Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Structure ; 32(9): 1394-1403.e5, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39013462

ABSTRACT

The scaffold proteins JIP1 and JIP2 intervene in the c-Jun N-terminal kinase (JNK) pathway to mediate signaling specificity by coordinating the simultaneous assembly of multiple kinases. Using NMR, we demonstrate that JIP1 and JIP2 heterodimerize via their SH3 domains with the affinity of heterodimerization being comparable to homodimerization. We present the high-resolution crystal structure of the JIP2-SH3 homodimer and the JIP1-JIP2-SH3 heterodimeric complex. The JIP2-SH3 structure reveals how charge differences in residues at its dimer interface lead to formation of compensatory hydrogen bonds and salt bridges, distinguishing it from JIP1-SH3. In the JIP1-JIP2-SH3 complex, structural features of each homodimer are employed to stabilize the heterodimer. Building on these insights, we identify key residues crucial for stabilizing the dimer of both JIP1 and JIP2. Through targeted mutations in cellulo, we demonstrate a functional role for the dimerization of the JIP1 and JIP2 scaffold proteins in activation of the JNK signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Models, Molecular , Protein Multimerization , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Binding Sites , Crystallography, X-Ray , Protein Binding
2.
Front Cell Dev Biol ; 12: 1371568, 2024.
Article in English | MEDLINE | ID: mdl-38606319

ABSTRACT

The mammalian brain, especially the cerebral cortex, has evolved to increase in size and complexity. The proper development of the cerebral cortex requires the coordination of several events, such as differentiation and migration, that are essential for forming a precise six-layered structure. We have previously reported that Cdk5-mediated phosphorylation of JIP1 at T205 modulates axonal out-growth. However, the spatiotemporal expression patterns and functions of these three genes (Cdk5, Cdk5r1 or p35, and Mapk8ip1 or JIP1) in distinct cell types during cortical development remain unclear. In this study, we analyzed single-cell RNA-sequencing data of mouse embryonic cortex and discovered that Cdk5, p35, and JIP1 are dynamically expressed in intermediate progenitors (IPs). Pseudotime analysis revealed that the expression of these three genes was concomitantly upregulated in IPs during neuronal migration and differentiation. By manipulating the expression of JIP1 and phospho-mimetic JIP1 using in utero electroporation, we showed that phosphorylated JIP1 at T205 affected the temporal migration of neurons.

3.
BMC Biol ; 20(1): 115, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35581583

ABSTRACT

BACKGROUND: Activated Cdk5 regulates a number of processes during nervous system formation, including neuronal differentiation, growth cone stabilization, and axonal growth. Cdk5 phosphorylates its downstream substrates located in axonal growth cones, where the highly expressed c-Jun N-terminal kinase (JNK)-interacting protein1 (JIP1) has been implicated as another important regulator of axonal growth. In addition, stringent control of the level of intracellular domain of Notch1 (Notch1-IC) plays a regulatory role in axonal outgrowth during neuronal differentiation. However, whether Cdk5-JIP1-Notch1 cooperate to regulate axonal outgrowth, and the mechanism of such joint contribution to this pathway, is presently unknown, and here we explore their potential interaction. RESULTS: Our interactome screen identified JIP1 as an interactor of p35, a Cdk5 activator, and we sought to explore the relationship between Cdk5 and JIP1 on the regulation of axonal outgrowth. We demonstrate that JIP1 phosphorylated by Cdk5 at Thr205 enhances axonal outgrowth and a phosphomimic JIP1 rescues the axonal outgrowth defects in JIP1-/- and p35-/- neurons. Axonal outgrowth defects caused by the specific increase of Notch1 in JIP1-/- neurons are rescued by Numb-mediated inhibition of Notch1. Finally, we demonstrate that Cdk5 phosphorylation of JIP1 further amplifies the phosphorylation status of yet another Cdk5 substrate E3-ubiquitin ligase Itch, resulting in increased Notch1 ubiquitination. CONCLUSIONS: Our findings identify a potentially critical signaling axis involving Cdk5-JIP1-Itch-Notch1, which plays an important role in the regulation of CNS development. Future investigation into the way this pathway integrates with additional pathways regulating axonal growth will further our knowledge of normal central nervous system development and pathological conditions.


Subject(s)
Neurons , Signal Transduction , Cells, Cultured , Neurons/metabolism , Phosphorylation , Signal Transduction/physiology
4.
Cells ; 9(10)2020 09 28.
Article in English | MEDLINE | ID: mdl-32998477

ABSTRACT

The c-Jun N-terminal kinase 3 (JNK3) is the JNK isoform mainly expressed in the brain. It is the most responsive to many stress stimuli in the central nervous system from ischemia to Aß oligomers toxicity. JNK3 activity is spatial and temporal organized by its scaffold protein, in particular JIP-1 and ß-arrestin-2, which play a crucial role in regulating different cellular functions in different cellular districts. Extensive evidence has highlighted the possibility of exploiting these adaptors to interfere with JNK3 signaling in order to block its action. JNK plays a key role in the first neurodegenerative event, the perturbation of physiological synapse structure and function, known as synaptic dysfunction. Importantly, this is a common mechanism in many different brain pathologies. Synaptic dysfunction and spine loss have been reported to be pharmacologically reversible, opening new therapeutic directions in brain diseases. Being JNK3-detectable at the peripheral level, it could be used as a disease biomarker with the ultimate aim of allowing an early diagnosis of neurodegenerative and neurodevelopment diseases in a still prodromal phase.


Subject(s)
Brain/metabolism , Mitogen-Activated Protein Kinase 10/genetics , Neurodegenerative Diseases/genetics , Neurodevelopmental Disorders/genetics , Biomarkers/metabolism , Brain/pathology , Humans , MAP Kinase Signaling System/genetics , Molecular Targeted Therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Phosphorylation
5.
Front Cell Dev Biol ; 7: 225, 2019.
Article in English | MEDLINE | ID: mdl-31681759

ABSTRACT

Retinal ganglion cells (RGCs) undergo apoptosis after injury. c-Jun N-terminal kinase (JNK)-interacting protein 1 (JIP1) is a scaffold protein that is relevant to JNK activation and a key molecule known to regulate neuronal apoptosis. However, the specific role of JIP1 in the apoptosis of RGCs is currently undefined. Here, we used JIP1 gene knockout (KO) mice to investigate the importance of JIP1-JNK signaling in the apoptosis of RGCs in a rotenone-induced injury model. In adult JIP1 KO mice, the number and electrophysiological functions of RGCs were not different from those of wild-type (WT) mice. Ablation of JIP1 attenuated the activation of JNK and the cleavage of caspase-3 in the retina after rotenone injury and contributed to a lower number of TUNEL-positive RGCs, a greater percentage of surviving RGCs, and a significant reduction in the electrophysiological functional loss of RGCs when compared to those in WT controls. We also found that JIP1 was located in the neurites of primary RGCs, but accumulated in soma in response to rotenone treatment. Moreover, the number of TUNEL-positive RGCs, the level of activation of JNK and the rate of cleavage of caspase-3 were reduced in primary JIP1-deficient RGCs after rotenone injury than in WT controls. Together, our results demonstrate that the JIP1-mediated activation of JNK contributes to the apoptosis of RGCs in a rotenone-induced injury model in vitro and in vivo, suggesting that JIP1 may be a potential therapeutic target for RGC degeneration.

6.
Behav Brain Res ; 359: 528-535, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30412737

ABSTRACT

Heme oxygenase (HO-1), which may be induced by Cobaltic protoporphyrin IX chloride (CoPPIX) or Rosiglitazone (Ros), is a neuroprotective agent that effectively reduces ischemic stroke. Previous studies have shown that the neuroprotective mechanisms of HO-1 are related to JNK signaling. The expression of HO-1 protects cells from death through the JNK signaling pathway. This study aimed to ascertain whether the neuroprotective effect of HO-1 depends on the assembly of the MLK3-MKK7-JNK3 signaling module scaffolded by JIP1 and further influences the JNK signal transmission through HO-1. Prior to the ischemia-reperfusion experiment, CoPPIX was injected through the lateral ventricle for 5 consecutive days or Ros was administered via intraperitoneal administration in the week prior to transient ischemia. Our results demonstrated that HO-1 could inhibit the assembly of the MLK3-MKK7-JNK3 signaling module scaffolded by JIP1 and could ultimately diminish the phosphorylation of JNK3. Furthermore, the inhibition of JNK3 phosphorylation downregulated the level of p-c-Jun and elevated neuronal cell death in the CA1 of the hippocampus. Taken together, these findings suggested that HO-1 could ameliorate brain injury by regulating the MLK3-MKK7-JNK3 signaling module, which was scaffolded by JIP1 and JNK signaling during cerebral ischemia/reperfusion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Brain Ischemia/enzymology , Heme Oxygenase (Decyclizing)/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinases/metabolism , Animals , Brain Ischemia/drug therapy , Brain Ischemia/pathology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/pathology , Cell Death/physiology , Disease Models, Animal , Down-Regulation , Male , Maze Learning/drug effects , Maze Learning/physiology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotection/drug effects , Neuroprotection/physiology , Neuroprotective Agents/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Rosiglitazone/pharmacology , Mitogen-Activated Protein Kinase Kinase Kinase 11
7.
J Cell Sci ; 131(6)2018 03 16.
Article in English | MEDLINE | ID: mdl-29487176

ABSTRACT

During muscle development, myonuclei undergo a complex set of movements that result in evenly spaced nuclei throughout the muscle cell. In Drosophila, two separate pools of Kinesin and Dynein work in synchrony to drive this process. However, how these two pools are specified is not known. Here, we investigate the role of Aplip1 (the Drosophila homolog of JIP1, JIP1 is also known as MAPK8IP1), a known regulator of both Kinesin and Dynein, in myonuclear positioning. Aplip1 localizes to the myotendinous junction and has genetically separable roles in myonuclear positioning and muscle stability. In Aplip1 mutant embryos, there was an increase in the percentage of embryos that had both missing and collapsed muscles. Via a separate mechanism, we demonstrate that Aplip1 regulates both the final position of and the dynamic movements of myonuclei. Aplip1 genetically interacts with both Raps (also known as Pins) and Kinesin to position myonuclei. Furthermore, Dynein and Kinesin localization are disrupted in Aplip1 mutants suggesting that Aplip1-dependent nuclear positioning requires Dynein and Kinesin. Taken together, these data are consistent with Aplip1 having a function in the regulation of Dynein- and Kinesin-mediated pulling of nuclei from the muscle end.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Muscle Development , Muscles/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Dyneins/genetics , Dyneins/metabolism , Kinesins/genetics , Kinesins/metabolism
8.
J Neurosci ; 38(15): 3708-3728, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29540552

ABSTRACT

The c-Jun N-terminal kinase (JNK) signal transduction pathway is implicated in learning and memory. Here, we examined the role of JNK activation mediated by the JNK-interacting protein 1 (JIP1) scaffold protein. We compared male wild-type mice with a mouse model harboring a point mutation in the Jip1 gene that selectively blocks JIP1-mediated JNK activation. These male mutant mice exhibited increased NMDAR currents, increased NMDAR-mediated gene expression, and a lower threshold for induction of hippocampal long-term potentiation. The JIP1 mutant mice also displayed improved hippocampus-dependent spatial memory and enhanced associative fear conditioning. These results were confirmed using a second JIP1 mutant mouse model that suppresses JNK activity. Together, these observations establish that JIP1-mediated JNK activation contributes to the regulation of hippocampus-dependent, NMDAR-mediated synaptic plasticity and learning.SIGNIFICANCE STATEMENT The results of this study demonstrate that c-Jun N-terminal kinase (JNK) activation induced by the JNK-interacting protein 1 (JIP1) scaffold protein negatively regulates the threshold for induction of long-term synaptic plasticity through the NMDA-type glutamate receptor. This change in plasticity threshold influences learning. Indeed, mice with defects in JIP1-mediated JNK activation display enhanced memory in hippocampus-dependent tasks, such as contextual fear conditioning and Morris water maze, indicating that JIP1-JNK constrains spatial memory. This study identifies JIP1-mediated JNK activation as a novel molecular pathway that negatively regulates NMDAR-dependent synaptic plasticity and memory.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Neuronal Plasticity , Spatial Memory , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Conditioning, Classical , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Point Mutation , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
9.
Artif Cells Nanomed Biotechnol ; 46(sup1): 684-692, 2018.
Article in English | MEDLINE | ID: mdl-29475393

ABSTRACT

Cationic liposomes have been investigated as non-viral vectors for gene delivery for more than a decade to overcome challenges associated with viral gene delivery. However, due to instability of liposomes, siRNA delivery is still a serious problem. In this study, we developed stealth PEGylated liposome formulations and focused on the effects of PEGylated liposomes on parameters related to size, zeta potential, polydispersity index, siRNA-loading efficiency and long-term stability of the siRNA-liposome complex. We were able to generate siRNA lipoplexes that could be very efficiently loaded, did not aggregate, could be stored at 4 °C for at least 6 months with only marginal release (1-5%) of siRNA and enhanced intracellular delivery of siRNA. Moreover, we could demonstrate that PEGylation positively contributed to all these parameters compared to liposomes, which were not PEGylated. The prepared lipoplex was successfully silenced J1P1 expression in MG-63 osteosarcoma cell line. In conclusion, our novel PEGylated liposomes have high potential for systemic delivery of siRNA and can improve in vivo stability of free siRNA and also siRNA lipoplexes.


Subject(s)
Liposomes/chemistry , Nanostructures/chemistry , Polyethylene Glycols/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Humans , Polyethylene Glycols/toxicity , Transfection
10.
Cell Rep ; 20(12): 2775-2783, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930674

ABSTRACT

Obesity is a major risk factor for the development of metabolic syndrome and type 2 diabetes. How obesity contributes to metabolic syndrome is unclear. Free fatty acid (FFA) activation of a non-receptor tyrosine kinase (SRC)-dependent cJun NH2-terminal kinase (JNK) signaling pathway is implicated in this process. However, the mechanism that mediates SRC-dependent JNK activation is unclear. Here, we identify a role for the scaffold protein JIP1 in SRC-dependent JNK activation. SRC phosphorylation of JIP1 creates phosphotyrosine interaction motifs that bind the SH2 domains of SRC and the guanine nucleotide exchange factor VAV. These interactions are required for SRC-induced activation of VAV and the subsequent engagement of a JIP1-tethered JNK signaling module. The JIP1 scaffold protein, therefore, plays a dual role in FFA signaling by coordinating upstream SRC functions together with downstream effector signaling by the JNK pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Stress, Physiological , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Animals , Diet, High-Fat , Enzyme Activation/drug effects , Insulin Resistance , Male , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice, Inbred C57BL , Palmitates/pharmacology , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Domains , Stress, Physiological/drug effects
11.
Proteins ; 85(2): 221-234, 2017 02.
Article in English | MEDLINE | ID: mdl-27891669

ABSTRACT

Mutations in the amyloid precursor protein (APP) are responsible for the formation of amyloid-ß peptides. These peptides play a role in Alzheimer's and other dementia-related diseases. The cargo binding domain of the kinesin-1 light chain motor protein (KLC1) may be responsible for transporting APP either directly or via interaction with C-jun N-terminal kinase-interacting protein 1 (JIP1). However, to date there has been no direct experimental or computational assessment of such binding at the atomistic level. We used molecular dynamics and free energy estimations to gauge the affinity for the binary complexes of KLC1, APP, and JIP1. We find that all binary complexes (KLC1:APP, KLC1:JIP1, and APP:JIP1) contain conformations with favorable binding free energies. For KLC1:APP the inclusion of approximate entropies reduces the favorability. This is likely due to the flexibility of the 42-residue APP protein. In all cases we analyze atomistic/residue driving forces for favorable interactions. Proteins 2017; 85:221-234. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Amyloid beta-Protein Precursor/chemistry , Microtubule-Associated Proteins/chemistry , Molecular Dynamics Simulation , Amino Acid Motifs , Binding Sites , Humans , Kinesins , Kinetics , Molecular Docking Simulation , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Thermodynamics
12.
Autophagy ; 10(11): 2079-81, 2014.
Article in English | MEDLINE | ID: mdl-25483967

ABSTRACT

Autophagy is a spatially regulated process in axons; autophagosomes form preferentially in the distal axon tip then move actively and processively toward the cell body. Despite the primarily unidirectional transport observed in live-cell imaging experiments, both anterograde-directed KIF5/kinesin-1 motors and retrograde-directed dynein motors are tightly associated with axonal autophagosomes. Here, we discuss our recent work identifying the scaffolding protein MAPK8IP1/JIP1 (mitogen-activated protein kinase 8 interacting protein 1) as a key regulator of autophagosome transport in neurons. MAPK8IP1 tightly coordinates motor activity to ensure the fidelity of retrograde autophagosome transport in the axon.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Neurons/metabolism , Neurons/pathology , Phagosomes , Axons/metabolism , Axons/physiology , Gene Expression Regulation , Humans , Kinesins/metabolism , MAP Kinase Signaling System , Microtubule-Associated Proteins/metabolism , Mutation , Phosphorylation , Vesicular Transport Proteins/metabolism
13.
J Neurosci ; 34(5): 1710-23, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24478353

ABSTRACT

Axon development and elongation require strictly controlled new membrane addition. Previously, we have shown the involvement of Rab10 in directional membrane insertion of plasmalemmal precursor vesicles (PPVs) during neuronal polarization and axonal growth. However, the mechanism responsible for PPV transportation remains unclear. Here we show that c-Jun N-terminal kinase-interacting protein 1 (JIP1) interacts with GTP-locked active form of Rab10 and directly connects Rab10 to kinesin-1 light chain (KLC). The kinesin-1/JIP1/Rab10 complex is required for anterograde transport of PPVs during axonal growth. Downregulation of JIP1 or KLC or disrupting the formation of this complex reduces anterograde transport of PPVs in developing axons and causes neuronal polarity defect. Furthermore, this complex plays an important role in neocortical neuronal polarization of rats in vivo. Thus, this study has demonstrated a mechanism underlying directional membrane trafficking involved in axon development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity/physiology , Neurons/physiology , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Axons/metabolism , Brain/cytology , Cells, Cultured , Cellular Structures/metabolism , Embryo, Mammalian , Female , Gene Expression Regulation/physiology , Humans , Ki-67 Antigen/metabolism , Kinesins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Microscopy, Confocal , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Protein Transport/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , T-Box Domain Proteins/metabolism , rab GTP-Binding Proteins/genetics
14.
J Cell Sci ; 127(Pt 1): 230-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24198394

ABSTRACT

The polarisation of developing neurons to form axons and dendrites is required for the establishment of neuronal connections leading to proper brain function. The protein kinase AKT and the MAP kinase scaffold protein JNK-interacting protein-1 (JIP1) are important regulators of axon formation. Here we report that JIP1 and AKT colocalise in axonal growth cones of cortical neurons and collaborate to promote axon growth. The loss of AKT protein from the growth cone results in the degradation of JIP1 by the proteasome, and the loss of JIP1 promotes a similar fate for AKT. Reduced protein levels of both JIP1 and AKT in the growth cone can be induced by glutamate and this coincides with reduced axon growth, which can be rescued by a stabilized mutant of JIP1 that rescues AKT protein levels. Taken together, our data reveal a collaborative relationship between JIP1 and AKT that is required for axon growth and can be regulated by changes in neuronal activity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental , Growth Cones/metabolism , Proto-Oncogene Proteins c-akt/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Embryo, Mammalian , Glutamic Acid/pharmacology , Growth Cones/drug effects , Growth Cones/ultrastructure , Mice , Mice, Inbred C57BL , Primary Cell Culture , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
15.
Adipocyte ; 1(3): 157-160, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-23700525

ABSTRACT

Abnormal adipokine production, along with defective uptake and metabolism of glucose within adipocytes, contributes to insulin resistance and altered glucose homeostasis. Recent research has highlighted one of the mechanisms that accounts for impaired production of adiponectin (ADIPOQ) and adipocyte glucose uptake in obesity. In adipocytes of human obese subjects and mice fed with a high fat diet, the level of the inducible cAMP early repressor (ICER) is diminished. Reduction of ICER elevates the cAMP response element binding protein (CREB) activity, which in turn increases the repressor activating transcription factor 3. In fine, the cascade triggers reduction in the ADIPOQ and GLUT4 levels, which ultimately hampers insulin-mediated glucose uptake. The c-Jun N-terminal kinase (JNK) interacting-protein 1, also called islet brain 1 (IB1), is a target of CREB/ICER that promotes JNK-mediated insulin resistance in adipocytes. A rise in IB1 and c-Jun levels accompanies the drop of ICER in white adipose tissues of obese mice when compared with mice fed with a chow diet. Other than the expression of ADIPOQ and glucose transport, decline in ICER expression might impact insulin signaling. Impairment of ICER is a critical issue that will need major consideration in future therapeutic purposes.

16.
Article in English | WPRIM (Western Pacific) | ID: wpr-651963

ABSTRACT

Activation of c-Jun N-terminal kinase (JNK) is associated with a wide range of disparate cellular responses to extracellular stimuli. In mammals, three JNK isoforms are known, and their differential regulation occurs in a stimulus- or a cell type-dependent manner. However, the underlying mechanism of this differential regulation has not been clearly elucidated. Here we demonstrated that JNK1 and JNK3 were activated in SH-SY5Y cells after treatment with H2O2 or UV. In SH-SY5Y cells overexpressing mJIP1, a splicing variant of a JNK scaffold protein JIP1, the H2O2-induced activities of both JNK1 and JNK3 were significantly suppressed. In the same cell line, however, UV-induced JNK1 activity was significantly suppressed, but JNK3 activity was not. During the RA-induced differentiation of SH-SY5Y cells, JNK1 was activated, whereas JNK3 was not, and this JNK1 activation was completely abolished in the cells overexpressing mJIP1. These results suggest that JIP1 plays a role in the regulation of the isoform-specific activation of JNKs in stimulus-dependent manner.


Subject(s)
Humans , Cell Line , JNK Mitogen-Activated Protein Kinases , Mammals , Neuroblastoma , Protein Isoforms
SELECTION OF CITATIONS
SEARCH DETAIL