Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.687
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 35: 501-521, 2019 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-31590586

RESUMEN

The dual leucine zipper-bearing kinase (DLK) and leucine zipper-bearing kinase (LZK) are evolutionarily conserved MAPKKKs of the mixed-lineage kinase family. Acting upstream of stress-responsive JNK and p38 MAP kinases, DLK and LZK have emerged as central players in neuronal responses to a variety of acute and traumatic injuries. Recent studies also implicate their function in astrocytes, microglia, and other nonneuronal cells, reflecting their expanding roles in the multicellular response to injury and in disease. Of particular note is the potential link of these kinases to neurodegenerative diseases and cancer. It is thus critical to understand the physiological contexts under which these kinases are activated, as well as the signal transduction mechanisms that mediate specific functional outcomes. In this review we first provide a historical overview of the biochemical and functional dissection of these kinases. We then discuss recent findings on regulating their activity to enhance cellular protection following injury and in disease, focusing on but not limited to the nervous system.


Asunto(s)
Leucina Zippers/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Neuronas/metabolismo , Estrés Fisiológico/genética , Animales , Axones/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/virología , Neuroglía/metabolismo , Neuronas/virología , Regeneración/genética , Regeneración/fisiología , Células Madre/metabolismo , Estrés Fisiológico/fisiología , Heridas y Lesiones/genética , Heridas y Lesiones/metabolismo
2.
Mol Cell ; 84(1): 142-155, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38118452

RESUMEN

Cellular homeostasis is continuously challenged by environmental cues and cellular stress conditions. In their defense, cells need to mount appropriate stress responses that, dependent on the cellular context, signaling intensity, and duration, may have diverse outcomes. The stress- and mitogen-activated protein kinase (SAPK/MAPK) system consists of well-characterized signaling cascades that sense and transduce an array of different stress stimuli into biological responses. However, the physical and chemical nature of stress signals and how these are sensed by individual upstream MAP kinase kinase kinases (MAP3Ks) remain largely ambiguous. Here, we review the existing knowledge of how individual members of the large and diverse group of MAP3Ks sense specific stress signals through largely non-redundant mechanisms. We emphasize the large knowledge gaps in assigning function and stress signals for individual MAP3K family members and touch on the potential of targeting this class of proteins for clinical benefit.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas Quinasa Quinasa PAM , Animales , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Transducción de Señal , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Mamíferos/metabolismo
3.
Immunity ; 54(8): 1807-1824.e14, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380064

RESUMEN

The transcription factor forkhead box O1 (FOXO1), which instructs the dark zone program to direct germinal center (GC) polarity, is typically inactivated by phosphatidylinositol 3-kinase (PI3K) signals. Here, we investigated how FOXO1 mutations targeting this regulatory axis in GC-derived B cell non-Hodgkin lymphomas (B-NHLs) contribute to lymphomagenesis. Examination of primary B-NHL tissues revealed that FOXO1 mutations and PI3K pathway activity were not directly correlated. Human B cell lines bearing FOXO1 mutations exhibited hyperactivation of PI3K and Stress-activated protein kinase (SAPK)/Jun amino-terminal kinase (JNK) signaling, and increased cell survival under stress conditions as a result of alterations in FOXO1 transcriptional affinities and activation of transcriptional programs characteristic of GC-positive selection. When modeled in mice, FOXO1 mutations conferred competitive advantage to B cells in response to key T-dependent immune signals, disrupting GC homeostasis. FOXO1 mutant transcriptional signatures were prevalent in human B-NHL and predicted poor clinical outcomes. Thus, rather than enforcing FOXO1 constitutive activity, FOXO1 mutations enable co-option of GC-positive selection programs during the pathogenesis of GC-derived lymphomas.


Asunto(s)
Linfocitos B/citología , Proteína Forkhead Box O1/genética , Centro Germinal/inmunología , Linfoma de Células B/patología , Animales , Linfocitos B/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular/genética , Supervivencia Celular/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Linfoma de Células B/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología
4.
Genes Dev ; 35(1-2): 133-146, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33334822

RESUMEN

The cJun NH2-terminal kinase (JNK) signaling pathway is activated by metabolic stress and promotes the development of metabolic syndrome, including hyperglycemia, hyperlipidemia, and insulin resistance. This integrated physiological response involves cross-talk between different organs. Here we demonstrate that JNK signaling in adipocytes causes an increased circulating concentration of the hepatokine fibroblast growth factor 21 (FGF21) that regulates systemic metabolism. The mechanism of organ crosstalk is mediated by a feed-forward regulatory loop caused by JNK-regulated FGF21 autocrine signaling in adipocytes that promotes increased expression of the adipokine adiponectin and subsequent hepatic expression of the hormone FGF21. The mechanism of organ cross-talk places circulating adiponectin downstream of autocrine FGF21 expressed by adipocytes and upstream of endocrine FGF21 expressed by hepatocytes. This regulatory loop represents a novel signaling paradigm that connects autocrine and endocrine signaling modes of the same hormone in different tissues.


Asunto(s)
Tejido Adiposo/fisiología , Comunicación Autocrina/genética , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica/genética , Transducción de Señal/genética , Adipocitos/metabolismo , Adiponectina/metabolismo , Tejido Adiposo/fisiopatología , Animales , Sistema Endocrino/metabolismo , Metabolismo Energético/genética , Retroalimentación Fisiológica/fisiología , Factores de Crecimiento de Fibroblastos/sangre , Hepatocitos/metabolismo , Resistencia a la Insulina/genética , Hígado/metabolismo , MAP Quinasa Quinasa 4/deficiencia , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones
5.
Immunity ; 48(5): 897-910.e7, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752064

RESUMEN

Intestinal infection triggers potent immune responses to combat pathogens and concomitantly drives epithelial renewal to maintain barrier integrity. Current models propose that epithelial renewal is primarily driven by damage caused by reactive oxygen species (ROS). Here we found that in Drosophila, the Imd-NF-κB pathway controlled enterocyte (EC) shedding upon infection, via a mechanism independent of ROS-associated apoptosis. Mechanistically, the Imd pathway synergized with JNK signaling to induce epithelial cell shedding specifically in the context of bacterial infection, requiring also the reduced expression of the transcription factor GATAe. Furthermore, cell-specific NF-κB responses enabled simultaneous production of antimicrobial peptides (AMPs) and epithelial shedding in different EC populations. Thus, the Imd-NF-κB pathway is central to the intestinal antibacterial response by mediating both AMP production and the maintenance of barrier integrity. Considering the similarities between Drosophila Imd signaling and mammalian TNFR pathway, our findings suggest the existence of an evolutionarily conserved genetic program in immunity-induced epithelial shedding.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Bacterias/inmunología , Infecciones Bacterianas/inmunología , Proteínas de Drosophila/inmunología , Células Epiteliales/inmunología , FN-kappa B/inmunología , Animales , Animales Modificados Genéticamente , Péptidos Catiónicos Antimicrobianos/metabolismo , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiología , Enterocitos/inmunología , Enterocitos/metabolismo , Enterocitos/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/inmunología , Factores de Transcripción GATA/metabolismo , Regulación de la Expresión Génica/inmunología , Mucosa Intestinal/citología , FN-kappa B/metabolismo , Transducción de Señal/inmunología
6.
Development ; 150(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37997694

RESUMEN

Identification of signaling events that contribute to innate spinal cord regeneration in zebrafish can uncover new targets for modulating injury responses of the mammalian central nervous system. Using a chemical screen, we identify JNK signaling as a necessary regulator of glial cell cycling and tissue bridging during spinal cord regeneration in larval zebrafish. With a kinase translocation reporter, we visualize and quantify JNK signaling dynamics at single-cell resolution in glial cell populations in developing larvae and during injury-induced regeneration. Glial JNK signaling is patterned in time and space during development and regeneration, decreasing globally as the tissue matures and increasing in the rostral cord stump upon transection injury. Thus, dynamic and regional regulation of JNK signaling help to direct glial cell behaviors during innate spinal cord regeneration.


Asunto(s)
Traumatismos de la Médula Espinal , Regeneración de la Medula Espinal , Animales , Larva , Mamíferos , Regeneración Nerviosa/fisiología , Neuroglía/fisiología , Médula Espinal , Pez Cebra/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos
7.
Proc Natl Acad Sci U S A ; 120(4): e2218373120, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36656864

RESUMEN

The HER2+ subtype of human breast cancer is associated with the malignant transformation of luminal ductal cells of the mammary epithelium. The sequence analysis of tumor DNA identifies loss of function mutations and deletions of the MAP2K4 and MAP2K7 genes that encode direct activators of the JUN NH2-terminal kinase (JNK). We report that in vitro studies of human mammary epithelial cells with CRISPR-induced mutations in the MAPK and MAP2K components of the JNK pathway caused no change in growth in 2D culture, but these mutations promoted epithelial cell proliferation in 3D culture. Analysis of gene expression signatures in 3D culture demonstrated similar changes caused by HER2 activation and JNK pathway loss. The mechanism of signal transduction cross-talk may be mediated, in part, by JNK-suppressed expression of integrin α6ß4 that binds HER2 and amplifies HER2 signaling. These data suggest that HER2 activation and JNK pathway loss may synergize to promote breast cancer. To test this hypothesis, we performed in vivo studies using a mouse model of HER2+ breast cancer with Cre/loxP-mediated ablation of genes encoding JNK (Mapk8 and Mapk9) and the MAP2K (Map2k4 and Map2k7) that activate JNK in mammary epithelial cells. Kaplan-Meier analysis of tumor development demonstrated that JNK pathway deficiency promotes HER2+-driven breast cancer. Collectively, these data identify JNK pathway genes as potential suppressors for HER2+ breast cancer.


Asunto(s)
Neoplasias de la Mama , Sistema de Señalización de MAP Quinasas , Humanos , Femenino , Neoplasias de la Mama/patología , Transducción de Señal , Transformación Celular Neoplásica/genética , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Línea Celular Tumoral
8.
J Biol Chem ; 300(5): 107263, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582451

RESUMEN

Synapse formation depends on the coordinated expression and regulation of scaffold proteins. The JNK family kinases play a role in scaffold protein regulation, but the nature of this functional interaction in dendritic spines requires further investigation. Here, using a combination of biochemical methods and live-cell imaging strategies, we show that the dynamics of the synaptic scaffold molecule SAP102 are negatively regulated by JNK inhibition, that SAP102 is a direct phosphorylation target of JNK3, and that SAP102 regulation by JNK is restricted to neurons that harbor mature synapses. We further demonstrate that SAP102 and JNK3 cooperate in the regulated trafficking of kainate receptors to the cell membrane. Specifically, we observe that SAP102, JNK3, and the kainate receptor subunit GluK2 exhibit overlapping expression at synaptic sites and that modulating JNK activity influences the surface expression of the kainate receptor subunit GluK2 in a neuronal context. We also show that SAP102 participates in this process in a JNK-dependent fashion. In summary, our data support a model in which JNK-mediated regulation of SAP102 influences the dynamic trafficking of kainate receptors to postsynaptic sites, and thus shed light on common pathophysiological mechanisms underlying the cognitive developmental defects associated with diverse mutations.


Asunto(s)
Espinas Dendríticas , Receptor de Ácido Kaínico GluK2 , Receptores de Ácido Kaínico , Animales , Humanos , Ratas , Membrana Celular/metabolismo , Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Hipocampo/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/genética , Neuronas/metabolismo , Neuropéptidos , Fosforilación , Transporte de Proteínas , Receptores de Ácido Kaínico/metabolismo , Receptores de Ácido Kaínico/genética , Sinapsis/metabolismo , Células Cultivadas
9.
J Biol Chem ; 300(7): 107486, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38897570

RESUMEN

Aberrant regulation of signal transduction pathways can adversely derail biological processes for tissue development. One such process is the embryonic eyelid closure that is dependent on the mitogen-activated protein kinase kinase kinase 1 (MAP3K1). Map3k1 KO in mice results in defective eyelid closure and an autosomal recessive eye-open at birth phenotype. We have shown that in utero exposure to dioxin, a persistent environmental toxicant, induces the same eye defect in Map3k1+/- heterozygous but not WT pups. Here, we explore the mechanisms of the Map3k1 (gene) and dioxin (environment) interactions (GxE) underlying defective eyelid closure. We show that, acting through the aryl hydrocarbon receptor, dioxin activates epidermal growth factor receptor signaling, which in turn depresses MAP3K1-dependent Jun N-terminal kinase (JNK) activity. The dioxin-mediated JNK repression is moderate but is exacerbated by Map3k1 heterozygosity. Therefore, dioxin exposed Map3k1+/- embryonic eyelids have a marked reduction of JNK activity, accelerated differentiation and impeded polarization in the epithelial cells. Knocking out Ahr or Egfr in eyelid epithelium attenuates the open-eye defects in dioxin-treated Map3k1+/- pups, whereas knockout of Jnk1 and S1pr that encodes the sphigosin-1-phosphate (S1P) receptors upstream of the MAP3K1-JNK pathway potentiates the dioxin toxicity. Our novel findings show that the crosstalk of aryl hydrocarbon receptor, epidermal growth factor receptor, and S1P-MAP3K1-JNK pathways determines the outcome of dioxin exposure. Thus, gene mutations targeting these pathways are potential risk factors for the toxicity of environmental chemicals.


Asunto(s)
Dioxinas , Receptores ErbB , Quinasa 1 de Quinasa de Quinasa MAP , Receptores de Hidrocarburo de Aril , Animales , Femenino , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Dioxinas/toxicidad , Receptores ErbB/metabolismo , Receptores ErbB/genética , Párpados/metabolismo , Párpados/anomalías , Interacción Gen-Ambiente , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Noqueados , Receptor Cross-Talk , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal/efectos de los fármacos
10.
Development ; 149(9)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35352808

RESUMEN

The establishment of the left-right axis is crucial for the placement, morphogenesis and function of internal organs. Left-right specification is proposed to be dependent on cilia-driven fluid flow in the embryonic node. Planar cell polarity (PCP) signalling is crucial for patterning of nodal cilia, yet downstream effectors driving this process remain elusive. We have examined the role of the JNK gene family, a proposed downstream component of PCP signalling, in the development and function of the zebrafish node. We show jnk1 and jnk2 specify length of nodal cilia, generate flow in the node and restrict southpaw to the left lateral plate mesoderm. Moreover, loss of asymmetric southpaw expression does not result in disturbances to asymmetric organ placement, supporting a model in which nodal flow may be dispensable for organ laterality. Later, jnk3 is required to restrict pitx2c expression to the left side and permit correct endodermal organ placement. This work uncovers multiple roles for the JNK gene family acting at different points during left-right axis establishment. It highlights extensive redundancy and indicates JNK activity is distinct from the PCP signalling pathway.


Asunto(s)
Tipificación del Cuerpo , Pez Cebra , Animales , Tipificación del Cuerpo/genética , Cilios/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
11.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35067712

RESUMEN

The regenerative ability of planarians relies on their adult pluripotent stem cell population. Although all stem cells express a piwi homolog, recently it has become possible to classify the piwi+ stem cell population into specialized subpopulations according to the expression of genes related to differentiation. However, piwi+ stem cells behave practically as a homogeneous population after amputation, during which stem cells show accelerated proliferation, named 'induced hyperproliferation'. Here, we show that plac8-A was expressed in almost all of the stem cells, and that a decrease of the plac8-A expression level led to induced hyperproliferation uniformly in a broad stem cell subpopulation after amputation. This reduction of plac8-A expression was caused by activated JNK signaling after amputation. Pharmacological inhibition of JNK signaling caused failure to induce hyperproliferation and resulted in regenerative defects. Such defects were abrogated by simultaneous knockdown of plac8-A expression. Thus, JNK-dependent suppression of plac8-A expression is indispensable for stem cell dynamics involved in regeneration. These findings suggest that plac8-A acts as a molecular switch of piwi+ stem cells for entry into the regenerative state after amputation.


Asunto(s)
Proteínas del Helminto/genética , Planarias/fisiología , Regeneración/fisiología , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proliferación Celular , Regulación hacia Abajo , Proteínas del Helminto/antagonistas & inhibidores , Proteínas del Helminto/metabolismo , Sistema de Señalización de MAP Quinasas , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Interferencia de ARN , ARN Bicatenario/metabolismo
12.
J Virol ; 98(4): e0015924, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38499512

RESUMEN

Equine herpesvirus type 8 (EHV-8) causes abortion and respiratory disease in horses and donkeys, leading to serious economic losses in the global equine industry. Currently, there is no effective vaccine or drug against EHV-8 infection, underscoring the need for a novel antiviral drug to prevent EHV-8-induced latent infection and decrease the pathogenicity of this virus. The present study demonstrated that hyperoside can exert antiviral effects against EHV-8 infection in RK-13 (rabbit kidney cells), MDBK (Madin-Darby bovine kidney), and NBL-6 cells (E. Derm cells). Mechanistic investigations revealed that hyperoside induces heme oxygenase-1 expression by activating the c-Jun N-terminal kinase/nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 axis, alleviating oxidative stress and triggering a downstream antiviral interferon response. Accordingly, hyperoside inhibits EHV-8 infection. Meanwhile, hyperoside can also mitigate EHV-8-induced injury in the lungs of infected mice. These results indicate that hyperoside may serve as a novel antiviral agent against EHV-8 infection.IMPORTANCEHyperoside has been reported to suppress viral infections, including herpesvirus, hepatitis B virus, infectious bronchitis virus, and severe acute respiratory syndrome coronavirus 2 infection. However, its mechanism of action against equine herpesvirus type 8 (EHV-8) is currently unknown. Here, we demonstrated that hyperoside significantly inhibits EHV-8 adsorption and internalization in susceptible cells. This process induces HO-1 expression via c-Jun N-terminal kinase/nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 axis activation, alleviating oxidative stress and triggering an antiviral interferon response. These findings indicate that hyperoside could be very effective as a drug against EHV-8.


Asunto(s)
Antivirales , Infecciones por Herpesviridae , Herpesvirus Équido 1 , Sistema de Señalización de MAP Quinasas , Quercetina , Animales , Bovinos , Ratones , Conejos , Antivirales/farmacología , Caballos , Interferones/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Quercetina/análogos & derivados , Quercetina/farmacología , Línea Celular
13.
FASEB J ; 38(2): e23373, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38217376

RESUMEN

Fatigue is a common phenomenon closely related to physical discomfort and numerous diseases, which is severely threatening the life quality and health of people. However, the exact mechanisms underlying fatigue are not fully characterized. Herein, we demonstrate that oxaloacetic acid (OAA), a crucial tricarboxylic acid cycle intermediate, modulates the muscle fatigue. The results showed that serum OAA level was positively correlated with fatigue state of mice. OAA-treated induced muscle fatigue impaired the exercise performance of mice. Mechanistically, OAA increased the c-Jun N-terminal kinase (JNK) phosphorylation and uncoupling protein 2 (UCP2) levels in skeletal muscle, which led to decreased energy substrate and enhanced glycolysis. On the other hand, OAA boosted muscle mitochondrial oxidative phosphorylation uncoupled with energy production. In addition, either UCP2 knockout or JNK inhibition totally reversed the effects of OAA on skeletal muscle. Therein, JNK mediated UCP2 activation with OAA-treated. Our studies reveal a novel role of OAA in skeletal muscle metabolism, which would shed light on the mechanism of muscle fatigue and weakness.


Asunto(s)
Mitocondrias , Ácido Oxaloacético , Humanos , Ratones , Animales , Ácido Oxaloacético/metabolismo , Ácido Oxaloacético/farmacología , Mitocondrias/metabolismo , Fosforilación Oxidativa , Ciclo del Ácido Cítrico , Músculo Esquelético/metabolismo , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteína Desacopladora 3/metabolismo , Metabolismo Energético
14.
FASEB J ; 38(1): e23347, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38095503

RESUMEN

The pathogenesis of osteoarthritis (OA) is still unclear. Fatty acid binding protein 4 (FABP4), a novel adipokine, has been found to play a role in OA. This study aimed to explore the role of NF-κB in FABP4-induced OA. In the in vivo study, four pairs of 12-week-old male FABP4 knockout (KO) and wild-type (WT) mice were included. The activation of NF-κB was assessed. In parallel, 24 6-week-old male C57/Bl6 mice were fed a high-fat diet (HFD) and randomly allocated to four groups: daily oral gavage with (1) PBS solution; (2) QNZ (NF-κB-specific inhibitor, 1 mg/kg/d); (3) BMS309403 (FABP4-specific inhibitor, 30 mg/kg/d); and (4) BMS309403 (30 mg/kg/d) + QNZ (1 mg/kg/d). The diet and treatment were sustained for 4 months. The knee joints were obtained to assess cartilage degradation, NF-κB activation, and subchondral bone sclerosis. In the in vitro study, a mouse chondrogenic cell line (ATDC5) was cultured. FABP4 was supplemented to stimulate chondrocytes, and the activation of NF-κB was investigated. In parallel, QNZ and NF-κB-specific siRNA were used to inhibit NF-κB. In vivo, the FABP4 WT mice had more significant NF-κB activation than the KO mice. Dual inhibition of FABP4 and NF-κB alleviated knee OA in mice. FABP4 has no significant effect on the activation of the JNK signaling pathway. In vitro, FABP4 directly activated NF-κB in chondrocytes. The use of QNZ and NF-κB-siRNA significantly alleviated the expression of catabolic markers of chondrocytes induced by FABP4. FABP4 induces chondrocyte degeneration by activating the NF-κB pathway.


Asunto(s)
FN-kappa B , Osteoartritis de la Rodilla , Animales , Masculino , Ratones , Condrocitos/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Interleucina-1beta/metabolismo , FN-kappa B/metabolismo , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , ARN Interferente Pequeño/genética , Transducción de Señal
15.
Mol Cell ; 68(1): 185-197.e6, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28943315

RESUMEN

Many infections and stress signals can rapidly activate the NLRP3 inflammasome to elicit robust inflammatory responses. This activation requires a priming step, which is thought to be mainly for upregulating NLRP3 transcription. However, recent studies report that the NLRP3 inflammasome can be activated independently of transcription, suggesting that the priming process has unknown essential regulatory steps. Here, we report that JNK1-mediated NLRP3 phosphorylation at S194 is a critical priming event and is essential for NLRP3 inflammasome activation. We show that NLRP3 inflammasome activation is disrupted in NLRP3-S194A knockin mice. JNK1-mediated NLRP3 S194 phosphorylation is critical for NLRP3 deubiquitination and facilitates its self-association and the subsequent inflammasome assembly. Importantly, we demonstrate that blocking S194 phosphorylation prevents NLRP3 inflammasome activation in cryopyrin-associated periodic syndromes (CAPS). Thus, our study reveals a key priming molecular event that is a prerequisite for NLRP3 inflammasome activation. Inhibiting NLRP3 phosphorylation could be an effective treatment for NLRP3-related diseases.


Asunto(s)
Inflamasomas/genética , Macrófagos/inmunología , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Choque Séptico/genética , Secuencia de Aminoácidos , Animales , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/inmunología , Escherichia coli/química , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inflamasomas/inmunología , Lipopolisacáridos/farmacología , Macrófagos/patología , Masculino , Ratones , Ratones Transgénicos , Proteína Quinasa 8 Activada por Mitógenos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Fosforilación , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Choque Séptico/inducido químicamente , Choque Séptico/mortalidad , Choque Séptico/patología , Transducción de Señal , Análisis de Supervivencia
16.
Cell Mol Life Sci ; 81(1): 195, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38653877

RESUMEN

The Notch pathway is an evolutionarily conserved signaling system that is intricately regulated at multiple levels and it influences different aspects of development. In an effort to identify novel components involved in Notch signaling and its regulation, we carried out protein interaction screens which identified non-muscle myosin II Zipper (Zip) as an interacting partner of Notch. Physical interaction between Notch and Zip was further validated by co-immunoprecipitation studies. Immunocytochemical analyses revealed that Notch and Zip co-localize within same cytoplasmic compartment. Different alleles of zip also showed strong genetic interactions with Notch pathway components. Downregulation of Zip resulted in wing phenotypes that were reminiscent of Notch loss-of-function phenotypes and a perturbed expression of Notch downstream targets, Cut and Deadpan. Further, synergistic interaction between Notch and Zip resulted in highly ectopic expression of these Notch targets. Activated Notch-induced tumorous phenotype of larval tissues was enhanced by over-expression of Zip. Notch-Zip synergy resulted in the activation of JNK pathway that consequently lead to MMP activation and proliferation. Taken together, our results suggest that Zip may play an important role in regulation of Notch signaling.


Asunto(s)
Proteínas de Drosophila , Proteínas de la Membrana , Cadenas Pesadas de Miosina , Receptores Notch , Transducción de Señal , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores Notch/metabolismo , Receptores Notch/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Alas de Animales/metabolismo , Alas de Animales/crecimiento & desarrollo , Drosophila/metabolismo , Drosophila/genética , Fenotipo , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Proliferación Celular , Miosina Tipo II/metabolismo , Miosina Tipo II/genética
17.
Cell Mol Life Sci ; 81(1): 303, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39008099

RESUMEN

Vitamin C (VC) serves as a pivotal nutrient for anti-oxidation process, metabolic responses, and stem cell differentiation. However, its precise contribution to placenta development and gestation remains obscure. Here, we demonstrated that physiological levels of VC act to stabilize Hand1, a key bHLH transcription factor vital for the development trajectory of trophoblast giant cell (TGC) lineages, thereby promoting the differentiation of trophoblast stem cells into TGC. Specifically, VC administration inactivated c-Jun N-terminal kinase (JNK) signaling, which directly phosphorylates Hand1 at Ser48, triggering the proteasomal degradation of Hand1. Conversely, a loss-of-function mutation at Ser48 on Hand1 not only significantly diminished both intrinsic and VC-induced stabilization of Hand1 but also underscored the indispensability of this residue. Noteworthy, the insufficiency of VC led to severe defects in the differentiation of diverse TGC subtypes and the formation of labyrinth's vascular network in rodent placentas, resulting in failure of maintenance of pregnancy. Importantly, VC deficiency, lentiviral knockdown of JNK or overexpression of Hand1 mutants in trophectoderm substantially affected the differentiation of primary and secondary TGC in E8.5 mouse placentas. Thus, these findings uncover the significance of JNK inactivation and consequential stabilization of Hand1 as a hitherto uncharacterized mechanism controlling VC-mediated placentation and perhaps maintenance of pregnancy.


Asunto(s)
Ácido Ascórbico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Proteínas Quinasas JNK Activadas por Mitógenos , Placentación , Trofoblastos , Animales , Femenino , Embarazo , Ácido Ascórbico/farmacología , Ácido Ascórbico/metabolismo , Placentación/genética , Ratones , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Diferenciación Celular/efectos de los fármacos , Trofoblastos/metabolismo , Trofoblastos/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Placenta/metabolismo , Fosforilación , Humanos , Ratones Endogámicos C57BL
18.
Am J Respir Crit Care Med ; 210(4): 435-443, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38484130

RESUMEN

Rationale: Idiopathic pulmonary fibrosis is a fatal and progressive disease with limited treatment options. Objectives: We sought to assess the efficacy and safety of CC-90001, an oral inhibitor of c-Jun N-terminal kinase 1, in patients with idiopathic pulmonary fibrosis. Methods: In a Phase 2, randomized (1:1:1), double-blind, placebo-controlled study (ClinicalTrials.gov ID: NCT03142191), patients received CC-90001 (200 or 400 mg) or placebo once daily for 24 weeks. Background antifibrotic treatment (pirfenidone) was allowed. The primary endpoint was change in the percentage of predicted FVC (ppFVC) from baseline to Week 24; secondary endpoints included safety. Measurements and Main Results: In total, 112 patients received at least one dose of study drug. The study was terminated early because of a strategic decision made by the sponsor. Ninety-one patients (81%) completed the study. The least-squares mean changes from baseline in ppFVC at Week 24 were -3.1% (placebo), -2.1% (200 mg), and -1.0% (400 mg); the differences compared with placebo were 1.1% (200 mg; 95% confidence interval: -2.1, 4.3; P = 0.50) and 2.2% (400 mg; 95% confidence interval: -1.1, 5.4; P = 0.19). Adverse event frequency was similar in patients in the combined CC-90001 arms versus placebo. The most common adverse events were nausea, diarrhea, and vomiting, which were more frequent in patients in CC-90001 arms versus placebo. Fewer patients in the CC-90001 arms than in the placebo arm experienced cough and dyspnea. Conclusions: Treatment with CC-90001 over 24 weeks led to numerical improvements in ppFVC in patients with idiopathic pulmonary fibrosis compared with placebo. CC-90001 was generally well tolerated, which was consistent with previous studies. Clinical trial registered with www.clinicaltrials.gov (NCT03142191).


Asunto(s)
Fibrosis Pulmonar Idiopática , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/fisiopatología , Método Doble Ciego , Masculino , Femenino , Anciano , Persona de Mediana Edad , Resultado del Tratamiento , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Adulto
19.
Proc Natl Acad Sci U S A ; 119(44): e2210434119, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36282921

RESUMEN

The cJun NH2-terminal kinase (JNK) signaling pathway in the liver promotes systemic changes in metabolism by regulating peroxisome proliferator-activated receptor α (PPARα)-dependent expression of the hepatokine fibroblast growth factor 21 (FGF21). Hepatocyte-specific gene ablation studies demonstrated that the Mapk9 gene (encoding JNK2) plays a key mechanistic role. Mutually exclusive inclusion of exons 7a and 7b yields expression of the isoforms JNK2α and JNK2ß. Here we demonstrate that Fgf21 gene expression and metabolic regulation are primarily regulated by the JNK2α isoform. To identify relevant substrates of JNK2α, we performed a quantitative phosphoproteomic study of livers isolated from control mice, mice with JNK deficiency in hepatocytes, and mice that express only JNK2α or JNK2ß in hepatocytes. We identified the JNK substrate retinoid X receptor α (RXRα) as a protein that exhibited JNK2α-promoted phosphorylation in vivo. RXRα functions as a heterodimeric partner of PPARα and may therefore mediate the effects of JNK2α signaling on Fgf21 expression. To test this hypothesis, we established mice with hepatocyte-specific expression of wild-type or mutated RXRα proteins. We found that the RXRα phosphorylation site Ser260 was required for suppression of Fgf21 gene expression. Collectively, these data establish a JNK-mediated signaling pathway that regulates hepatic Fgf21 expression.


Asunto(s)
Síndrome Metabólico , PPAR alfa , Animales , Ratones , Proteínas Portadoras/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Síndrome Metabólico/metabolismo , Ratones Noqueados , Fosforilación , PPAR alfa/genética , PPAR alfa/metabolismo , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , MAP Quinasa Quinasa 4/metabolismo
20.
BMC Biol ; 22(1): 98, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38679694

RESUMEN

BACKGROUND: The ability of animals to regenerate damaged tissue is a complex process that involves various cellular mechanisms. As animals age, they lose their regenerative abilities, making it essential to understand the underlying mechanisms that limit regenerative ability during aging. Drosophila melanogaster wing imaginal discs are epithelial structures that can regenerate after tissue injury. While significant research has focused on investigating regenerative responses during larval stages our comprehension of the regenerative potential of pupal wings and the underlying mechanisms contributing to the decline of regenerative responses remains limited. RESULTS: Here, we explore the temporal dynamics during pupal development of the proliferative response triggered by the induction of cell death, a typical regenerative response. Our results indicate that the apoptosis-induced proliferative response can continue until 34 h after puparium formation (APF), beyond this point cell death alone is not sufficient to induce a regenerative response. Under normal circumstances, cell proliferation ceases around 24 h APF. Interestingly, the failure of reinitiating the cell cycle beyond this time point is not attributed to an incapacity to activate the JNK pathway. Instead, our results suggest that the function of the ecdysone-responsive transcription factor E93 is involved in limiting the apoptosis-induced proliferative response during pupal development. CONCLUSIONS: Our study shows that apoptosis can prolong the proliferative period of cells in the wing during pupal development as late as 34 h APF, at least 10 h longer than during normal development. After this time point, the regenerative response is diminished, a process mediated in part by the ecdysone-responsive transcription factor E93.


Asunto(s)
Apoptosis , Proliferación Celular , Proteínas de Drosophila , Drosophila melanogaster , Pupa , Regeneración , Factores de Transcripción , Alas de Animales , Animales , Alas de Animales/crecimiento & desarrollo , Alas de Animales/fisiología , Drosophila melanogaster/fisiología , Drosophila melanogaster/crecimiento & desarrollo , Pupa/crecimiento & desarrollo , Pupa/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Regeneración/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA