Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
2.
Int J Mol Sci ; 23(5)2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-35269949

RESUMEN

ß-thalassemia is a hematologic disease that may be associated with significant morbidity and mortality. Increased expression of HBG1/2 can ameliorate the severity of ß-thalassemia. Compared to the unaffected population, some ß-thalassemia patients display elevated HBG1/2 expression levels in their red blood cells. However, the magnitude of up-regulation does not reach the threshold of self-healing, and thus, the molecular mechanism underlying HBG1/2 expression in the context of HBB-deficiency requires further elucidation. Here, we performed a multi-omics study examining chromatin accessibility, transcriptome, proteome, and phosphorylation patterns in the HBB homozygous knockout of the HUDEP2 cell line (HBB-KO). We found that up-regulation of HBG1/2 in HBB-KO cells was not induced by the H3K4me3-mediated genetic compensation response. Deletion of HBB in human erythroid progenitor cells resulted in increased ROS levels and production of oxidative stress, which led to an increased rate of apoptosis. Furthermore, in response to oxidative stress, slower cell cycle progression and proliferation were observed. In addition, stress erythropoiesis was initiated leading to increased intracellular HBG1/2 expression. This molecular model was also validated in the single-cell transcriptome of hematopoietic stem cells from ß-hemoglobinopathy patients. These findings further the understanding of HBG1/2 gene regulatory networks and provide novel clinical insights into ß-thalassemia phenotypic diversity.


Asunto(s)
Talasemia beta , Recuento de Eritrocitos , Eritrocitos/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Humanos
3.
STAR Protoc ; 2(4): 100915, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34755118

RESUMEN

Neuronal loss resulting from progressive neurodegeneration is a major pathological feature of Alzheimer's disease (AD). Here, we present a protocol to detect neurodegeneration, neuronal apoptosis, and neuronal loss in 5XFAD mouse strain, which is a well-established model for interrogating the molecular mechanism of neuronal death in AD. This protocol describes the use of the neurodegenerative marker Fluro-Jade C, cleaved caspase-3 immunofluorescent staining and Nissl staining for the analysis of neurodegeneration and neuronal loss in 5XFAD mice. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021).


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo , Disfunción Cognitiva/patología , Histocitoquímica/métodos , Animales , Apoptosis/fisiología , Encéfalo/citología , Encéfalo/patología , Masculino , Ratones , Ratones Transgénicos , Microscopía
4.
iScience ; 24(9): 102942, 2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34430820

RESUMEN

Alzheimer's disease (AD) is the most common progressive neurodegenerative disease. However, the underlying molecular mechanism is incompletely understood. Here we report that the pro-apoptotic protein BAD as a key regulator for neuronal apoptosis, neuroinflammation and Aß clearance in AD. BAD pro-apoptotic activity is significantly increased in neurons of AD patients and 5XFAD mice. Conversely, genetic disruption of Bad alleles restores spatial learning and memory deficits in 5XFAD mice. Mechanistically, phosphorylation and inactivation of BAD by neurotropic factor-activated Akt is abrogated in neurons under AD condition. Through reactive oxygen species (ROS)-oxidized mitochondrial DNA (mtDNA) axis, BAD also promotes microglial NLRP3 inflammasome activation, thereby skewing microglia toward neuroinflammatory microglia to inhibit microglial phagocytosis of Aß in AD mice. Our results support a model in which BAD contributes to AD pathologies by driving neuronal apoptosis and neuroinflammation but suppressing microglial phagocytosis of Aß, suggesting that BAD is a potential therapeutic target for AD.

