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










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 209(9): 1788-1795, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36113884

RESUMO

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.


Assuntos
Proteínas F-Box , Animais , Camundongos , Abatacepte , Aloenxertos , Citocinas/metabolismo , Modelos Animais de Doenças , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Rejeição de Enxerto , Pulmão/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , RNA Mensageiro , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
2.
JCI Insight ; 4(19)2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31578312

RESUMO

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.


Assuntos
Pulmão/metabolismo , Pneumonia/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Adulto , Idoso , Animais , Citocinas/metabolismo , Feminino , Humanos , Imunidade Inata , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Simulação de Acoplamento Molecular , Pneumonia/imunologia , Pneumonia/patologia , Ligação Proteica , Transcriptoma
3.
Respir Res ; 18(1): 131, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28659178

RESUMO

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.


Assuntos
Azitromicina/farmacologia , Inflamassomos/metabolismo , Monócitos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , RNA Mensageiro/metabolismo , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Azitromicina/uso terapêutico , Linhagem Celular , Células HEK293 , Humanos , Inflamassomos/antagonistas & inibidores , Inflamassomos/genética , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/metabolismo , Monócitos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Estabilidade Proteica/efeitos dos fármacos , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética
4.
J Exp Med ; 213(6): 1029-46, 2016 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-27162139

RESUMO

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.


Assuntos
Pulmão/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Fibrose Pulmonar/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Pulmão/patologia , Camundongos , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Inibidoras de STAT Ativados/genética , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-theta , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Ubiquitina-Proteína Ligases/genética
5.
Sci Transl Med ; 7(295): 295ra109, 2015 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-26157031

RESUMO

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.


Assuntos
Imunidade Inata/fisiologia , Inflamação/fisiopatologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/fisiopatologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Técnicas de Silenciamento de Genes , Lesão Pulmonar/microbiologia , Camundongos , Fosforilação , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Pseudomonas/patogenicidade
6.
Am J Respir Cell Mol Biol ; 52(5): 622-33, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25286244

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Rejeição de Enxerto/etiologia , Mediadores da Inflamação/metabolismo , Interleucina-17/metabolismo , Transplante de Pulmão/efeitos adversos , Pulmão/metabolismo , Neutrófilos/metabolismo , Pneumonia/etiologia , Proteínas com Domínio T/metabolismo , Doença Aguda , Aloenxertos , Animais , Anticorpos/farmacologia , Ligante de CD40/imunologia , Ligante de CD40/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Quimiotaxia de Leucócito , Modelos Animais de Doenças , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Rejeição de Enxerto/prevenção & controle , Histocompatibilidade , Mediadores da Inflamação/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/imunologia , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Pneumonia/prevenção & controle , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
7.
PLoS One ; 9(12): e115937, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25549252

RESUMO

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.


Assuntos
Forsythia/química , Lignina/farmacologia , Inibidores da Fosfodiesterase 4/isolamento & purificação , Animais , Simulação por Computador , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Inflamação/tratamento farmacológico , Lignina/isolamento & purificação , Lesão Pulmonar/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Inibidores da Fosfodiesterase 4/química , Inibidores da Fosfodiesterase 4/farmacologia , Inibidores da Fosfodiesterase 4/uso terapêutico , Plantas Medicinais/química , Choque Séptico/tratamento farmacológico , Relação Estrutura-Atividade
8.
Cell Rep ; 7(2): 476-487, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24703837

RESUMO

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.


Assuntos
Antígeno B7-2/metabolismo , Proteínas F-Box/metabolismo , Mitocôndrias/metabolismo , Pneumonia/metabolismo , Proteínas Quinases/metabolismo , Adolescente , Adulto , Animais , Antígeno B7-2/genética , Estudos de Casos e Controles , Linhagem Celular , Células Cultivadas , Criança , Estabilidade Enzimática , Proteínas F-Box/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas Quinases/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L534-42, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24441869

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal , Fibroblastos/citologia , Pulmão/metabolismo , MicroRNAs/genética , Miofibroblastos/metabolismo , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Actinas/metabolismo , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Movimento Celular/genética , Proliferação de Células , Células Cultivadas , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Proteína HMGA2/metabolismo , Proteína HMGB2/metabolismo , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Queratina-19/metabolismo , Pulmão/citologia , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/genética , Proteína A4 de Ligação a Cálcio da Família S100 , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Cicatrização/genética , Proteína da Zônula de Oclusão-1/metabolismo
10.
J Immunol ; 191(10): 5247-55, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24123678

RESUMO

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.


Assuntos
Benzilaminas/uso terapêutico , Proteínas F-Box/metabolismo , Inflamação/tratamento farmacológico , Piridinas/uso terapêutico , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Animais , Benzilaminas/farmacologia , Linhagem Celular , Colite/tratamento farmacológico , Citocinas/biossíntese , Citocinas/metabolismo , Proteínas F-Box/antagonistas & inibidores , Inflamação/imunologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/tratamento farmacológico , Piridinas/farmacologia , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo
11.
Nat Immunol ; 14(5): 470-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23542741

RESUMO

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.


