Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37873169

RESUMO

Targeted protein degradation by the ubiquitin-proteasome system is an essential mechanism regulating cellular division. The kinase PLK1 coordinates protein degradation at the G2/M phase of the cell cycle by promoting the binding of substrates to the E3 ubiquitin ligase SCFßTrCP. However, the magnitude to which PLK1 shapes the mitotic proteome has not been characterized. Combining deep, quantitative proteomics with pharmacologic PLK1 inhibition (PLK1i), we identified more than 200 proteins whose abundances were increased by PLK1i at G2/M. We validate many new PLK1-regulated proteins, including several substrates of the cell cycle E3 SCFCyclin F, demonstrating that PLK1 promotes proteolysis through at least two distinct SCF-family E3 ligases. Further, we found that the protein kinase A anchoring protein AKAP2 is cell cycle regulated and that its mitotic degradation is dependent on the PLK1/ßTrCP-signaling axis. Interactome analysis revealed that the strongest interactors of AKAP2 function in signaling networks regulating proliferation, including MAPK, AKT, and Hippo. Altogether, our data demonstrate that PLK1 coordinates a widespread program of protein breakdown at G2/M. We propose that dynamic proteolytic changes mediated by PLK1 integrate proliferative signals with the core cell cycle machinery during cell division. This has potential implications in malignancies where PLK1 is aberrantly regulated.

2.
Elife ; 102021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34851822

RESUMO

Cell cycle gene expression programs fuel proliferation and are universally dysregulated in cancer. The retinoblastoma (RB)-family of proteins, RB1, RBL1/p107, and RBL2/p130, coordinately represses cell cycle gene expression, inhibiting proliferation, and suppressing tumorigenesis. Phosphorylation of RB-family proteins by cyclin-dependent kinases is firmly established. Like phosphorylation, ubiquitination is essential to cell cycle control, and numerous proliferative regulators, tumor suppressors, and oncoproteins are ubiquitinated. However, little is known about the role of ubiquitin signaling in controlling RB-family proteins. A systems genetics analysis of CRISPR/Cas9 screens suggested the potential regulation of the RB-network by cyclin F, a substrate recognition receptor for the SCF family of E3 ligases. We demonstrate that RBL2/p130 is a direct substrate of SCFcyclin F. We map a cyclin F regulatory site to a flexible linker in the p130 pocket domain, and show that this site mediates binding, stability, and ubiquitination. Expression of a mutant version of p130, which cannot be ubiquitinated, severely impaired proliferative capacity and cell cycle progression. Consistently, we observed reduced expression of cell cycle gene transcripts, as well a reduced abundance of cell cycle proteins, analyzed by quantitative, iterative immunofluorescent imaging. These data suggest a key role for SCFcyclin F in the CDK-RB network and raise the possibility that aberrant p130 degradation could dysregulate the cell cycle in human cancers.


Assuntos
Ciclinas/genética , Proteína p130 Retinoblastoma-Like/genética , Retinoblastoma/genética , Fator de Células-Tronco/genética , Ciclinas/metabolismo , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Proteína p130 Retinoblastoma-Like/metabolismo , Fator de Células-Tronco/metabolismo
3.
Mol Cell Biol ; 41(2)2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33168699

RESUMO

The ubiquitin-proteasome system is essential for cell cycle progression. Cyclin F is a cell cycle-regulated substrate adapter F-box protein for the Skp1, CUL1, and F-box protein (SCF) family of E3 ubiquitin ligases. Despite its importance in cell cycle progression, identifying cyclin F-bound SCF complex (SCFCyclin F) substrates has remained challenging. Since cyclin F overexpression rescues a yeast mutant in the cdc4 gene, we considered the possibility that other genes that genetically modify cdc4 mutant lethality could also encode cyclin F substrates. We identified the mitochondrial and cytosolic deacylating enzyme sirtuin 5 (SIRT5) as a novel cyclin F substrate. SIRT5 has been implicated in metabolic processes, but its connection to the cell cycle is not known. We show that cyclin F interacts with and controls the ubiquitination, abundance, and stability of SIRT5. We show SIRT5 knockout results in a diminished G1 population and a subsequent increase in both S and G2/M. Global proteomic analyses reveal cyclin-dependent kinase (CDK) signaling changes congruent with the cell cycle changes in SIRT5 knockout cells. Together, these data demonstrate that SIRT5 is regulated by cyclin F and suggest a connection between SIRT5, cell cycle regulation, and metabolism.


Assuntos
Proteínas de Ciclo Celular/genética , Ciclo Celular/genética , Proteínas F-Box/genética , Regulação Fúngica da Expressão Gênica , Processamento de Proteína Pós-Traducional , Proteínas Ligases SKP Culina F-Box/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Sirtuínas/genética , Ubiquitina-Proteína Ligases/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Perfilação da Expressão Gênica , Genes Letais , Células HEK293 , Células HeLa , Humanos , Mutação , Proteínas Ligases SKP Culina F-Box/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Sirtuínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
PLoS Biol ; 18(12): e3000975, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33306668

RESUMO

The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase and critical regulator of cell cycle progression. Despite its vital role, it has remained challenging to globally map APC/C substrates. By combining orthogonal features of known substrates, we predicted APC/C substrates in silico. This analysis identified many known substrates and suggested numerous candidates. Unexpectedly, chromatin regulatory proteins are enriched among putative substrates, and we show experimentally that several chromatin proteins bind APC/C, oscillate during the cell cycle, and are degraded following APC/C activation, consistent with being direct APC/C substrates. Additional analysis revealed detailed mechanisms of ubiquitylation for UHRF1, a key chromatin regulator involved in histone ubiquitylation and DNA methylation maintenance. Disrupting UHRF1 degradation at mitotic exit accelerates G1-phase cell cycle progression and perturbs global DNA methylation patterning in the genome. We conclude that APC/C coordinates crosstalk between cell cycle and chromatin regulatory proteins. This has potential consequences in normal cell physiology, where the chromatin environment changes depending on proliferative state, as well as in disease.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Cromatina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ciclossomo-Complexo Promotor de Anáfase/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Cromatina/genética , Simulação por Computador , Células HEK293 , Células HeLa , Humanos , Processamento de Proteína Pós-Traducional , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitinação
5.
ACS Chem Biol ; 15(8): 2164-2174, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32589399

RESUMO

Thiopeptide antibiotics are emerging clinical candidates that exhibit potent antibacterial activity against a variety of intracellular pathogens, including Mycobacterium tuberculosis (Mtb). Many thiopeptides directly inhibit bacterial growth by disrupting protein synthesis. However, recent work has shown that one thiopeptide, thiostrepton (TSR), can also induce autophagy in infected macrophages, which has the potential to be exploited for host-directed therapies against intracellular pathogens, such as Mtb. To better define the therapeutic potential of this class of antibiotics, we studied the host-directed effects of a suite of natural thiopeptides that spans five structurally diverse thiopeptide classes, as well as several analogs. We discovered that thiopeptides as a class induce selective autophagic removal of mitochondria, known as mitophagy. This activity is independent of other biological activities, such as proteasome inhibition or antibiotic activity. We also find that many thiopeptides exhibit potent activity against intracellular Mtb in macrophage infection models. However, the thiopeptide-induced mitophagy occurs outside of pathogen-containing autophagosomes and does not appear to contribute to thiopeptide control of intracellular Mtb. These results expand basic understanding of thiopeptide biology and provide key guidance for the development of new thiopeptide antibiotics and host-directed therapeutics.


Assuntos
Mitofagia/efeitos dos fármacos , Peptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Compostos de Sulfidrila/química , Animais , Antibacterianos/farmacologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Proteína Forkhead Box M1/metabolismo , Camundongos , Mycobacterium tuberculosis/efeitos dos fármacos , Peptídeos/química , Fosforilação , Células RAW 264.7
6.
Mol Biol Cell ; 31(8): 725-740, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31995441

RESUMO

E2F8 is a transcriptional repressor that antagonizes E2F1 at the crossroads of the cell cycle, apoptosis, and cancer. Previously, we discovered that E2F8 is a direct target of the APC/C ubiquitin ligase. Nevertheless, it remains unknown how E2F8 is dynamically controlled throughout the entirety of the cell cycle. Here, using newly developed human cell-free systems that recapitulate distinct inter-mitotic and G1 phases and a continuous transition from prometaphase to G1, we reveal an interlocking dephosphorylation switch coordinating E2F8 degradation with mitotic exit and the activation of APC/CCdh1. Further, we uncover differential proteolysis rates for E2F8 at different points within G1 phase, accounting for its accumulation in late G1 while APC/CCdh1 is still active. Finally, we demonstrate that the F-box protein Cyclin F regulates E2F8 in G2-phase. Altogether, our data define E2F8 regulation throughout the cell cycle, illuminating an extensive coordination between phosphorylation, ubiquitination and transcription in mammalian cell cycle.


Assuntos
Ciclo Celular/fisiologia , Proteínas Repressoras/metabolismo , Motivos de Aminoácidos , Subunidade Apc1 do Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Sistema Livre de Células , Ciclinas/metabolismo , Fator de Transcrição E2F1/metabolismo , Fase G1/fisiologia , Fase G2/fisiologia , Células HeLa , Humanos , Mitose/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteólise , Proteínas Recombinantes/metabolismo , Ubiquitinação
7.
Cell Rep ; 26(11): 3076-3086.e6, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30865895

RESUMO

The transcription factor FOXM1 contributes to cell cycle progression and is significantly upregulated in basal-like breast cancer (BLBC). Despite its importance in normal and cancer cell cycles, we lack a complete understanding of mechanisms that regulate FOXM1. We identified USP21 in an RNAi-based screen for deubiquitinases that control FOXM1 abundance. USP21 increases the stability of FOXM1, and USP21 binds and deubiquitinates FOXM1 in vivo and in vitro, indicating a direct enzyme-substrate relationship. Depleting USP21 downregulates the FOXM1 transcriptional network and causes a significant delay in cell cycle progression. Significantly, USP21 depletion sensitized BLBC cell lines and mouse xenograft tumors to paclitaxel, an anti-mitotic, frontline therapy in BLBC treatment. USP21 is the most frequently amplified deubiquitinase in BLBC patient tumors, and its amplification co-occurs with the upregulation of FOXM1 protein. Altogether, these data suggest a role for USP21 in the proliferation and potentially treatment of FOXM1-high, USP21-high BLBC.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Ciclo Celular , Resistencia a Medicamentos Antineoplásicos , Proteína Forkhead Box M1/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Paclitaxel/uso terapêutico , Ubiquitinação , Animais , Feminino , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ubiquitina Tiolesterase/metabolismo
8.
Int J Mol Sci ; 19(12)2018 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-30563203

RESUMO

Mitogen-activated protein kinase phosphatase (Mkp)-1 exerts its anti-inflammatory activities during Gram-negative sepsis by deactivating p38 and c-Jun N-terminal kinase (JNK). We have previously shown that Mkp-1+/+ mice, but not Mkp-1-/- mice, exhibit hypertriglyceridemia during severe sepsis. However, the regulation of hepatic lipid stores and the underlying mechanism of lipid dysregulation during sepsis remains an enigma. To understand the molecular mechanism underlying the sepsis-associated metabolic changes and the role of Mkp-1 in the process, we infected Mkp-1+/+ and Mkp-1-/- mice with Escherichia coli i.v., and assessed the effects of Mkp-1 deficiency on tissue lipid contents. We also examined the global gene expression profile in the livers via RNA-seq. We found that in the absence of E. coli infection, Mkp-1 deficiency decreased liver triglyceride levels. Upon E. coli infection, Mkp-1+/+ mice, but not Mkp-1-/- mice, developed hepatocyte ballooning and increased lipid deposition in the livers. E. coli infection caused profound changes in the gene expression profile of a large number of proteins that regulate lipid metabolism in wildtype mice, while these changes were substantially disrupted in Mkp-1-/- mice. Interestingly, in Mkp-1+/+ mice E. coli infection resulted in downregulation of genes that facilitate fatty acid synthesis but upregulation of Cd36 and Dgat2, whose protein products mediate fatty acid uptake and triglyceride synthesis, respectively. Taken together, our studies indicate that sepsis leads to a substantial change in triglyceride metabolic gene expression programs and Mkp-1 plays an important role in this process.


Assuntos
Fosfatase 1 de Especificidade Dupla/deficiência , Infecções por Escherichia coli/genética , Perfilação da Expressão Gênica/métodos , Metabolismo dos Lipídeos , Sepse/genética , Animais , Infecções por Escherichia coli/metabolismo , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Fígado/química , Redes e Vias Metabólicas , Camundongos , Sepse/metabolismo , Sepse/microbiologia , Análise de Sequência de RNA , Triglicerídeos/metabolismo
9.
Cell Rep ; 20(13): 3212-3222, 2017 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-28954236

RESUMO

The oncogenic AKT kinase is a key regulator of apoptosis, cell growth, and cell-cycle progression. Despite its important role in proliferation, it remains largely unknown how AKT is mechanistically linked to the cell cycle. We show here that cyclin F, a substrate receptor F-box protein for the SCF (Skp1/Cul1/F-box) family of E3 ubiquitin ligases, is a bona fide AKT substrate. Cyclin F expression oscillates throughout the cell cycle, a rare feature among the 69 human F-box proteins, and all of its known substrates are involved in proliferation. AKT phosphorylation of cyclin F enhances its stability and promotes assembly into productive E3 ligase complexes. Importantly, expression of mutant versions of cyclin F that cannot be phosphorylated by AKT impair cell-cycle entry. Our data suggest that cyclin F transmits mitogen signaling through AKT to the core cell-cycle machinery. This discovery has potential implications for proliferative control in malignancies where AKT is activated.


Assuntos
Ciclo Celular/fisiologia , Ciclinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Fosforilação , Transdução de Sinais
10.
Mol Cell Biol ; 37(13)2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28416635

RESUMO

The oncogenic transcription factor FoxM1 plays a vital role in cell cycle progression, is activated in numerous human malignancies, and is linked to chromosome instability. We characterize here a cullin 4-based E3 ubiquitin ligase and its substrate receptor, VprBP/DCAF1 (CRL4VprBP), which we show regulate FoxM1 ubiquitylation and degradation. Paradoxically, we also found that the substrate receptor VprBP is a potent FoxM1 activator. VprBP depletion reduces expression of FoxM1 target genes and impairs mitotic entry, whereas ectopic VprBP expression strongly activates a FoxM1 transcriptional reporter. VprBP binding to CRL4 is reduced during mitosis, and our data suggest that VprBP activation of FoxM1 is ligase independent. This implies a nonproteolytic activation mechanism that is reminiscent of, yet distinct from, the ubiquitin-dependent transactivation of the oncoprotein Myc by other E3s. Significantly, VprBP protein levels were upregulated in high-grade serous ovarian patient tumors, where the FoxM1 signature is amplified. These data suggest that FoxM1 abundance and activity are controlled by VprBP and highlight the functional repurposing of E3 ligase substrate receptors independent of the ubiquitin system.


Assuntos
Proteínas de Transporte/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Proteína Forkhead Box M1/metabolismo , Neoplasias Ovarianas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Transporte/genética , Ciclo Celular , Instabilidade Cromossômica , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Feminino , Proteína Forkhead Box M1/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Proteínas Serina-Treonina Quinases , Proteólise , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
11.
PLoS One ; 11(1): e0147113, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26760500

RESUMO

There is currently tremendous interest in developing anti-cancer therapeutics targeting cell signaling pathways important for both cancer cell metabolism and growth. Several epidemiological studies have shown that diabetic patients taking metformin have a decreased incidence of pancreatic cancer. This has prompted efforts to evaluate metformin, a drug with negligible toxicity, as a therapeutic modality in pancreatic cancer. Preclinical studies in cell line xenografts and one study in patient-derived xenograft (PDX) models were promising, while recently published clinical trials showed no benefit to adding metformin to combination therapy regimens for locally advanced and metastatic pancreatic cancer. PDX models in which patient tumors are directly engrafted into immunocompromised mice have been shown to be excellent preclinical models for biomarker discovery and therapeutic development. We evaluated the response of four PDX tumor lines to metformin treatment and found that all four of our PDX lines were resistant to metformin. We found that the mechanisms of resistance may occur through lack of sustained activation of adenosine monophosphate-activated protein kinase (AMPK) or downstream reactivation of the mammalian target of rapamycin (mTOR). Moreover, combined treatment with metformin and mTOR inhibitors failed to improve responses in cell lines, which further indicates that metformin alone or in combination with mTOR inhibitors will be ineffective in patients, and that resistance to metformin may occur through multiple pathways. Further studies are required to better understand these mechanisms of resistance and inform potential combination therapies with metformin and existing or novel therapeutics.


Assuntos
Hipoglicemiantes/farmacologia , Metformina/farmacologia , Neoplasias Pancreáticas/patologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Expressão Gênica , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa , Transdução de Sinais , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Am J Transl Res ; 1(3): 267-82, 2009 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-19956437

RESUMO

Tripterygium wilfordii Hook F. has been used for centuries in traditional Chinese medicine to treat rheumatoid arthritis, an autoimmune disease associated with increased production of the pro-inflammatory cytokine, tumor necrosis factor (TNF)-alpha. Triptolide is a compound originally purified from T. wilfordii Hook F. and has potent anti-inflammatory and immunosuppressant activities. In this study, we investigated the effect of triptolide on the global gene expression patterns of macrophages treated with lipopolysaccharide (LPS). We found that LPS stimulation resulted in >5-fold increase in expression of 117 genes, and triptolide caused a >50% inhibition in 47 of the LPS-inducible 117 genes. A large portion of the genes that were strongly induced by LPS and significantly inhibited by triptolide were pro-inflammatory cytokine and chemokine genes, including TNF-alpha, IL-1beta, and IL-6. Interestingly, LPS also induced the expression of micro-RNA-155 (miR-155) precursor, BIC, which was inhibited by triptolide. Confirming the cDNA array results, we demonstrated that triptolide blocked the induction of these pro-inflammatory cytokines as well as miR-155 in a dose-dependent manner. Profound inhibition of pro-inflammatory cytokine expression was observed at concentrations as low as 10-50 nM. However, triptolide neither inhibited the phosphorylation or degradation of IkappaBalpha after LPS stimulation, nor affected the DNA-binding activity of NF-kappaB. Surprisingly, we found that triptolide not only inhibited NF-kappaB-regulated reporter transcription, but also dramatically blocked the activity of other transcription factors. Our study offers a plausible explanation of the therapeutic mechanism of T. wilfordii Hook F.

13.
J Immunol ; 183(11): 7411-9, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19890037

RESUMO

MAPKs are crucial for TNF-alpha and IL-6 production by innate immune cells in response to TLR ligands. MAPK phosphatase 1 (Mkp-1) deactivates p38 and JNK, abrogating the inflammatory response. We have previously demonstrated that Mkp-1(-/-) mice exhibit exacerbated inflammatory cytokine production and increased mortality in response to challenge with LPS and heat-killed Staphylococcus aureus. However, the function of Mkp-1 in host defense during live Gram-negative bacterial infection remains unclear. We challenged Mkp-1(+/+) and Mkp-1(-/-) mice with live Escherichia coli i.v. to examine the effects of Mkp-1 deficiency on animal survival, bacterial clearance, metabolic activity, and cytokine production. We found that Mkp-1 deficiency predisposed animals to accelerated mortality and was associated with more robust production of TNF-alpha, IL-6 and IL-10, greater bacterial burden, altered cyclooxygenase-2 and iNOS expression, and substantial changes in the mobilization of energy stores. Likewise, knockout of Mkp-1 also sensitized mice to sepsis caused by cecal ligation and puncture. IL-10 inhibition by neutralizing Ab or genetic deletion alleviated increased bacterial burden. Treatment with the bactericidal antibiotic gentamicin, given 3 h after Escherichia coli infection, protected Mkp-1(+/+) mice from septic shock but had no effect on Mkp-1(-/-) mice. Thus, during Gram-negative bacterial sepsis Mkp-1 not only plays a critical role in the regulation of cytokine production but also orchestrates the bactericidal activities of the innate immune system and controls the metabolic response to stress.


Assuntos
Fosfatase 1 de Especificidade Dupla/imunologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/metabolismo , Inflamação/imunologia , Sepse/imunologia , Animais , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/imunologia , Fosfatase 1 de Especificidade Dupla/deficiência , Ensaio de Imunoadsorção Enzimática , Escherichia coli , Glucose/metabolismo , Glicogênio/metabolismo , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/metabolismo , Hiperlipidemias/metabolismo , Hiperlipidemias/microbiologia , Inflamação/metabolismo , Inflamação/microbiologia , Interleucina-10/biossíntese , Interleucina-10/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Metabolismo dos Lipídeos/imunologia , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/imunologia , Sepse/microbiologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
14.
J Biol Chem ; 284(40): 27123-34, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19651781

RESUMO

Inducible nitric-oxide (NO) synthase (iNOS) plays a critical role in the eradication of intracellular pathogens. However, the excessive production of NO by iNOS has also been implicated in the pathogenesis of septic shock syndrome. Previously, we have demonstrated that mice deficient in mitogen-activated protein kinase phosphatase-1 (MKP-1) exhibit exaggerated inflammatory responses and rapidly succumb to lipopolysaccharide (LPS). In response to LPS, MKP-1(-/-) mice produce greater amounts of inflammatory cytokines and NO than do wild-type mice, and the MKP-1(-/-) mice exhibit severe hypotension. To understand the molecular basis for the increase in NO production, we studied the role of MKP-1 in the regulation of iNOS expression. We found that LPS challenge elicited a stronger iNOS induction in MKP-1 knock-out mice than in wild-type mice. Likewise, LPS treatment also resulted in greater iNOS expression in macrophages from MKP-1(-/-) mice than in macrophages from wild-type mice. Both accelerated gene transcription and enhanced mRNA stability contribute to the increases in iNOS expression in LPS-stimulated MKP-1(-/-) macrophages. We found that STAT-1, a transcription factor known to mediate iNOS induction by interferon-gamma, was more potently activated by LPS in MKP-1(-/-) macrophages than in wild-type cells. MicroRNA array analysis indicated that microRNA (miR)-155 expression was increased in MKP-1-deficient macrophages compared with wild-type macrophages. Transfection of miR-155 attenuated the expression of Suppressor of Cytokine Signal (SOCS)-1 and enhanced the expression of iNOS. Our results suggest that MKP-1 may negatively regulate iNOS expression by controlling the expression of miR-155 and consequently the STAT pathway via SOCS-1.


Assuntos
Fosfatase 1 de Especificidade Dupla/metabolismo , Regulação Enzimológica da Expressão Gênica , Óxido Nítrico Sintase Tipo II/genética , Animais , Linhagem Celular , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Técnicas de Inativação de Genes , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação/efeitos dos fármacos , Processamento Pós-Transcricional do RNA , Estabilidade de RNA , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Fatores de Transcrição STAT/metabolismo , Ativação Transcricional , Tirosina/metabolismo
15.
Life Sci ; 83(19-20): 671-80, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18845168

RESUMO

AIMS: We have previously shown that glucocorticoids induce the expression of MAP kinase phosphatase (Mkp)(a)-1 in innate immune cells. Since Mkp-1 is a critical negative regulator of the innate immune response, we hypothesize that Mkp-1 plays a significant role in the anti-inflammatory action of glucocorticoids. The specific aim of the present study is to understand the role of Mkp-1 in the anti-inflammatory function of glucocorticoids. MAIN METHODS: Wild-type and Mkp-1(-/-) mice were treated with different doses of dexamethasone and then challenged with different doses of lipopolysaccharide (LPS). The survival and blood cytokines were assessed. The effects of dexamethasone on cytokine production in wild-type and Mkp-1(-/-) primary macrophages ex vivo were also examined. KEY FINDINGS: We found that dexamethasone induced the expression of Mkp-1 in vivo. Dexamethasone treatment completely protected wild-type mice from the mortality caused by a relatively high dose of LPS. However, dexamethasone treatment offered only a partial protection to Mkp-1(-/-) mice. Dexamethasone attenuated TNF-alpha production in both wild-type and Mkp-1(-/-) mice challenged with LPS, although TNF-alpha production in Mkp-1(-/-) mice was significantly more robust than that in wild-type mice. Dexamethasone pretreatment shortened the duration of p38 and JNK activation in LPS-stimulated wild-type macrophages, but had little effect on p38 or JNK activation in similarly treated Mkp-1(-/-) macrophages. SIGNIFICANCE: Our results indicate that the inhibition of p38 and JNK activities by glucocorticoids is mediated by enhanced Mkp-1 expression. These results demonstrate that dexamethasone exerts its anti-inflammatory effects through both Mkp-1-dependent and Mkp-1-indepent mechanisms.


Assuntos
Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Fosfatase 1 de Especificidade Dupla/fisiologia , Endotoxemia/enzimologia , Endotoxemia/prevenção & controle , Animais , Northern Blotting , Western Blotting , Citocinas/biossíntese , Fosfatase 1 de Especificidade Dupla/genética , Ativação Enzimática/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Lipopolissacarídeos/toxicidade , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/enzimologia , Camundongos , Camundongos Knockout , Baço/citologia , Baço/efeitos dos fármacos , Baço/enzimologia
16.
Cell Signal ; 19(7): 1372-82, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17512700

RESUMO

Mitogen-activated protein (MAP) kinase cascades are signal transduction pathways that play pivotal regulatory roles in the biosynthesis of pro-inflammatory cytokines. MAP kinase phosphatase (MKP)-1, an archetypal member of the MKP family, is essential for the dephosphorylation/deactivation of MAP kinases p38 and JNK. Earlier studies conducted using cultured immortalized macrophages provided compelling evidence indicating that MKP-1 deactivates p38 and JNK, thereby limiting pro-inflammatory cytokine biosynthesis in innate immune cells exposed to microbial components. Recent studies employing MKP-1 knockout mice have confirmed the central function of MKP-1 in the feedback control of p38 and JNK activity as well as the crucial physiological function of MKP-1 as a negative regulator of the synthesis of pro-inflammatory cytokines in vivo. MKP-1 was shown to be a major feedback regulator of the innate immune response and to play a critical role in preventing septic shock and multi-organ dysfunction during pathogenic infection. In this review, we will update the studies on the biochemical properties and the regulation of MKP-1, and summarize our understanding on the physiological function of this key phosphatase in the innate immune response.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Imunidade Inata/imunologia , Fosfoproteínas Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , Proteínas de Ciclo Celular/química , Citocinas/biossíntese , Fosfatase 1 de Especificidade Dupla , Humanos , Proteínas Imediatamente Precoces/química , Fatores Imunológicos/metabolismo , Inflamação/microbiologia , Fosfoproteínas Fosfatases/química , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/química
17.
J Immunol ; 178(8): 5312-20, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17404316

RESUMO

MAPK phosphatase (MKP)-1 is an archetypal member of the dual specificity protein phosphatase family that dephosphorylates MAPK. We have previously demonstrated that MKP-1 acts as a negative regulator of p38 and JNK in immortalized macrophages after stimulation with peptidoglycan isolated from Gram-positive bacteria. To define the physiological function of MKP-1 during Gram-positive bacterial infection, we studied the innate immune responses to Gram-positive bacteria using Mkp-1 knockout (KO) mice. We found that Mkp-1(-/-) macrophages exhibited prolonged activation of p38 and JNK, but not of ERK, following exposure to either peptidoglycan or lipoteichoic acid. Compared with wild-type (WT) macrophages, Mkp-1(-/-) macrophages produced more proinflammatory cytokines such as TNF-alpha and IL-6. Moreover, after challenge with peptidoglycan, lipoteichoic acid, live or heat-killed Staphylococcus aureus bacteria, Mkp-1 KO mice also mounted a more robust production of cytokines and chemokines, including TNF-alpha, IL-6, IL-10, and MIP-1alpha, than did WT mice. Accordingly, Mkp-1 KO mice also exhibited greater NO production, more robust neutrophil infiltration, and more severe organ damage than did WT mice. Surprisingly, WT and Mkp-1 KO mice exhibited no significant difference in either bacterial load or survival rates when infected with live S. aureus. However, in response to challenge with heat-killed S. aureus, Mkp-1 KO mice exhibited a substantially higher mortality rate compared with WT mice. Our studies indicate that MKP-1 plays a critical role in the inflammatory response to Gram-positive bacterial infection. MKP-1 serves to limit the inflammatory reaction by inactivating JNK and p38, thus preventing multiorgan failure caused by exaggerated inflammatory responses.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Infecções por Bactérias Gram-Positivas/imunologia , Proteínas Imediatamente Precoces/fisiologia , Inflamação/etiologia , Fosfoproteínas Fosfatases/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Animais , Parede Celular/imunologia , Citocinas/biossíntese , Fosfatase 1 de Especificidade Dupla , Proteínas Imediatamente Precoces/deficiência , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Peptidoglicano/farmacologia , Peroxidase/metabolismo , Fosfoproteínas Fosfatases/deficiência , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/deficiência , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/mortalidade , Ácidos Teicoicos/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Am J Physiol Cell Physiol ; 293(2): C632-40, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17442735

RESUMO

L-Arginine (L-arg) is metabolized to nitric oxide (NO) by inducible NO synthase (iNOS) or to urea and L-ornithine (L-orn) by arginase. NO is involved in the inflammatory response, whereas arginase is the first step in polyamine and proline synthesis necessary for tissue repair and wound healing. Mitogen-activated protein kinases (MAPK) mediate LPS-induced iNOS expression, and MAPK phosphatase-1 (MKP-1) plays a crucial role in limiting MAPK signaling in macrophages. We hypothesized that MKP-1, by attenuating iNOS expression, acts as a switch changing L-arg metabolism from NO production to L-orn production after endotoxin administration. To test this hypothesis, we performed studies in RAW264.7 macrophages stably transfected with an MKP-1 expression vector in thioglyollate-elicited peritoneal macrophages harvested from wild-type and Mkp-1(-/-) mice, as well as in vivo in wild-type and Mkp-1(-/-) mice. We found that overexpression of MKP-1 resulted in lower iNOS expression and NO production but greater urea production in response to LPS. Although deficiency of MKP-1 resulted in greater iNOS expression and NO production and lower urea production in response to LPS, neither the overexpression nor the deficiency of MKP-1 had any substantial effect on the expression of the arginases.


Assuntos
Arginase/metabolismo , Arginina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , Arginase/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular , Fosfatase 1 de Especificidade Dupla , Regulação Enzimológica da Expressão Gênica , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Inflamação/enzimologia , Inflamação/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Ornitina/metabolismo , Fosfoproteínas Fosfatases/deficiência , Fosfoproteínas Fosfatases/genética , Inibidores de Proteínas Quinases/farmacologia , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/deficiência , Proteínas Tirosina Fosfatases/genética , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , Ureia/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Biochemistry ; 45(49): 14740-8, 2006 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-17144666

RESUMO

Inhibitor of apoptosis (IAP) proteins regulate programmed cell death by inhibiting members of the caspase family of proteases. The X-chromosome-linked IAP (XIAP) contains three baculovirus IAP repeat (BIR) domains, which bind directly to the N-termini of target proteins including those of caspases-3, -7, and -9. In the present study, we defined the consensus sequences of the motifs that interact with the three BIR domains in an unbiased manner. A combinatorial peptide library containing four random residues at the N-terminus was constructed and screened using BIR domains as probes. We found that the BIR3 domain binds a highly specific motif containing an alanine or valine at the N-terminus (P1 position), an arginine or proline at the P3 position, and a hydrophobic residue (Phe, Ile, and Tyr) at the P4 position. The BIR2-binding motif is less stringent. Although it still requires an N-terminal alanine, it tolerates a wide variety of amino acids at P2-P4 positions. The BIR1 failed to bind to any peptides in the library. SPR analysis of individually synthesized peptides confirmed the library screening results. Database searches with the BIR2- and BIR3-binding consensus sequences revealed a large number of potential target proteins. The combinatorial library method should be readily applicable to other BIR domains or other types of protein modular domains.


Assuntos
Técnicas de Química Combinatória/métodos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/química , Sequência de Aminoácidos , Sequência de Bases , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Caspases/metabolismo , Sequência Consenso , Primers do DNA , Humanos , Oligopeptídeos/química , Biblioteca de Peptídeos , Reação em Cadeia da Polimerase , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
20.
J Exp Med ; 203(1): 131-40, 2006 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-16380513

RESUMO

Septic shock is a leading cause of morbidity and mortality. However, genetic factors predisposing to septic shock are not fully understood. Excessive production of proinflammatory cytokines, particularly tumor necrosis factor (TNF)-alpha, and the resultant severe hypotension play a central role in the pathophysiological process. Mitogen-activated protein (MAP) kinase cascades are crucial in the biosynthesis of proinflammatory cytokines. MAP kinase phosphatase (MKP)-1 is an archetypal member of the dual specificity protein phosphatase family that dephosphorylates MAP kinase. Thus, we hypothesize that knockout of the Mkp-1 gene results in prolonged MAP kinase activation, augmented cytokine production, and increased susceptibility to endotoxic shock. Here, we show that knockout of Mkp-1 substantially sensitizes mice to endotoxic shock induced by lipopolysaccharide (LPS) challenge. We demonstrate that upon LPS challenge, Mkp-1-/- cells exhibit prolonged p38 and c-Jun NH2-terminal kinase activation as well as enhanced TNF-alpha and interleukin (IL)-6 production compared with wild-type cells. After LPS challenge, Mkp-1 knockout mice produce dramatically more TNF-alpha, IL-6, and IL-10 than do wild-type mice. Consequently, Mkp-1 knockout mice develop severe hypotension and multiple organ failure, and exhibit a remarkable increase in mortality. Our studies demonstrate that MKP-1 is a pivotal feedback control regulator of the innate immune responses and plays a critical role in suppressing endotoxin shock.


Assuntos
Proteínas de Ciclo Celular/imunologia , Proteínas Imediatamente Precoces/imunologia , Fosfoproteínas Fosfatases/imunologia , Proteínas Tirosina Fosfatases/imunologia , Choque Séptico/prevenção & controle , Animais , Proteínas de Ciclo Celular/genética , Células Cultivadas , Citocinas/imunologia , Células Dendríticas/imunologia , Fosfatase 1 de Especificidade Dupla , Proteínas Imediatamente Precoces/deficiência , Proteínas Imediatamente Precoces/genética , Imunidade Inata , Lipopolissacarídeos , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfoproteínas Fosfatases/deficiência , Fosfoproteínas Fosfatases/genética , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/deficiência , Proteínas Tirosina Fosfatases/genética , Choque Séptico/mortalidade , Baço/citologia , Baço/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...