Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33798093

RESUMEN

The c-Jun N-terminal kinase (JNK) signaling pathway mediates adaptation to stress signals and has been associated with cell death, cell proliferation, and malignant transformation in the liver. However, up to now, its function was experimentally studied mainly in young mice. By generating mice with combined conditional ablation of Jnk1 and Jnk2 in liver parenchymal cells (LPCs) (JNK1/2LPC-KO mice; KO, knockout), we unraveled a function of the JNK pathway in the regulation of liver homeostasis during aging. Aging JNK1/2LPC-KO mice spontaneously developed large biliary cysts that originated from the biliary cell compartment. Mechanistically, we could show that cyst formation in livers of JNK1/2LPC-KO mice was dependent on receptor-interacting protein kinase 1 (RIPK1), a known regulator of cell survival, apoptosis, and necroptosis. In line with this, we showed that RIPK1 was overexpressed in the human cyst epithelium of a subset of patients with polycystic liver disease. Collectively, these data reveal a functional interaction between JNK signaling and RIPK1 in age-related progressive cyst development. Thus, they provide a functional linkage between stress adaptation and programmed cell death (PCD) in the maintenance of liver homeostasis during aging.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades de los Conductos Biliares/etiología , Enfermedades de los Conductos Biliares/metabolismo , Caspasa 8/metabolismo , Quistes/etiología , Quistes/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Animales , Apoptosis , Biopsia , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Inmunohistoquímica , Inmunofenotipificación , Hepatopatías/etiología , Hepatopatías/metabolismo , Ratones , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Necroptosis
2.
Mol Neurobiol ; 55(5): 4437-4452, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-28664455

RESUMEN

The activation of c-Jun-N-terminal kinases (JNK) pathway has been largely associated with the pathogenesis and the neuronal death that occur in neurodegenerative diseases. Altogether, this justifies why JNKs have become a focus of screens for new therapeutic strategies. The aim of the present study was to identify the role of the different JNK isoforms (JNK1, JNK2, and JNK3) in apoptosis and inflammation after induction of brain damage. To address this aim, we induced excitotoxicity in wild-type and JNK knockout mice (jnk1 -/- , jnk2 -/- , and jnk3 -/- ) via an intraperitoneal injection of kainic acid, an agonist of glutamic-kainate-receptors, that induce status epilepticus.Each group of animals was divided into two treatments: a single intraperitoneal dose of saline solution, used as a control, and a single intraperitoneal dose (30 mg/kg) of kainic acid. Our results reported a significant decrease in neuronal degeneration in the hippocampus of jnk1 -/- and jnk3 -/- mice after kainic acid treatment, together with reduced or unaltered expression of several apoptotic genes compared to WT treated mice. In addition, both jnk1 -/- and jnk3 -/- mice exhibited a reduction in glial reactivity, as shown by the lower expression of inflammatory genes and a reduction of JNK phosphorylation. In addition, in jnk3 -/- mice, the c-Jun phosphorylation was also diminished.Collectively, these findings provide compelling evidence that the absence of JNK1 or JNK3 isoforms confers neuroprotection against neuronal damage induced by KA and evidence, for the first time, the implication of JNK1 in excitotoxicity. Accordingly, JNK1 and/or JNK3 are promising targets for the prevention of cell death and inflammation during epileptogenesis.


Asunto(s)
Epilepsia del Lóbulo Temporal/enzimología , Proteína Quinasa 10 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Fármacos Neuroprotectores/metabolismo , Animales , Apoptosis/genética , Activación Enzimática , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/patología , Hipocampo/patología , Inflamación/patología , Isoenzimas/metabolismo , Ácido Kaínico , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Fosforilación
3.
Respir Res ; 17(1): 101, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27506464

RESUMEN

BACKGROUND: Pseudomonas aeruginosa (PA) is the single-most common pathogen of ventilator-associated pneumonia (VAP). Large quantities of PA in the trachea of ventilated patients are associated with an increased risk of death. However, the role of PA colonization in PA VAP-induced lung injury remains elusive. This study examined the effect and mechanism of PA colonization in VAP-induced lung injury. METHODS: C57BL/6 wild-type (WT) and c-Jun N-terminal kinase knockout (JNK1(-/-)) mice received mechanical ventilation for 3 h at 2 days after receiving nasal instillation of PA (1 × 10(6) colony forming unit) or normal saline. RESULTS: Intranasal instillation of PA or mechanical ventilation induced the expression of interleukin-6 (IL-6) in the lungs. Phospho-JNK protein expression in the lungs was significantly increased in mice receiving mechanical ventilation after PA instillation as compared with those receiving ventilation alone. Mechanical ventilation after PA instillation significantly increased the expression of tumor necrosis factor-α (TNF-α), IL-1ß, and macrophage inflammatory protein-2 (MIP-2) proteins; neutrophil sequestration; and TNF-α, IL-1ß, and IL-6 levels in the lungs of WT mice, but not in JNK1(-/-) mice. CONCLUSION: PA colonization plays an important role in PA VAP-induced lung injury through the induction of JNK1-mediated inflammation. PA-induced VAP causes lung injury through JNK signaling pathway in the lungs. JNK inhibition in ICU patients with higher percentages of PA colonization may reduce VAP-induced lung injury and mortality.


Asunto(s)
Pulmón/microbiología , Neumonía Asociada al Ventilador/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmón/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Infiltración Neutrófila , Fosforilación , Neumonía Asociada al Ventilador/genética , Neumonía Asociada al Ventilador/metabolismo , Neumonía Asociada al Ventilador/prevención & control , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/prevención & control , Factor de Necrosis Tumoral alfa/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 36(6): 1122-31, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27102962

RESUMEN

OBJECTIVE: The c-Jun NH2-terminal kinases (JNK) are regulated by a wide variety of cellular stresses and have been implicated in apoptotic signaling. Macrophages express 2 JNK isoforms, JNK1 and JNK2, which may have different effects on cell survival and atherosclerosis. APPROACH AND RESULTS: To dissect the effect of macrophage JNK1 and JNK2 on early atherosclerosis, Ldlr(-/-) mice were reconstituted with wild-type, Jnk1(-/-), and Jnk2(-/-) hematopoietic cells and fed a high cholesterol diet. Jnk1(-/-)→Ldlr(-/-) mice have larger atherosclerotic lesions with more macrophages and fewer apoptotic cells than mice transplanted with wild-type or Jnk2(-/-) cells. Moreover, genetic ablation of JNK to a single allele (Jnk1(+/-)/Jnk2(-/-) or Jnk1(-/-)/Jnk2(+/-)) in marrow of Ldlr(-/-) recipients further increased atherosclerosis compared with Jnk1(-/-)→Ldlr(-/-) and wild-type→Ldlr(-/-) mice. In mouse macrophages, anisomycin-mediated JNK signaling antagonized Akt activity, and loss of Jnk1 gene obliterated this effect. Similarly, pharmacological inhibition of JNK1, but not JNK2, markedly reduced the antagonizing effect of JNK on Akt activity. Prolonged JNK signaling in the setting of endoplasmic reticulum stress gradually extinguished Akt and Bad activity in wild-type cells with markedly less effects in Jnk1(-/-) macrophages, which were also more resistant to apoptosis. Consequently, anisomycin increased and JNK1 inhibitors suppressed endoplasmic reticulum stress-mediated apoptosis in macrophages. We also found that genetic and pharmacological inhibition of phosphatase and tensin homolog abolished the JNK-mediated effects on Akt activity, indicating that phosphatase and tensin homolog mediates crosstalk between these pathways. CONCLUSIONS: Loss of Jnk1, but not Jnk2, in macrophages protects them from apoptosis, increasing cell survival, and this accelerates early atherosclerosis.


Asunto(s)
Aorta/enzimología , Enfermedades de la Aorta/enzimología , Apoptosis , Aterosclerosis/enzimología , Células de la Médula Ósea/enzimología , Macrófagos/enzimología , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Receptores de LDL/deficiencia , Animales , Aorta/efectos de los fármacos , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apoptosis/efectos de los fármacos , Aterosclerosis/genética , Aterosclerosis/patología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Supervivencia Celular , Células Cultivadas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Predisposición Genética a la Enfermedad , Hipercolesterolemia/enzimología , Hipercolesterolemia/genética , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Placa Aterosclerótica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de LDL/genética , Transducción de Señal , Proteína Letal Asociada a bcl/metabolismo
5.
Mol Neurobiol ; 53(9): 6183-6193, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26558630

RESUMEN

c-Jun N-terminal kinases (JNKs), which belong to a mitogen-activated protein kinase (MAPK) family, are involved in the regulation of several physiological functions in mammals and act as mediators of apoptosis, obesity, and memory storage in the brain, including the processes of neuronal de- and regeneration. JNK subfamily is encoded by three separate but related genes: jnk1, jnk2, and jnk3, giving rise to at least ten distinct splice variants of the JNK proteins. JNK3 is thought to be a major contributor to neurodegeneration in mammalian brain. The role of JNK1 in the pathological processes affecting cognitive function, especially in diseases such as Alzheimer's disease (AD), is less clear. In order to evaluate the effects of JNK1 deficiency in an experimental model of familial Alzheimer's disease, double transgenic APPswe/PS1dE9 mice were crossed with the JNK1 heterozygous deficient animals (jnk1+/-). As expected, a ∼50 % reduction in JNK1 protein levels was observed in the hippocampi of 9-month-old APPswe/PS1dE9/jnk1+/- mice, compared with the APPswe/PS1dE9 group. JNK1 deficiency resulted in reduced BACE1 expression, suggesting alterations in amyloidogenic pathway. However, no significant inter-group differences in the total number of ß-amyloid plaques were observed in the hippocampal region. In addition, protein levels of PPAR gamma coactivator-1α (PGC-1α), a molecule involved in mitochondrial biogenesis and energy homeostasis, were decreased in 9-month-old APPswe/PS1dE9 mice but not in APPswe/PS1dE9/jnk1+/- animals. Furthermore, JNK1 deficiency did not have an effect on pro-inflammatory marker expression in the hippocampus. Heterozygous deficiency of JNK1 results in the decrease of BACE1 protein levels, which is not accompanied by the reduction in the total number of ß-amyloid plaques in the hippocampi of APPswe/PS1dE9 mice. Moreover, PGC-1α expression is restored in APPswe/PS1dE9/jnk1+/- animals, which indicates a possible role of JNK1 in brain mitochondrial regulation. Nevertheless, our results suggest that partial inhibition of JNK1 is not sufficient to prevent the neuropathological processes in this model. It may be necessary to inhibit both the JNK1 and JNK3 simultaneously, especially as previous studies suggest that JNK3 contributes to AD neuropathology.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Hipocampo/enzimología , Hipocampo/patología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Heterocigoto , Humanos , Inflamación/genética , Inflamación/patología , Ratones Transgénicos , Mitocondrias/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Presenilina-1/metabolismo , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
6.
Gastroenterology ; 150(4): 968-81, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26708719

RESUMEN

BACKGROUND & AIMS: c-Jun N-terminal kinase (JNK) 1 and JNK2 are expressed in hepatocytes and have overlapping and distinct functions. JNK proteins are activated via phosphorylation in response to acetaminophen- or carbon tetrachloride (CCl4)-induced liver damage; the level of activation correlates with the degree of injury. SP600125, a JNK inhibitor, has been reported to block acetaminophen-induced liver injury. We investigated the role of JNK in drug-induced liver injury (DILI) in liver tissue from patients and in mice with genetic deletion of JNK in hepatocytes. METHODS: We studied liver sections from patients with DILI (due to acetaminophen, phenprocoumon, nonsteroidal anti-inflammatory drugs, or autoimmune hepatitis) or patients without acute liver failure (controls) collected from a DILI Biobank in Germany. Levels of total and activated (phosphorylated) JNK were measured by immunohistochemistry and Western blotting. Mice with hepatocyte-specific deletion of Jnk1 (Jnk1(Δhepa)) or combination of Jnk1 and Jnk2 (Jnk(Δhepa)), as well as Jnk1-floxed C57BL/6 (control) mice, were given injections of CCl4 (to induce fibrosis) or acetaminophen (to induce toxic liver injury). We performed gene expression microarray and phosphoproteomic analyses to determine mechanisms of JNK activity in hepatocytes. RESULTS: Liver samples from DILI patients contained more activated JNK, predominantly in nuclei of hepatocytes and in immune cells, than healthy tissue. Administration of acetaminophen to Jnk(Δhepa) mice produced a greater level of liver injury than that observed in Jnk1(Δhepa) or control mice, based on levels of serum markers and microscopic and histologic analysis of liver tissues. Administration of CCl4 also induced stronger hepatic injury in Jnk(Δhepa) mice, based on increased inflammation, cell proliferation, and fibrosis progression, compared with Jnk1(Δhepa) or control mice. Hepatocytes from Jnk(Δhepa) mice given acetaminophen had an increased oxidative stress response, leading to decreased activation of adenosine monophosphate-activated protein kinase, total protein adenosine monophosphate-activated protein kinase levels, and pJunD and subsequent necrosis. Administration of SP600125 before or with acetaminophen protected Jnk(Δhepa) and control mice from liver injury. CONCLUSIONS: In hepatocytes, JNK1 and JNK2 appear to have combined effects in protecting mice from CCl4- and acetaminophen-induced liver injury. It is important to study the tissue-specific functions of both proteins, rather than just JNK1, in the onset of toxic liver injury. JNK inhibition with SP600125 shows off-target effects.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Hepatocitos/enzimología , Fallo Hepático Agudo/prevención & control , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Acetaminofén , Animales , Tetracloruro de Carbono , Estudios de Casos y Controles , Muerte Celular , Proliferación Celular , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Perfilación de la Expresión Génica , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/enzimología , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Estrés Oxidativo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Factores de Tiempo , Adulto Joven
7.
FEBS Lett ; 589(23): 3686-90, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26519559

RESUMEN

TAp63γ is a homologue of tumor suppressor p53 and functions as a transcriptional factor playing key roles in cell cycle and cell apoptosis. In the present work, we find that JNK1 can physically interact with N-terminal transactivation domain (TAD) of TAp63. Overexpression of JNK1 inhibits TAp63γ-mediated transcription, while knockdown or inhibition of endogenous JNK1 increases transactivity of TAp63γ. Further study reveals that Ser12 site in TAD is critical for JNK1-mediated inhibition of TAp63γ. This JNK1-mediated inhibition can impair pro-apoptotic activity of TAp63γ. Together, we report a novel regulation of TAp63γ transactivity and pro-apoptotic activity mediated by JNK1.


Asunto(s)
Apoptosis , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Estructura Terciaria de Proteína , Factores de Transcripción/química , Activación Transcripcional , Proteínas Supresoras de Tumor/química
8.
PLoS One ; 9(10): e109750, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25285524

RESUMEN

Stathmin 1 performs a critical function in cell proliferation by regulating microtubule polymerization. This proliferative function is thought to explain the frequent overexpression of stathmin in human cancer and its correlation with a bad prognosis. Whether stathmin also functions in cell death pathways is unclear. Stathmin regulates microtubules in part by binding free tubulin, a process inhibited by stathmin phosphorylation from kinases including c-Jun N-terminal kinase (JNK). The involvement of JNK activation both in stathmin phosphorylation, and in hepatocellular resistance to oxidative stress, led to an examination of the role of stathmin/JNK crosstalk in oxidant-induced hepatocyte death. Oxidative stress from menadione-generated superoxide induced JNK-dependent stathmin phosphorylation at Ser-16, Ser-25 and Ser-38 in hepatocytes. A stathmin knockdown sensitized hepatocytes to both apoptotic and necrotic cell death from menadione without altering levels of oxidant generation. The absence of stathmin during oxidative stress led to JNK overactivation that was the mechanism of cell death as a concomitant knockdown of JNK1 or JNK2 blocked death. Hepatocyte death from JNK overactivation was mediated by the effects of JNK on mitochondria. Mitochondrial outer membrane permeabilization occurred in stathmin knockdown cells at low concentrations of menadione that triggered apoptosis, whereas mitochondrial ß-oxidation and ATP homeostasis were compromised at higher, necrotic menadione concentrations. Stathmin therefore mediates hepatocyte resistance to death from oxidative stress by down regulating JNK and maintaining mitochondrial integrity. These findings demonstrate a new mechanism by which stathmin promotes cell survival and potentially tumor growth.


Asunto(s)
Regulación hacia Abajo , Hepatocitos/citología , Hepatocitos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Estrés Oxidativo , Estatmina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HEK293 , Hepatocitos/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Necrosis/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Estatmina/deficiencia , Estatmina/genética , Vitamina K 3/farmacología
9.
J Hepatol ; 61(6): 1376-84, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25064435

RESUMEN

BACKGROUND & AIMS: Free cholesterol (FC) accumulates in non-alcoholic steatohepatitis (NASH) but not in simple steatosis. We sought to establish how FC causes hepatocyte injury. METHODS: In NASH-affected livers from diabetic mice, subcellular FC distribution (filipin fluorescence) was established by subcellular marker co-localization. We loaded murine hepatocytes with FC by incubation with low-density lipoprotein (LDL) and studied the effects of FC on JNK1 activation, mitochondrial injury and cell death and on the amplifying roles of the high-mobility-group-box 1 (HMGB1) protein and the Toll-like receptor 4 (TLR4). RESULTS: In NASH, FC localized to hepatocyte plasma membrane, mitochondria and ER. This was reproduced in FC-loaded hepatocytes. At 40 µM LDL, hepatocyte FC increased to cause LDH leakage, apoptosis and necrosis associated with JNK1 activation (c-Jun phosphorylation), mitochondrial membrane pore transition, cytochrome c release, oxidative stress (GSSG:GSH ratio) and ATP depletion. Mitochondrial swelling and crystae disarray were evident by electron microscopy. Jnk1(-/-) and Tlr4(-/-) hepatocytes were refractory to FC lipotoxicity; JNK inhibitors (1-2 µM CC-401, CC-930) blocked apoptosis and necrosis. Cyclosporine A and caspase-3 inhibitors protected FC-loaded hepatocytes, confirming mitochondrial cell death pathways; in contrast, 4-phenylbutyric acid, which improves ER folding capacity did not protect FC-loaded hepatocytes. HMGB1 was released into the culture medium of FC-loaded wild type (WT) but not Jnk1(-/-) or Tlr4(-/-) hepatocytes, while anti-HMGB1 anti-serum prevented JNK activation and FC lipotoxicity in WT hepatocytes. CONCLUSIONS: These novel findings show that mitochondrial FC deposition causes hepatocyte apoptosis and necrosis by activating JNK1; inhibition of which could be a novel therapeutic approach in NASH. Further, there is a tight link between JNK1-dependent HMGB1 secretion from lipotoxic hepatocytes and a paracrine cytolytic effect on neighbouring cholesterol-loaded hepatocytes operating via TLR4.


Asunto(s)
Colesterol/metabolismo , Proteína HMGB1/metabolismo , Hepatocitos/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Receptor Toll-Like 4/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/fisiología , Células Cultivadas , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Femenino , Hepatocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Estrés Oxidativo/fisiología , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
10.
Atherosclerosis ; 235(2): 613-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24956536

RESUMEN

OBJECTIVE: The c-Jun N-terminal kinase (JNK) family regulates fundamental physiological processes including apoptosis and metabolism. Although JNK2 is known to promote foam cell formation during atherosclerosis, the potential role of JNK1 is uncertain. We examined the potential influence of JNK1 and its negative regulator, MAP kinase phosphatase-1 (MKP-1), on endothelial cell (EC) injury and early lesion formation using hypercholesterolemic LDLR(-/-) mice. METHODS AND RESULTS: To assess the function of JNK1 in early atherogenesis, we measured EC apoptosis and lesion formation in LDLR(-/-) or LDLR(-/-)/JNK1(-/-) mice exposed to a high fat diet for 6 weeks. En face staining using antibodies that recognise active, cleaved caspase-3 (apoptosis) or using Sudan IV (lipid deposition) revealed that genetic deletion of JNK1 reduced EC apoptosis and lesion formation in hypercholesterolemic mice. By contrast, although EC apoptosis was enhanced in LDLR(-/-)/MKP-1(-/-) mice compared to LDLR(-/-) mice, lesion formation was unaltered. CONCLUSION: We conclude that JNK1 is required for EC apoptosis and lipid deposition during early atherogenesis. Thus pharmacological inhibitors of JNK may reduce atherosclerosis by preventing EC injury as well as by influencing foam cell formation.


Asunto(s)
Células Endoteliales/patología , Hipercolesterolemia/fisiopatología , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Dieta Alta en Grasa , Fosfatasa 1 de Especificidad Dual/deficiencia , Células Endoteliales/metabolismo , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/metabolismo
11.
Gut ; 63(7): 1159-72, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24037431

RESUMEN

OBJECTIVE: The c-Jun N-terminal kinase-1 (Jnk1) gene has been shown to be involved in liver fibrosis. Here, we aimed to investigate the molecular mechanism and define the cell type involved in mediating the Jnk1-dependent effect on liver fibrogenesis. DESIGN: Jnk1(f/f) wildtype (WT), Jnk1(-/-) and Jnk1(Δhepa) (hepatocyte-specific deletion of Jnk1) mice were subjected to (i) bile duct ligation (BDL) and (ii) CCl4-induced liver fibrosis. Additionally, we performed bone marrow transplantations (BMT), isolated primary hepatic stellate cells (HSCs), studied their activation in vitro and investigated human diseased liver samples. RESULTS: Phosphorylated Jnk was expressed in myofibroblasts, epithelial and inflammatory cells during the progression of fibrogenesis in humans and mice. In mice, liver transaminases, alkaline phosphatase, bilirubin and liver histology revealed reduced injury in Jnk1(-/-) compared with WT and Jnk1(Δhepa) mice correlating with lower hepatocyte cell death and proliferation. Consequently, parameters of liver fibrosis such as Sirius red staining and collagen IA1 and α-smooth muscle actin expression were downregulated in Jnk1(-/-) compared with WT and Jnk1(Δhepa) livers, 4 weeks after CCl4 or BDL. BMT experiments excluded bone marrow-derived cells from having a major impact on the Jnk1-dependent effect on fibrogenesis, while primary HSCs from Jnk1(-/-) livers showed reduced transdifferentiation and extracellular matrix production. Moreover, Jnk1 ablation caused a reduced lifespan and poor differentiation of HSCs into matrix-producing myofibroblasts. CONCLUSIONS: Jnk1 in HSCs, but not in hepatocytes, significantly contribute to liver fibrosis development, identifying Jnk1 in HSCs as a profibrotic kinase and a promising cell-directed target for liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/enzimología , Cirrosis Hepática/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Trasplante de Médula Ósea , Transdiferenciación Celular , Enfermedad Crónica , Regulación hacia Abajo , Hepatocitos/enzimología , Humanos , Cirrosis Hepática/etiología , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Regulación hacia Arriba
12.
Proc Natl Acad Sci U S A ; 110(6): 2377-82, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341606

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of registered blindness among the elderly and affects over 30 million people worldwide. It is well established that oxidative stress, inflammation, and apoptosis play critical roles in pathogenesis of AMD. In advanced wet AMD, although, most of the severe vision loss is due to bleeding and exudation of choroidal neovascularization (CNV), and it is well known that vascular endothelial growth factor (VEGF) plays a pivotal role in the growth of the abnormal blood vessels. VEGF suppression therapy improves visual acuity in AMD patients. However, there are unresolved issues, including safety and cost. Here we show that mice lacking c-Jun N-terminal kinase 1 (JNK1) exhibit decreased inflammation, reduced CNV, lower levels of choroidal VEGF, and impaired choroidal macrophage recruitment in a murine model of wet AMD (laser-induced CNV). Interestingly, we also detected a substantial reduction in choroidal apoptosis of JNK1-deficient mice. Intravitreal injection of a pan-caspase inhibitor reduced neovascularization in the laser-induced CNV model, suggesting that apoptosis plays a role in laser-induced pathological angiogenesis. Intravitreal injection of a specific JNK inhibitor decreased choroidal VEGF expression and reduced pathological CNV. These results suggest that JNK1 plays a key role in linking oxidative stress, inflammation, macrophage recruitment apoptosis, and VEGF production in wet AMD and pharmacological JNK inhibition offers a unique and alternative avenue for prevention and treatment of AMD.


Asunto(s)
Neovascularización Coroidal/prevención & control , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Inhibidores de Cisteína Proteinasa/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/patología , Degeneración Macular/patología , Degeneración Macular/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Estrés Oxidativo , Factor A de Crecimiento Endotelial Vascular/biosíntesis
13.
Science ; 339(6116): 218-22, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23223452

RESUMEN

The cJun NH(2)-terminal kinase (JNK) signaling pathway contributes to inflammation and plays a key role in the metabolic response to obesity, including insulin resistance. Macrophages are implicated in this process. To test the role of JNK, we established mice with selective JNK deficiency in macrophages. We report that feeding a high-fat diet to control and JNK-deficient mice caused similar obesity, but only mice with JNK-deficient macrophages remained insulin-sensitive. The protection of mice with macrophage-specific JNK deficiency against insulin resistance was associated with reduced tissue infiltration by macrophages. Immunophenotyping demonstrated that JNK was required for pro-inflammatory macrophage polarization. These studies demonstrate that JNK in macrophages is required for the establishment of obesity-induced insulin resistance and inflammation.


Asunto(s)
Inflamación/fisiopatología , Resistencia a la Insulina , Macrófagos/enzimología , Macrófagos/inmunología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Obesidad/fisiopatología , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa , Técnica de Clampeo de la Glucosa , Inmunofenotipificación , Inflamación/inmunología , Islotes Pancreáticos/patología , Sistema de Señalización de MAP Quinasas , Activación de Macrófagos , Macrófagos/fisiología , Ratones , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Obesidad/inmunología
14.
Free Radic Biol Med ; 53(5): 1170-80, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22749809

RESUMEN

Binge alcohol drinking induces hepatic steatosis. Recent studies showed that chronic ethanol-induced fatty liver was, at least in part, CYP2E1 dependent. The mechanism of acute alcohol-induced steatosis and whether CYP2E1 plays any role are still unclear. Increasing oxidative stress by alcohol can activate the JNK MAP kinase signaling pathway, suggesting that JNK might be a target for prevention of alcohol-induced steatosis. We used CYP2E1 knockout (KO) mice, a JNK inhibitor, and JNK1 or JNK2 knockout mice to test the role of CYP2E1, JNK, and the individual role of JNK1 and JNK2 in acute alcohol-induced steatosis. In wild-type (WT) mice, acute alcohol activates CYP2E1 and increases oxidative stress, which reciprocally increases activation of the JNK signaling pathway. Acute alcohol-induced fatty liver and oxidative stress were blunted in CYP2E1 KO mice and by the JNK inhibitor in WT mice. The antioxidant N-acetylcysteine decreased the acute alcohol-induced oxidative stress, the activation of JNK, and the steatosis but not the activation of CYP2E1. Acute alcohol decreased autophagy and increased expression of SREBP, effects blocked by the JNK inhibitor. Acute alcohol-induced fatty liver was the same in JNK1 and JNK2 KO mice as in WT mice; thus either JNK1 or JNK2 per se is sufficient for induction of steatosis by acute alcohol. The results show that acute alcohol elevation of CYP2E1, oxidative stress, and activation of JNK interact to lower autophagy and increase lipogenic SREBP resulting in fatty liver.


Asunto(s)
Autofagia , Citocromo P-450 CYP2E1/metabolismo , Hígado Graso Alcohólico/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Estrés Oxidativo , Animales , Citocromo P-450 CYP2E1/deficiencia , Etanol/administración & dosificación , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/deficiencia
15.
J Clin Invest ; 122(4): 1529-40, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22378044

RESUMEN

Integrin signaling critically contributes to the progression, growth, and therapy resistance of malignant tumors. Here, we show that targeting of ß1 integrins with inhibitory antibodies enhances the sensitivity to ionizing radiation and delays the growth of human head and neck squamous cell carcinoma cell lines in 3D cell culture and in xenografted mice. Mechanistically, dephosphorylation of focal adhesion kinase (FAK) upon inhibition of ß1 integrin resulted in dissociation of a FAK/cortactin protein complex. This, in turn, downregulated JNK signaling and induced cell rounding, leading to radiosensitization. Thus, these findings suggest that robust and selective pharmacological targeting of ß1 integrins may provide therapeutic benefit to overcome tumor cell resistance to radiotherapy.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Cortactina/fisiología , Quinasa 1 de Adhesión Focal/fisiología , Neoplasias de Cabeza y Cuello/radioterapia , Integrina beta1/fisiología , Proteínas de Neoplasias/fisiología , Tolerancia a Radiación/fisiología , Secuencias de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Cortactina/química , Femenino , Quinasa 1 de Adhesión Focal/química , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Integrina beta1/inmunología , Masculino , Ratones , Ratones Desnudos , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/fisiología , Complejos Multiproteicos , Proteínas de Neoplasias/química , Trasplante de Neoplasias , Fosforilación , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Transducción de Señal , Células Tumorales Cultivadas/efectos de la radiación
16.
Cancer Res ; 72(2): 472-81, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22127926

RESUMEN

cJun NH(2)-terminal kinase (JNK) signaling has been implicated in the developmental morphogenesis of epithelial organs. In this study, we employed a compound deletion of the murine Jnk1 and Jnk2 genes in the mammary gland to evaluate the requirement for these ubiquitously expressed genes in breast development and tumorigenesis. JNK1/2 was not required for breast epithelial cell proliferation or motility. However, JNK1/2 deficiency caused increased branching morphogenesis and defects in the clearance of lumenal epithelial cells. In the setting of breast cancer development, JNK1/2 deficiency significantly increased tumor formation. Together, these findings established that JNK signaling is required for normal mammary gland development and that it has a suppressive role in mammary tumorigenesis.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Animales , Procesos de Crecimiento Celular/fisiología , Movimiento Celular/fisiología , Femenino , Expresión Génica , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética
17.
Am J Pathol ; 179(4): 1884-93, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21839715

RESUMEN

The roles of the c-Jun N-terminal kinases (JNKs) in inflammatory arthritis have been investigated; however, the roles of each isotype (ie, JNK1 and JNK2) in rheumatoid arthritis and conclusions about whether inhibition of one or both is necessary for amelioration of disease are unclear. By using JNK1- or JNK2-deficient mice in the collagen-induced arthritis and the KRN T-cell receptor transgenic mouse on C57BL/6 nonobese diabetic (K/BxN) serum transfer arthritis models, we demonstrate that JNK1 deficiency results in protection from arthritis, as judged by clinical score and histological evaluation in both models of inflammatory arthritis. In contrast, abrogation of JNK2 exacerbates disease. In collagen-induced arthritis, the distinct roles of the JNK isotypes can, at least in part, be explained by altered regulation of CD86 expression in JNK1- or JNK2-deficient macrophages in response to microbial products, thereby affecting T-cell-mediated immunity. The protection from K/BxN serum-induced arthritis in Jnk1(-/-) mice can also be explained by inept macrophage function because adoptive transfer of wild-type macrophages to Jnk1(-/-) mice restored disease susceptibility. Thus, our results provide a possible explanation for the modest therapeutic effects of broad JNK inhibitors and suggest that future therapies should selectively target the JNK1 isoform.


Asunto(s)
Artritis Experimental/enzimología , Artritis Experimental/patología , Inflamación/enzimología , Inflamación/patología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Animales , Artritis Experimental/complicaciones , Artritis Experimental/inmunología , Antígeno B7-2/metabolismo , Colágeno Tipo II/inmunología , Modelos Animales de Enfermedad , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Inflamación/complicaciones , Articulaciones/enzimología , Articulaciones/inmunología , Articulaciones/patología , Macrófagos/enzimología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Suero , Regulación hacia Arriba
18.
Neurosci Res ; 71(3): 266-77, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21824499

RESUMEN

c-Jun N-terminal kinase (JNK) mediates neuronal death in response to stress and injury in the CNS and peripheral nervous system. Here, we show that JNK also regulates retrograde axonal degeneration (axonal dieback) after spinal cord injury (SCI) in mice. Activated phospho-JNK was highly expressed in damaged corticospinal tract (CST) axons after thoracic SCI by hemisection. Local administration of SP600125, a JNK inhibitor, prevented accumulation of amyloid-ß precursor protein and retraction of the severed CST axons as well as preserved the axonal arbors rostral to the injury site. The treatment with SP600125 also improved functional recovery of the hindlimbs, assessed by Basso mouse scale open-field scores and the grid-walking test. In Jnk1(-/-) and Jnk3(-/-) mice, we observed prevention of axonal degeneration and enhancement of motor recovery after SCI. These results indicate that both JNK1 and JNK3 induce axonal degeneration and limit motor recovery after SCI. Thus, a JNK inhibitor may be a suitable therapeutic agent for SCI.


Asunto(s)
Proteína Quinasa 10 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Recuperación de la Función , Traumatismos de la Médula Espinal/enzimología , Animales , Antracenos/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 10 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 10 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Parálisis/enzimología , Parálisis/genética , Parálisis/fisiopatología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/genética , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Degeneración Walleriana/enzimología , Degeneración Walleriana/patología , Degeneración Walleriana/fisiopatología
19.
Hear Res ; 278(1-2): 77-85, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21554942

RESUMEN

Jun N-terminal kinase (JNK) is a multifunctional protein kinase crucial for neuronal apoptosis as well as neurite growth. We have previously shown that JNK activity is correlated with spiral ganglion neuron (SGN) apoptosis following hair cell loss in rats (Alam et al., 2007) implying that JNK inhibition may have therapeutic potential to protect SGNs in deaf individuals. Here we investigated the role of JNK in neurite outgrowth from cultured neonatal rat and mouse SGNs. We show that JNK is required for initial growth of neurites and for continued extension of already established neurites. The effect of JNK inhibition on neurite growth is rapid and is also rapidly reversible after washout of the inhibitor. Using phosphoJNK immunoreactivity as an indicator, we show that JNK is activated in growth cones within 30 min after transfer to medium lacking neurotrophic stimuli (5 K medium) but activation in the nucleus and soma requires hours. By transfecting epitope-tagged JNK1, JNK2, or JNK3 isoforms into SGNs, we found that all are present in the nucleus and cytoplasm and that there is no preferential redistribution to the nucleus after transfer to 5 K medium. Cotransfection of dominant-negative (dn) JNK1 and JNK2 into SGNs reduced neurite growth, although transfection of dnJNK1 or dnJNK2 alone had no significant effect. SGNs cultured from JNK3(-/-) mice showed reduced neurite growth that was further reduced by transfection of dnJNK1 and dnJNK2. This indicates that all three JNK isoforms promote SGN neurite growth although there may be functional redundancy between JNK1 and JNK2.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Neuritas/enzimología , Neuritas/ultraestructura , Ganglio Espiral de la Cóclea/enzimología , Ganglio Espiral de la Cóclea/inervación , Animales , Células Cultivadas , Activación Enzimática , Cinética , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 10 Activada por Mitógenos/deficiencia , Proteína Quinasa 10 Activada por Mitógenos/genética , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Neuronas/enzimología , Neuronas/ultraestructura , Fosforilación , Ratas , Ganglio Espiral de la Cóclea/ultraestructura , Fracciones Subcelulares/enzimología , Transfección
20.
J Biol Chem ; 286(25): 22227-34, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21540183

RESUMEN

SIRT1 is involved in the pathogenesis of obesity, diabetes, and aging. However, it is not clear how SIRT1 activity is regulated by intracellular kinases in cells. In this study, we investigated SIRT1 phosphorylation and protein degradation in response to JNK1 activation in obese mice. Mouse SIRT1 is phosphorylated by JNK1 at Ser-46 (Ser-47 in human SIRT1), which is one of the four potential residues targeted by JNK1. The phosphorylation induces a brief activation of SIRT1 function and degradation of SIRT1 thereafter by the proteasome. Ubiquitination occurs in SIRT1 protein after the phosphorylation. Mutation of Ser-46 to alanine prevents the phosphorylation, ubiquitination, and degradation. In vivo, SIRT1 undergoes an extensive degradation in hepatocytes in obesity as a consequence of persistent activation of JNK1. The degradation leads to inhibition of SIRT1 function, which contributes to development of hepatic steatosis. The degradation disappears in obesity when JNK1 is inactivated in mice. JNK2 exhibits an opposite activity in the regulation of SIRT1 degradation. The JNK1-SIRT1 pathway provides a new molecular mechanism for the pathogenesis of hepatic steatosis in obesity.


Asunto(s)
Hígado Graso/complicaciones , Hígado Graso/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Obesidad/complicaciones , Sirtuina 1/metabolismo , Animales , Línea Celular , Grasas de la Dieta/efectos adversos , Activación Enzimática , Hígado Graso/enzimología , Técnicas de Inactivación de Genes , Histona Desacetilasas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Fosforilación , Ratas , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...