Assuntos
Proteínas F-Box/metabolismo , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Sepse/imunologia , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Animais , Ceco/imunologia , Ceco/cirurgia , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Motivos F-Box/genética , Proteínas F-Box/genética , Humanos , Imunomodulação , Inflamação/genética , Camundongos , Camundongos Endogâmicos C57BL , Polimorfismo Genético , Estabilidade Proteica , Infecções por Pseudomonas/genética , Pseudomonas aeruginosa/genética , RNA Interferente Pequeno/genética , Sepse/genética , Transgenes/genética
12.
FASEB J ; 27(7): 2611-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23512198

RESUMO

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.


Assuntos
Movimento Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas F-Box/metabolismo , Neuropeptídeos/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas F-Box/genética , Humanos , Immunoblotting , Camundongos , Mutação , Neuropeptídeos/genética , Fosforilação , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteólise , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Serina/genética , Serina/metabolismo , Ubiquitinação , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP
13.
Cell Cycle ; 12(4): 663-73, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23370391

RESUMO

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.


Assuntos
Calmodulina/genética , Citocinese/genética , Proteínas F-Box/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Telófase/genética , Animais , Apoptose/efeitos dos fármacos , Aurora Quinase B , Aurora Quinases , Sítios de Ligação , Calmodulina/metabolismo , Linhagem Celular Tumoral , Segregação de Cromossomos/efeitos dos fármacos , Citocinese/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Proteínas F-Box/metabolismo , Humanos , Imidazóis/farmacologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Telófase/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos
14.
J Biol Chem ; 288(9): 6306-16, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23319590

RESUMO

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.


Assuntos
Proliferação de Células , Proteínas F-Box/metabolismo , Histona Acetiltransferases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ubiquitina/metabolismo , Acetilação , Animais , Linhagem Celular , Proteínas F-Box/genética , Inativação Gênica , Histona Acetiltransferases/genética , Histonas/genética , Histonas/metabolismo , Humanos , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Camundongos , Complexo de Endopeptidases do Proteassoma/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
15.
Cell Cycle ; 11(4): 721-9, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22306998

RESUMO

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.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Ciclo Celular/fisiologia , Mitose/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Aurora Quinase A , Aurora Quinases , Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Transferência Ressonante de Energia de Fluorescência , Humanos , Camundongos , Mitose/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Ligases SKP Culina F-Box/genética , Ubiquitinação/genética , Ubiquitinação/fisiologia
16.
Blood ; 119(13): 3132-41, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22323446

RESUMO

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.


Assuntos
Proliferação de Células , Ciclina D2/metabolismo , Proteínas F-Box/fisiologia , Leucemia/patologia , Animais , Apoptose/genética , Apoptose/fisiologia , Calmodulina/metabolismo , Calmodulina/fisiologia , Células Cultivadas , Regulação para Baixo/fisiologia , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Regulação Leucêmica da Expressão Gênica , Humanos , Células K562 , Leucemia/genética , Leucemia/metabolismo , Camundongos , Proteólise , Transfecção , Células U937 , Ubiquitinação/genética
17.
Cell Cycle ; 10(20): 3487-94, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22024926

RESUMO

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.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Ciclina D3/metabolismo , Proteínas F-Box/metabolismo , Mitose/fisiologia , Modelos Biológicos , Ubiquitina-Proteína Ligases/metabolismo , Análise de Variância , Animais , Linhagem Celular , Ciclina D3/genética , Fluorescência , Immunoblotting , Imunoprecipitação , Camundongos , Interferência de RNA , Fatores de Tempo , Ubiquitinação
18.
Mol Cell Biol ; 31(9): 1905-20, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21343341

RESUMO

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.


Assuntos
Calmodulina/metabolismo , Proteínas F-Box/metabolismo , Interações Hospedeiro-Patógeno , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Chaperonina com TCP-1/metabolismo , Complexo de Golgi/metabolismo , Complexo de Golgi/microbiologia , Pulmão/metabolismo , Pulmão/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilcolinas/metabolismo , Infecções por Pseudomonas/microbiologia
19.
J Biol Chem ; 286(4): 2719-27, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21068446

RESUMO

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.


Assuntos
1,2-Dipalmitoilfosfatidilcolina/biossíntese , 1-Acilglicerofosfocolina O-Aciltransferase/metabolismo , Lipopolissacarídeos/farmacologia , Surfactantes Pulmonares/metabolismo , Mucosa Respiratória/metabolismo , Ubiquitinação/efeitos dos fármacos , Proteínas Contendo Repetições de beta-Transducina/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/genética , 1-Acilglicerofosfocolina O-Aciltransferase/genética , Animais , Células Cultivadas , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Camundongos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Sepse/genética , Sepse/metabolismo , Ubiquitinação/genética , Proteínas Contendo Repetições de beta-Transducina/genética
20.
Nat Med ; 16(10): 1120-1127, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20852622

RESUMO

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.


Assuntos
Adenosina Trifosfatases/fisiologia , Cardiolipinas/fisiologia , Lesão Pulmonar/etiologia , Pneumonia Bacteriana/complicações , Animais , Sítios de Ligação , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Pulmão/metabolismo , Pulmão/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transferência de Fosfolipídeos , Pneumonia Bacteriana/metabolismo , Surfactantes Pulmonares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...