5.
Immunity ; 54(6): 1168-1185.e8, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34038747

RESUMEN

Chronic inflammation plays a central role in hepatocellular carcinoma (HCC), but the contribution of hepatocytes to tumor-associated inflammation is not clear. Here, we report that the zinc finger transcription factor Miz1 restricted hepatocyte-driven inflammation to suppress HCC, independently of its transcriptional activity. Miz1 was downregulated in HCC mouse models and a substantial fraction of HCC patients. Hepatocyte-specific Miz1 deletion in mice generated a distinct sub-group of hepatocytes that produced pro-inflammatory cytokines and chemokines, which skewed the polarization of the tumor-infiltrating macrophages toward pro-inflammatory phenotypes to promote HCC. Mechanistically, Miz1 sequestrated the oncoprotein metadherin (MTDH), preventing MTDH from promoting transcription factor nuclear factor κB (NF-κB) activation. A distinct sub-group of pro-inflammatory cytokine-producing hepatocytes was also seen in a subset of HCC patients. In addition, Miz1 expression inversely correated with disease recurrence and poor prognosis in HCC patients. Our findings identify Miz1 as a tumor suppressor that prevents hepatocytes from driving inflammation in HCC.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma Hepatocelular/metabolismo , Hepatocitos/metabolismo , Inflamación/metabolismo , Neoplasias Hepáticas/metabolismo , Activación de Macrófagos/fisiología , Proteínas Inhibidoras de STAT Activados/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Línea Celular , Línea Celular Tumoral , Quimiocinas/metabolismo , Regulación hacia Abajo/fisiología , Femenino , Células HEK293 , Hepatocitos/patología , Humanos , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Dedos de Zinc/fisiología
6.
Elife ; 92020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33270017

RESUMEN

The resistance of synovial sublining macrophages to apoptosis has a crucial role in joint inflammation and destruction in rheumatoid arthritis (RA). However, the underlying mechanism is incompletely understood. Here we report that inactivation of the pro-apoptotic BCL-2 family protein BAD is essential for survival of synovial sublining macrophage in RA. Genetic disruption of Bad leads to more severe joint inflammation and cartilage and bone damage with reduced apoptosis of synovial sublining macrophages in collagen-induced arthritis (CIA) and TNFα transgenic (TNF-Tg) mouse models. Conversely, Bad3SA/3SA mice, in which BAD can no longer be inactivated by phosphorylation, are protected from collagen-induced arthritis. Mechanistically, phosphorylation-mediated inactivation of BAD specifically protects synovial sublining macrophages from apoptosis in highly inflammatory environment of arthritic joints in CIA and TNF-Tg mice, and in patients with RA, thereby contributing to RA pathology. Our findings put forward a model in which inactivation of BAD confers the apoptosis resistance on synovial sublining macrophages, thereby contributing to the development of arthritis, suggesting that BAD may be a potential therapeutic target for RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Macrófagos/fisiología , Osteoartritis/inducido químicamente , Proteína Letal Asociada a bcl/metabolismo , Adulto , Anciano , Animales , Artritis Reumatoide/genética , Trasplante de Médula Ósea , Colágeno/toxicidad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Osteoartritis/metabolismo , Proteína Letal Asociada a bcl/genética
7.
EMBO Rep ; 21(5): e48566, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32239614

RESUMEN

Progenitor cells at the basal layer of skin epidermis play an essential role in maintaining tissue homeostasis and enhancing wound repair in skin. The proliferation, differentiation, and cell death of epidermal progenitor cells have to be delicately regulated, as deregulation of this process can lead to many skin diseases, including skin cancers. However, the underlying molecular mechanisms involved in skin homeostasis remain poorly defined. In this study, with quantitative proteomics approach, we identified an important interaction between KDF1 (keratinocyte differentiation factor 1) and IKKα (IκB kinase α) in differentiating skin keratinocytes. Ablation of either KDF1 or IKKα in mice leads to similar but striking abnormalities in skin development, particularly in skin epidermal differentiation. With biochemical and mouse genetics approach, we further demonstrate that the interaction of IKKα and KDF1 is essential for epidermal differentiation. To probe deeper into the mechanisms, we find that KDF1 associates with a deubiquitinating protease USP7 (ubiquitin-specific peptidase 7), and KDF1 can regulate skin differentiation through deubiquitination and stabilization of IKKα. Taken together, our study unravels an important molecular mechanism underlying epidermal differentiation and skin tissue homeostasis.


Asunto(s)
Diferenciación Celular , Células Epidérmicas/citología , Quinasa I-kappa B , Queratinocitos , Proteínas/metabolismo , Animales , Epidermis , Quinasa I-kappa B/genética , Ratones , Ubiquitinación
8.
Proc Natl Acad Sci U S A ; 115(45): E10682-E10691, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30337485

RESUMEN

Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by low platelet count which can cause fatal hemorrhage. ITP patients with antiplatelet glycoprotein (GP) Ib-IX autoantibodies appear refractory to conventional treatments, and the mechanism remains elusive. Here we show that the platelets undergo apoptosis in ITP patients with anti-GPIbα autoantibodies. Consistent with these findings, the anti-GPIbα monoclonal antibodies AN51 and SZ2 induce platelet apoptosis in vitro. We demonstrate that anti-GPIbα antibody binding activates Akt, which elicits platelet apoptosis through activation of phosphodiesterase (PDE3A) and PDE3A-mediated PKA inhibition. Genetic ablation or chemical inhibition of Akt or blocking of Akt signaling abolishes anti-GPIbα antibody-induced platelet apoptosis. We further demonstrate that the antibody-bound platelets are removed in vivo through an apoptosis-dependent manner. Phosphatidylserine (PS) exposure on apoptotic platelets results in phagocytosis of platelets by macrophages in the liver. Notably, inhibition or genetic ablation of Akt or Akt-regulated apoptotic signaling or blockage of PS exposure protects the platelets from clearance. Therefore, our findings reveal pathogenic mechanisms of ITP with anti-GPIbα autoantibodies and, more importantly, suggest therapeutic strategies for thrombocytopenia caused by autoantibodies or other pathogenic factors.


Asunto(s)
Apoptosis , Plaquetas/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Púrpura Trombocitopénica Idiopática/patología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glicoproteínas/inmunología , Humanos , Hígado/metabolismo , Macrófagos/metabolismo , Fagocitosis , Hidrolasas Diéster Fosfóricas/metabolismo , Púrpura Trombocitopénica Idiopática/enzimología , Transducción de Señal
9.
Cell Res ; 28(7): 701-718, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29795446

RESUMEN

The inflammatory cytokine TNFα plays a crucial role in the pathology of many inflammatory and infectious diseases. However, the mechanism underlying TNFα cytotoxicity in these diseases is incompletely understood. Here we report that the pro-apoptotic BCL-2 family member BAD mediates TNFα cytotoxicity despite concurrent activation of IKK and NF-κB in vitro by inducing apoptosis in cultured cells and in vivo by eliciting tissue damage of multiple organs and contributing to mortality in septic shock. At high doses, TNFα significantly inactivates RhoA through activation of the Src-p190GAP pathway, resulting in massive actin stress fiber destabilization, followed by substantial BAD release from the cytoskeleton to the cytosol. Under this condition, activated IKK fails to phosphorylate all cytosolic BAD, allowing translocation of non-phosphorylated BAD to mitochondria to trigger apoptosis. Polymicrobial infection utilizes the same mechanism as high-dose TNFα to elicit apoptosis-associated tissue damage of multiple organs. Consequently, loss of Bad or elimination of BAD pro-apoptotic activity protects mice from tissue damage of multiple organs and reduces mortality rates. Our results support a model in which BAD mediates TNFα cytotoxicity despite concurrent activation of the IKK-NF-κB pathway in cultured mammalian cells and in septic shock.


Asunto(s)
Apoptosis , Choque Séptico/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteína Letal Asociada a bcl/fisiología , Animales , Células Cultivadas , Coinfección/inmunología , Quinasa I-kappa B/metabolismo , Mutación con Pérdida de Función , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Proteína Letal Asociada a bcl/genética , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
10.
J Clin Invest ; 127(12): 4338-4351, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29083324

RESUMEN

Apoptosis delimits platelet life span in the circulation and leads to storage lesion, which severely limits the shelf life of stored platelets. Moreover, accumulating evidence indicates that platelet apoptosis provoked by various pathological stimuli results in thrombocytopenia in many common diseases. However, little is known about how platelet apoptosis is initiated or regulated. Here, we show that PKA activity is markedly reduced in platelets aged in vitro, stored platelets, and platelets from patients with immune thrombocytopenia (ITP), diabetes, and bacterial infections. Inhibition or genetic ablation of PKA provoked intrinsic programmed platelet apoptosis in vitro and rapid platelet clearance in vivo. PKA inhibition resulted in dephosphorylation of the proapoptotic protein BAD at Ser155, resulting in sequestration of prosurvival protein BCL-XL in mitochondria and subsequent apoptosis. Notably, PKA activation protected platelets from apoptosis induced by storage or pathological stimuli and elevated peripheral platelet levels in normal mice and in a murine model of ITP. Therefore, these findings identify PKA as a homeostatic regulator of platelet apoptosis that determines platelet life span and survival. Furthermore, these results suggest that regulation of PKA activity represents a promising strategy for extending platelet shelf life and has profound implications for the treatment of platelet number-related diseases and disorders.


Asunto(s)
Apoptosis , Plaquetas/enzimología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Animales , Infecciones Bacterianas/enzimología , Infecciones Bacterianas/genética , Infecciones Bacterianas/patología , Plaquetas/patología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Activación Enzimática/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Púrpura Trombocitopénica Idiopática/enzimología , Púrpura Trombocitopénica Idiopática/genética , Púrpura Trombocitopénica Idiopática/patología , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
11.
PLoS Pathog ; 13(7): e1006534, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28753655

RESUMEN

As a major diarrheagenic human pathogen, enterohemorrhagic Escherichia coli (EHEC) produce attaching and effacing (A/E) lesions, characterized by the formation of actin pedestals, on mammalian cells. A bacterial T3SS effector NleL from EHEC O157:H7 was recently shown to be a HECT-like E3 ligase in vitro, but its biological functions and host targets remain elusive. Here, we report that NleL is required to effectively promote EHEC-induced A/E lesions and bacterial infection. Furthermore, human c-Jun NH2-terminal kinases (JNKs) were identified as primary substrates of NleL. NleL-induced JNK ubiquitylation, particularly mono-ubiquitylation at the Lys 68 residue of JNK, impairs JNK's interaction with an upstream kinase MKK7, thus disrupting JNK phosphorylation and activation. This subsequently suppresses the transcriptional activity of activator protein-1 (AP-1), which modulates the formation of the EHEC-induced actin pedestals. Moreover, JNK knockdown or inhibition in host cells complements NleL deficiency in EHEC infection. Thus, we demonstrate that the effector protein NleL enhances the ability of EHEC to infect host cells by targeting host JNK, and elucidate an inhibitory role of ubiquitylation in regulating JNK phosphorylation.


Asunto(s)
Adhesión Bacteriana , Escherichia coli Enterohemorrágica/fisiología , Infecciones por Escherichia coli/enzimología , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Escherichia coli Enterohemorrágica/genética , Infecciones por Escherichia coli/genética , Proteínas de Escherichia coli/genética , Células HeLa , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/química , Proteínas Quinasas JNK Activadas por Mitógenos/genética , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Fosforilación , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
12.
J Gerontol A Biol Sci Med Sci ; 72(11): 1492-1500, 2017 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-28498894

RESUMEN

Death from chronic lung disease is increasing and chronic obstructive pulmonary disease has become the third leading cause of death in the United States in the past decade. Both chronic and acute lung diseases disproportionately affect elderly individuals, making it likely that these diseases will become more frequent and severe as the worldwide population ages. Chronic lung diseases are associated with substantial morbidity, frequently resulting in exercise limiting dyspnea, immobilization, and isolation. Therefore, effective strategies to prevent or treat lung disease are likely to increase healthspan as well as life span. This review summarizes the findings of a joint workshop sponsored by the NIA and NHLBI that brought together investigators focused on aging and lung biology. These investigators encouraged the use of genetic systems and aged animals in the study of lung disease and the development of integrative systems-based platforms that can dynamically incorporate data sets that describe the genomics, transcriptomics, epigenomics, metabolomics, and proteomics of the aging lung in health and disease. Further research was recommended to integrate benchmark biological hallmarks of aging in the lung with the pathobiology of acute and chronic lung diseases with divergent pathologies for which advanced age is the most important risk factor.


Asunto(s)
Envejecimiento/fisiología , Enfermedades Pulmonares/terapia , Humanos , Enfermedades Pulmonares/fisiopatología , Metabolómica/métodos , National Heart, Lung, and Blood Institute (U.S.) , Estados Unidos
13.
Proc Natl Acad Sci U S A ; 112(31): 9644-9, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195787

RESUMEN

The serine-threonine kinase Akt is a key regulator of cell proliferation and survival, glucose metabolism, cell mobility, and tumorigenesis. Activation of Akt by extracellular stimuli such as insulin centers on the interaction of Akt with PIP3 on the plasma membrane, where it is subsequently phosphorylated and activated by upstream protein kinases. However, it is not known how Akt is recruited to the plasma membrane upon stimulation. Here we report that ubiquitin-like protein 4A (Ubl4A) plays a crucial role in insulin-induced Akt plasma membrane translocation. Ubl4A knockout newborn mice have defective Akt-dependent glycogen synthesis and increased neonatal mortality. Loss of Ubl4A results in the impairment of insulin-induced Akt translocation to the plasma membrane and activation. Akt binds actin-filaments and colocalizes with actin-related protein 2 and 3 (Arp2/3) complex in the membrane ruffles and lamellipodia. Ubl4A directly interacts with Arp2/3 to accelerate actin branching and networking, allowing Akt to be in close proximity to the plasma membrane for activation upon insulin stimulation. Our finding reveals a new mechanism by which Akt is recruited to the plasma membrane for activation, thereby providing a missing link in Akt signaling.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Membrana Celular/enzimología , Insulina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ubiquitinas/metabolismo , Actinas/metabolismo , Animales , Animales Recién Nacidos , Membrana Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Embrión de Mamíferos/citología , Activación Enzimática/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Glucógeno/biosíntesis , Proteínas Fluorescentes Verdes/metabolismo , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Ubiquitinas/deficiencia
14.
Mol Cell Biol ; 33(11): 2252-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23530055

RESUMEN

Cellular processes are tightly controlled through well-coordinated signaling networks that respond to conflicting cues, such as reactive oxygen species (ROS), endoplasmic reticulum (ER) stress signals, and survival factors to ensure proper cell function. We report here a direct interaction between inhibitor of κB kinase (IKK) and apoptosis signal-regulating kinase 1 (ASK1), unveiling a critical node at the junction of survival, inflammation, and stress signaling networks. IKK can be activated by growth factor stimulation or tumor necrosis factor alpha engagement. IKK forms a complex with and phosphorylates ASK1 at a sensor site, Ser967, leading to the recruitment of 14-3-3, counteracts stress signal-triggered ASK1 activation, and suppresses ASK1-mediated functions. An inhibitory role of IKK in JNK signaling has been previously reported to depend on NF-κB-mediated gene expression. Our data suggest that IKK has a dual role: a transcription-dependent and a transcription-independent action in controlling the ASK1-JNK axis, coupling IKK to ROS and ER stress response. Direct phosphorylation of ASK1 by IKK also defines a novel IKK phosphorylation motif. Because of the intimate involvement of ASK1 in diverse diseases, the IKK/ASK1 interface offers a promising target for therapeutic development.


Asunto(s)
Quinasa I-kappa B/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/fisiología , Células COS/efectos de los fármacos , Células COS/metabolismo , Chlorocebus aethiops , Cromonas/farmacología , Peróxido de Hidrógeno/farmacología , Quinasa I-kappa B/genética , MAP Quinasa Quinasa Quinasa 5/genética , Ratones , Morfolinas/farmacología , Neuritas/fisiología , Células PC12 , Fosforilación , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Serina/metabolismo , Transducción de Señal
15.
Nat Immunol ; 14(5): 461-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23525087

RESUMEN

Inflammation is essential for host defense but can cause tissue damage and organ failure if unchecked. How the inflammation is resolved remains elusive. Here we report that the transcription factor Miz1 was required for terminating lipopolysaccharide (LPS)-induced inflammation. Genetic disruption of the Miz1 POZ domain, which is essential for the transactivation or repression activity of Miz1, resulted in hyperinflammation, lung injury and greater mortality in LPS-treated mice but a lower bacterial load and mortality in mice with Pseudomonas aeruginosa pneumonia. Loss of the Miz1 POZ domain prolonged the expression of proinflammatory cytokines. After stimulation, Miz1 was phosphorylated at Ser178, which was required for recruitment of the histone deacetylase HDAC1 to repress transcription of the gene encoding C/EBP-δ, an amplifier of inflammation. Our data provide a long-sought mechanism underlying the resolution of LPS-induced inflammation.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Lesión Pulmonar Aguda/genética , Animales , Citocinas/metabolismo , Represión Enzimática/genética , Histona Desacetilasa 1/metabolismo , Tolerancia Inmunológica , Inflamación/genética , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutagénesis Sitio-Dirigida , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Fosforilación , Proteínas Inhibidoras de STAT Activados/genética , Infecciones por Pseudomonas/genética , Proteínas Represoras/genética , Activación Transcripcional/genética , Ubiquitina-Proteína Ligasas
16.
Cell ; 152(1-2): 304-15, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23332762

RESUMEN

The IκB kinase complex (IKK) is a key regulator of immune responses, inflammation, cell survival, and tumorigenesis. The prosurvival function of IKK centers on activation of the transcription factor NF-κB, whose target gene products inhibit caspases and prevent prolonged JNK activation. Here, we report that inactivation of the BH3-only protein BAD by IKK independently of NF-κB activation suppresses TNFα-induced apoptosis. TNFα-treated Ikkß(-/-) mouse embryonic fibroblasts (MEFs) undergo apoptosis significantly faster than MEFs deficient in both RelA and cRel due to lack of inhibition of BAD by IKK. IKK phosphorylates BAD at serine-26 (Ser26) and primes it for inactivation. Elimination of Ser26 phosphorylation promotes BAD proapoptotic activity, thereby accelerating TNFα-induced apoptosis in cultured cells and increasing mortality in animals. Our results reveal that IKK inhibits TNFα-induced apoptosis through two distinct but cooperative mechanisms: activation of the survival factor NF-κB and inactivation of the proapoptotic BH3-only BAD protein.


Asunto(s)
Apoptosis , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Animales , Fibroblastos/citología , Quinasa I-kappa B/genética , Ratones , Ratones Noqueados , Fosforilación , Serina/metabolismo , Proteína Letal Asociada a bcl/química , Proteína Letal Asociada a bcl/genética , Proteína bcl-X/metabolismo
17.
PLoS One ; 7(11): e48611, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23139803

RESUMEN

A-to-I RNA editing catalyzed by the two main members of the adenosine deaminase acting on RNA (ADAR) family, ADAR1 and ADAR2, represents a RNA-based recoding mechanism implicated in a variety of cellular processes. Previously we have demonstrated that the expression of ADAR2 in pancreatic islet ß-cells is responsive to the metabolic cues and ADAR2 deficiency affects regulated cellular exocytosis. To investigate the molecular mechanism by which ADAR2 is metabolically regulated, we found that in cultured ß-cells and primary islets, the stress-activated protein kinase JNK1 mediates the upregulation of ADAR2 in response to changes of the nutritional state. In parallel with glucose induction of ADAR2 expression, JNK phosphorylation was concurrently increased in insulin-secreting INS-1 ß-cells. Pharmacological inhibition of JNKs or siRNA knockdown of the expression of JNK1 prominently suppressed glucose-augmented ADAR2 expression, resulting in decreased efficiency of ADAR2 auto-editing. Consistently, the mRNA expression of Adar2 was selectively reduced in the islets from JNK1 null mice in comparison with that of wild-type littermates or JNK2 null mice, and ablation of JNK1 diminished high-fat diet-induced Adar2 expression in the islets from JNK1 null mice. Furthermore, promoter analysis of the mouse Adar2 gene identified a glucose-responsive region and revealed the transcription factor c-Jun as a driver of Adar2 transcription. Taken together, these results demonstrate that JNK1 serves as a crucial component in mediating glucose-responsive upregulation of ADAR2 expression in pancreatic ß-cells. Thus, the JNK1 pathway may be functionally linked to the nutrient-sensing actions of ADAR2-mediated RNA editing in professional secretory cells.


Asunto(s)
Adenosina Desaminasa/genética , Glucosa/farmacología , Células Secretoras de Insulina/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Edición de ARN/genética , Regulación hacia Arriba/efectos de los fármacos , Adenosina Desaminasa/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Dieta Alta en Grasa , Activación Enzimática/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Noqueados , Obesidad/enzimología , Obesidad/genética , Obesidad/patología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Edición de ARN/efectos de los fármacos , Ratas , Regulación hacia Arriba/genética
18.
Proc Natl Acad Sci U S A ; 109(1): 191-6, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22184250

RESUMEN

The transcription factor zinc-finger protein Miz1 represses TNF-α-induced JNK activation and the repression is relieved upon TNF-α stimulation. However, the underlying mechanism is incompletely understood. Here we report that Miz1 interferes with the ubiquitin conjugating enzyme (E2) Ubc13 for binding to the RING domain of TNF-receptor associated factor 2 (TRAF2), thereby inhibiting the ubiquitin ligase (E3) activity of TRAF2 and suppressing TNF-α-induced JNK activation. Upon TNF-α stimulation, Miz1 rapidly undergoes K48-linked polyubiquitination at Lys388 and Lys472 residues and subsequent proteasomal degradation in a TRAF2-dependent manner. Replacement of Lysine 388 and Lysine 472 by arginines generates a nondegradable Miz1 mutant, which significantly suppresses TNF-α-induced JNK1 activation and inflammation. Thus, our results reveal a molecular mechanism by which the repression of TNF-α-induced JNK activation by Miz1 is de-repressed by its own site-specific ubiquitination and degradation, which may account for the temporal control of TNF-α-JNK signaling.


Asunto(s)
Inflamación/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitinación/efectos de los fármacos , Animales , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Inflamación/genética , Inflamación/patología , Mediadores de Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/deficiencia , Lisina/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Modelos Biológicos , Proteínas Nucleares/deficiencia , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Inhibidoras de STAT Activados/deficiencia , Proteolisis/efectos de los fármacos , Factor 2 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
19.
Mol Cell ; 42(5): 557-8, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21658597

RESUMEN

In this issue of Molecular Cell,Choksi et al. (2011) report the identification of an NF-κB-independent ATIA (anti-TNFα-induced apoptosis)-Thioredoxin 2 axis that inhibits TNFα- and hypoxia-induced apoptosis through elimination of excessive reactive oxygen species directly.

20.
Diabetes ; 60(2): 486-95, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21270260

RESUMEN

OBJECTIVE: Obesity is associated with increased activation of the c-Jun NH(2)-terminal kinase (JNK) in several metabolic organs, including adipose tissue, liver, and skeletal muscle. In this study, we aimed to define the role of JNK activation in adipose tissue in the development of obesity-related insulin resistance. RESEARCH DESIGN AND METHODS: Transgenic mice with adipose tissue-specific overexpression of dominant-negative JNK (ap2-dn-JNK) under the transcriptional control of the aP2 gene promoter were generated and subjected to metabolic characterization together with the wild-type littermates. RESULTS: On a high-fat diet (HFD), the ap2-dn-JNK mice displayed a marked suppression of both JNK1 and JNK2 activation in their adipose tissue, accompanied by a marked reduction in weight gain, fat mass, and size of the adipocytes. The transgenic mice were resistant to the deleterious impact of an HFD on systemic insulin sensitivity, glucose tolerance, and hepatic steatosis. Reduced hepatic gluconeogenesis was evident in in vivo and ex vivo studies and showed greater insulin-induced glucose uptake in skeletal muscles. These changes were accompanied by reduced macrophage infiltration in adipose tissue, decreased production of proinflammatory adipokines, and increased expression of adiponectin. Indirect calorimetry analysis showed that the transgenic mice had significant increases in oxygen consumption and reductions in respiration exchange rates compared with their wild-type littermates. CONCLUSIONS: Selective suppression of JNK activation in adipose tissue alone is sufficient to counteract HFD-induced obesity and its associated metabolic dysregulations, in part through an increase in energy expenditure and a decrease in systemic inflammation.


Asunto(s)
Tejido Adiposo/metabolismo , Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Análisis de Varianza , Animales , Calorimetría Indirecta , Dieta , Grasas de la Dieta/metabolismo , Hígado Graso/metabolismo , Inmunohistoquímica , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ratones , Ratones Transgénicos , Obesidad/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA