyme as Ser(178) within a phosphodegron was identified as a putative molecular recognition site for glycogen synthase kinase-3ß (GSK-3ß) phosphorylation that recruits ß-TrCP docking within the enzyme. ß-TrCP ubiquitinates LPCAT1 at an acceptor site (Lys(221)), as substitution of Lys(221) with Arg abrogated LPCAT1 polyubiquitination. LPS profoundly reduced immunoreactive LPCAT1 levels and impaired lung surfactant mechanics, effects that were overcome by siRNA to ß-TrCP and GSK-3ß or LPCAT1 gene transfer, respectively. Thus, LPS appears to destabilize the LPCAT1 protein by GSK-3ß-mediated phosphorylation within a canonical phosphodegron for ß-TrCP docking and site-specific ubiquitination. LPCAT1 is the first lipogenic substrate for ß-TrCP, and the results suggest that modulation of the GSK-3ß-SCFß(TrCP) E3 ligase effector pathway might be a unique strategy to optimize dipalmitoylphosphatidylcholine levels in sepsis.


Asunto(s)
1,2-Dipalmitoilfosfatidilcolina/biosíntesis , 1-Acilglicerofosfocolina O-Aciltransferasa/metabolismo , Lipopolisacáridos/farmacología , Surfactantes Pulmonares/metabolismo , Mucosa Respiratoria/metabolismo , Ubiquitinación/efectos de los fármacos , Proteínas con Repetición de beta-Transducina/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/genética , 1-Acilglicerofosfocolina O-Aciltransferasa/genética , Animales , Células Cultivadas , Estabilidad de Enzimas/efectos de los fármacos , Estabilidad de Enzimas/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Ratones , Fosforilación/efectos de los fármacos , Fosforilación/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Sepsis/genética , Sepsis/metabolismo , Ubiquitinación/genética , Proteínas con Repetición de beta-Transducina/genética
11.
JCI Insight ; 4(19)2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31578312

RESUMEN

Dysregulated proinflammatory cytokine release has been implicated in the pathogenesis of several life-threatening acute lung illnesses such as pneumonia, sepsis, and acute respiratory distress syndrome. Suppressors of cytokine signaling proteins, particularly SOCS2, have recently been described as antiinflammatory mediators. However, the regulation of SOCS2 protein has not been described. Here we describe a mechanism of SOCS2 regulation by the action of the ubiquitin E3 ligase KIAA0317. KIAA0317-mediated degradation of SOCS2 exacerbated inflammation in vitro, and depletion of KIAA0317 in vivo ameliorated pulmonary inflammation. KIAA0317-knockout mice exhibited resistance to LPS-induced pulmonary inflammation, while KIAA03017 reexpression mitigated this effect. We uncovered a small molecule inhibitor of KIAA0317 protein (BC-1365) that prevented SOCS2 degradation and attenuated LPS- and P. aeruginosa-induced lung inflammation in vivo. These studies show KIAA0317 to be a critical mediator of pulmonary inflammation through its degradation of SOCS2 and a potential candidate target for therapeutic inhibition.


Asunto(s)
Pulmón/metabolismo , Neumonía/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Adulto , Anciano , Animales , Citocinas/metabolismo , Femenino , Humanos , Inmunidad Innata , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Simulación del Acoplamiento Molecular , Neumonía/inmunología , Neumonía/patología , Unión Proteica , Transcriptoma
12.
J Exp Med ; 213(6): 1029-46, 2016 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-27162139

RESUMEN

The E3 small ubiquitin-like modifier (SUMO) protein ligase protein inhibitor of activated STAT 4 (PIAS4) is a pivotal protein in regulating the TGFß pathway. In this study, we discovered a new protein isoform encoded by KIAA0317, termed fibrosis-inducing E3 ligase 1 (FIEL1), which potently stimulates the TGFß signaling pathway through the site-specific ubiquitination of PIAS4. FIEL1 targets PIAS4 using a double locking mechanism that is facilitated by the kinases PKCζ and GSK3ß. Specifically, PKCζ phosphorylation of PIAS4 and GSK3ß phosphorylation of FIEL1 are both essential for the degradation of PIAS4. FIEL1 protein is highly expressed in lung tissues from patients with idiopathic pulmonary fibrosis (IPF), whereas PIAS4 protein levels are significantly reduced. FIEL1 overexpression significantly increases fibrosis in a bleomycin murine model, whereas FIEL1 knockdown attenuates fibrotic conditions. Further, we developed a first-in-class small molecule inhibitor toward FIEL1 that is highly effective in ameliorating fibrosis in mice. This study provides a basis for IPF therapeutic intervention by modulating PIAS4 protein abundance.


Asunto(s)
Pulmón/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Fibrosis Pulmonar/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Bleomicina/efectos adversos , Bleomicina/farmacología , Línea Celular , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Pulmón/patología , Ratones , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Inhibidoras de STAT Activados/genética , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteína Quinasa C-theta , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Ubiquitina-Proteína Ligasas/genética
13.
Sci Transl Med ; 7(295): 295ra109, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26157031

RESUMEN

Invading pathogens may trigger overactivation of the innate immune system, which results in the release of large amounts of proinflammatory cytokines (cytokine storm) and leads to the development of pulmonary edema, multiorgan failure, and shock. PIAS1 is a multifunctional and potent anti-inflammatory protein that negatively regulates several key inflammatory pathways such as Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and nuclear factor κB (NF-κB). We discovered a ubiquitin E3 ligase, HECTD2, which ubiquitinated and mediated the degradation of PIAS1, thus increasing inflammation in an experimental pneumonia model. We found that GSK3ß phosphorylation of PIAS1 provided a phosphodegron for HECTD2 targeting. We also identified a mislocalized HECTD2 polymorphism, HECTD2(A19P), that was present in 8.5% of the population and functioned to reduce inflammation. This polymorphism prevented HECTD2/PIAS1 nuclear interaction, thus preventing PIAS1 degradation. The HECTD2(A19P) polymorphism was also protective toward acute respiratory distress syndrome (ARDS). We then developed a small-molecule inhibitor, BC-1382, that targeted HECTD2 and attenuated lipopolysaccharide (LPS)- and Pseudomonas aeruginosa-induced lung inflammation. These studies describe an unreported innate immune pathway and suggest that mutation or antagonism of the E3 ligase HECTD2 results in reduced severity of lung inflammation by selectively modulating the abundance of the anti-inflammatory protein PIAS1.


Asunto(s)
Inmunidad Innata/fisiología , Inflamación/fisiopatología , Lesión Pulmonar/inmunología , Lesión Pulmonar/fisiopatología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Técnicas de Silenciamiento del Gen , Lesión Pulmonar/microbiología , Ratones , Fosforilación , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Pseudomonas/patogenicidad
14.
PLoS One ; 9(12): e115937, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25549252

RESUMEN

Cyclic adenosine monophosphate (cAMP) is a crucial intracellular second messenger molecule that converts extracellular molecules to intracellular signal transduction pathways generating cell- and stimulus-specific effects. Importantly, specific phosphodiesterase (PDE) subtypes control the amplitude and duration of cAMP-induced physiological processes and are therefore a prominent pharmacological target currently used in a variety of fields. Here we tested the extracts from traditional Chinese medicine, Forsythia suspense seeds, which have been used for more than 2000 years to relieve respiratory symptoms. Using structural-functional analysis we found its major lignin, Forsynthin, acted as an immunosuppressant by inhibiting PDE4 in inflammatory and immune cell. Moreover, several novel, selective small molecule derivatives of Forsythin were tested in vitro and in murine models of viral and bacterial pneumonia, sepsis and cytokine-driven systemic inflammation. Thus, pharmacological targeting of PDE4 may be a promising strategy for immune-related disorders characterized by amplified host inflammatory response.


Asunto(s)
Forsythia/química , Lignina/farmacología , Inhibidores de Fosfodiesterasa 4/aislamiento & purificación , Animales , Simulación por Computador , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Inflamación/tratamiento farmacológico , Lignina/aislamiento & purificación , Lesión Pulmonar/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa 4/farmacología , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Plantas Medicinales/química , Choque Séptico/tratamiento farmacológico , Relación Estructura-Actividad
15.
Cell Rep ; 7(2): 476-487, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24703837

RESUMEN

Acute lung injury (ALI) is linked to mitochondrial injury, resulting in impaired cellular oxygen utilization; however, it is unknown how these events are linked on the molecular level. Cardiolipin, a mitochondrial-specific lipid, is generated by cardiolipin synthase (CLS1). Here, we show that S. aureus activates a ubiquitin E3 ligase component, Fbxo15, that is sufficient to mediate proteasomal degradation of CLS1 in epithelia, resulting in decreased cardiolipin availability and disrupted mitochondrial function. CLS1 is destabilized by the phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1), which binds CLS1 to phosphorylate and regulates CLS1 disposal. Like Fbxo15, PINK1 interacts with and regulates levels of CLS1 through a mechanism dependent upon Thr219. S. aureus infection upregulates this Fbxo15-PINK1 pathway to impair mitochondrial integrity, and Pink1 knockout mice are less prone to S. aureus-induced ALI. Thus, ALI-associated disruption of cellular bioenergetics involves bioeffectors that utilize a phosphodegron to elicit ubiquitin-mediated disposal of a key mitochondrial enzyme.


Asunto(s)
Antígeno B7-2/metabolismo , Proteínas F-Box/metabolismo , Mitocondrias/metabolismo , Neumonía/metabolismo , Proteínas Quinasas/metabolismo , Adolescente , Adulto , Animales , Antígeno B7-2/genética , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Niño , Estabilidad de Enzimas , Proteínas F-Box/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas Quinasas/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
16.
Cell Cycle ; 12(4): 663-73, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23370391

RESUMEN

Aurora B kinase is an integral regulator of cytokinesis as it stabilizes the intercellular canal within the midbody to ensure proper chromosomal segregation during cell division. Here we identified an E3 ligase subunit, F box protein FBXL2, that by recognizing a calmodulin binding signature within Aurora B, ubiquitinates and removes the kinase from the midbody. Calmodulin, by competing with the F box protein for access to the calmodulin binding signature, protected Aurora B from FBXL2. Calmodulin co-localized with Aurora B on the midbody, preserved Aurora B levels in cells, and stabilized intercellular canals during delayed abscission. Genetic or pharmaceutical depletion of endogenous calmodulin significantly reduced Aurora B protein levels at the midbody resulting in tetraploidy and multi-spindle formation. The calmodulin inhibitor, calmidazolium, reduced Aurora B protein levels resulting in tetraploidy, mitotic arrest, and apoptosis of tumorigenic cells and profoundly inhibiting tumor formation in athymic nude mice. These observations indicate molecular interplay between Aurora B and calmodulin in telophase and suggest that calmodulin acts as a checkpoint sensor for chromosomal segregation errors during mitosis.


Asunto(s)
Calmodulina/genética , Citocinesis/genética , Proteínas F-Box/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Telofase/genética , Animales , Apoptosis/efectos de los fármacos , Aurora Quinasa B , Aurora Quinasas , Sitios de Unión , Calmodulina/metabolismo , Línea Celular Tumoral , Segregación Cromosómica/efectos de los fármacos , Citocinesis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Proteínas F-Box/metabolismo , Humanos , Imidazoles/farmacología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Telofase/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
17.
Cell Cycle ; 11(4): 721-9, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22306998

RESUMEN

Aurora family kinases play pivotal roles in several steps during mitosis. Specifically, Aurora A kinase is an important regulator of bipolar mitotic spindle formation and chromosome segregation. Like other members of the Aurora family, Aurora A kinase is also regulated by post-translational modifications. Here, we show that a previously undescribed E3 ligase component belonging to the SCF (Skp-Cullin1-F-box protein) E3 ligase family, SCFFBXL7, impairs cell proliferation by mediating Aurora A polyubiquitination and degradation. Both Aurora A and FBXL7 co-localize within the centrosome during spindle formation. FBXL7 ectopic expression led to G(2)/M phase arrest in transformed epithelia, resulting in the appearance of tetraploidy and mitotic arrest with circular monopolar spindles and multipolar spindle formation. Interestingly, FBXL7 specifically interacts with Aurora A during mitosis but not in interphase, suggesting a regulatory role for FBXL7 in controlling Aurora A abundance during mitosis.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Ciclo Celular/fisiología , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Aurora Quinasa A , Aurora Quinasas , Ciclo Celular/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Transferencia Resonante de Energía de Fluorescencia , Humanos , Ratones , Mitosis/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Ligasas SKP Cullina F-box/genética , Ubiquitinación/genética , Ubiquitinación/fisiología
18.
Mol Cell Biol ; 31(9): 1905-20, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21343341

RESUMEN

Calmodulin is a universal calcium-sensing protein that has pleiotropic effects. Here we show that calmodulin inhibits a new SCF (Skp1-Cullin-F-box) E3 ligase component, FBXL2. During Pseudomonas aeruginosa infection, SCF (FBXL2) targets the key enzyme, CCTα, for its monoubiquitination and degradation, thereby reducing synthesis of the indispensable membrane and surfactant component, phosphatidylcholine. P. aeruginosa triggers calcium influx and calcium-dependent activation of FBXL2 within the Golgi complex, where it engages CCTα. FBXL2 through its C terminus binds to the CCTα IQ motif. FBXL2 knockdown increases CCTα levels and phospholipid synthesis. The molecular interaction of FBXL2 with CCTα is opposed by calmodulin, which traffics to the Golgi complex, binds FBXL2 (residues 80 to 90) via its C terminus, and vies with the ligase for occupancy within the IQ motif. These observations were recapitulated in murine models of P. aeruginosa-induced surfactant deficiency, where calmodulin gene transfer reduced FBXL2 actions by stabilizing CCTα and lessening the severity of inflammatory lung injury. The results provide a unique model of calcium-regulated intermolecular competition between an E3 ligase subunit and an antagonist that is critically relevant to pneumonia and lipid homeostasis.


Asunto(s)
Calmodulina/metabolismo , Proteínas F-Box/metabolismo , Interacciones Huésped-Patógeno , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiología , Proteínas Ligasas SKP Cullina F-box/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Chaperonina con TCP-1/metabolismo , Aparato de Golgi/metabolismo , Aparato de Golgi/microbiología , Pulmón/metabolismo , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfatidilcolinas/metabolismo , Infecciones por Pseudomonas/microbiología
19.
Cell Cycle ; 10(20): 3487-94, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22024926

RESUMEN

Mitotic progression is regulated by ubiquitin E3 ligase complexes to carefully orchestrate eukaryotic cell division. Here, we show that a relatively new E3 ligase component belonging to the SCF (Skip-Cullin1-F-box protein) E3 ligase family, SCF (FBXL2) , impairs cell proliferation by mediating cyclin D3 polyubiquitination and degradation. Both cyclin D3 and FBXL2 colocalize within the centrosome. FBXL2 overexpression led to G 2/M-phase arrest in transformed epithelia, resulting in the appearance of supernumerary centrosomes, tetraploidy and nuclei where condensed chromosomes are arranged on circular monopolar spindles typical of mitotic arrest. RNAi-mediated knockdown of cyclin D3 recapitulated effects of SCF (FBXL2) expression. SCF (FBXL2) impaired the ability of cyclin D3 to associate with centrosomal assembly proteins [Aurora A, polo-like kinase 4 (Plk4), CDK11]. Thus, these results suggest a role for SCF (FBXL2) in regulating the fidelity of cellular division.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Ciclina D3/metabolismo , Proteínas F-Box/metabolismo , Mitosis/fisiología , Modelos Biológicos , Ubiquitina-Proteína Ligasas/metabolismo , Análisis de Varianza , Animales , Línea Celular , Ciclina D3/genética , Fluorescencia , Immunoblotting , Inmunoprecipitación , Ratones , Interferencia de ARN , Factores de Tiempo , Ubiquitinación
20.
Nat Med ; 16(10): 1120-1127, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20852622

RESUMEN

Pneumonia remains the leading cause of death from infection in the US, yet fundamentally new conceptual models underlying its pathogenesis have not emerged. We show that humans and mice with bacterial pneumonia have markedly elevated amounts of cardiolipin, a rare, mitochondrial-specific phospholipid, in lung fluid and find that it potently disrupts surfactant function. Intratracheal cardiolipin administration in mice recapitulates the clinical phenotype of pneumonia, including impaired lung mechanics, modulation of cell survival and cytokine networks and lung consolidation. We have identified and characterized the activity of a unique cardiolipin transporter, the P-type ATPase transmembrane lipid pump Atp8b1, a mutant version of which is associated with severe pneumonia in humans and mice. Atp8b1 bound and internalized cardiolipin from extracellular fluid via a basic residue-enriched motif. Administration of a peptide encompassing the cardiolipin binding motif or Atp8b1 gene transfer in mice lessened bacteria-induced lung injury and improved survival. The results unveil a new paradigm whereby Atp8b1 is a cardiolipin importer whose capacity to remove cardiolipin from lung fluid is exceeded during inflammation or when Atp8b1 is defective. This discovery opens the door for new therapeutic strategies directed at modulating the abundance or molecular interactions of cardiolipin in pneumonia.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Cardiolipinas/fisiología , Lesión Pulmonar/etiología , Neumonía Bacteriana/complicaciones , Animales , Sitios de Unión , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Pulmón/metabolismo , Pulmón/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas de Transferencia de Fosfolípidos , Neumonía Bacteriana/metabolismo , Surfactantes Pulmonares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA