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










Publication year range
1.
Adv Sci (Weinh) ; 10(20): e2205949, 2023 07.
Article in English | MEDLINE | ID: mdl-37166058

ABSTRACT

Isocitrate dehydrogenase (IDH) mutation, a known pathologic classifier, initiates metabolic reprogramming in glioma cells and has been linked to the reaction status of glioma-associated microglia/macrophages (GAMs). However, it remains unclear how IDH genotypes contribute to GAM phenotypes. Here, it is demonstrated that gliomas expressing mutant IDH determine M1-like polarization of GAMs, while archetypal IDH induces M2-like polarization. Intriguingly, IDH-mutant gliomas secrete excess cholesterol, resulting in cholesterol-rich, pro-inflammatory GAMs without altering their cholesterol biosynthesis, and simultaneously exhibiting low levels of tumoral cholesterol due to expression remodeling of cholesterol transport molecules, particularly upregulation of ABCA1 and downregulation of LDLR. Mechanistically, a miR-19a/LDLR axis-mediated novel post-transcriptional regulation of cholesterol uptake is identified, modulated by IDH mutation, and influencing tumor cell proliferation and invasion. IDH mutation-induced PERK activation enhances cholesterol export from glioma cells via the miR-19a/LDLR axis and ABCA1/APOE upregulation. Further, a synthetic PERK activator, CCT020312 is introduced, which markedly stimulates cholesterol efflux from IDH wild-type glioma cells, induces M1-like polarization of GAMs, and consequently suppresses glioma cell invasion. The findings reveal an essential role of the PERK/miR-19a/LDLR signaling pathway in orchestrating gliomal cholesterol transport and the subsequent phenotypes of GAMs, thereby highlighting a novel potential target pathway for glioma therapy.


Subject(s)
Brain Neoplasms , Glioma , MicroRNAs , Humans , Brain Neoplasms/metabolism , Cholesterol , Glioma/metabolism , Isocitrate Dehydrogenase/genetics , Microglia/metabolism , MicroRNAs/genetics
2.
Front Mol Neurosci ; 16: 1071327, 2023.
Article in English | MEDLINE | ID: mdl-36969556

ABSTRACT

Depression is a common recurrent psychiatric disorder with a high lifetime prevalence and suicide rate. At present, although several traditional clinical drugs such as fluoxetine and ketamine, are widely used, medications with a high efficiency and reduced side effects are of urgent need. Our group has recently reported that a single administration of salmon calcitonin (sCT) could ameliorate a depressive-like phenotype via the amylin signaling pathway in a mouse model established by chronic restraint stress (CRS). However, the molecular mechanism underlying the antidepressant effect needs to be addressed. In this study, we investigated the antidepressant potential of sCT applied chronically and its underlying mechanism. In addition, using transcriptomics, we found the MAPK signaling pathway was upregulated in the hippocampus of CRS-treated mice. Further phosphorylation levels of ERK/p38/JNK kinases were also enhanced, and sCT treatment was able only to downregulate the phosphorylation level of p38/JNK, with phosphorylated ERK level unaffected. Finally, we found that the antidepressant effect of sCT was blocked by p38 agonists rather than JNK agonists. These results provide a mechanistic explanation of the antidepressant effect of sCT, suggesting its potential for treating the depressive disorder in the clinic.

3.
Elife ; 102021 10 12.
Article in English | MEDLINE | ID: mdl-34636321

ABSTRACT

Lung squamous cell carcinoma (LSCC) is a considerable global health burden, with an incidence of over 600,000 cases per year. Treatment options are limited, and patient's 5-year survival rate is less than 5%. The ubiquitin-specific protease 28 (USP28) has been implicated in tumourigenesis through its stabilization of the oncoproteins c-MYC, c-JUN, and Δp63. Here, we show that genetic inactivation of Usp28-induced regression of established murine LSCC lung tumours. We developed a small molecule that inhibits USP28 activity in the low nanomole range. While displaying cross-reactivity against the closest homologue USP25, this inhibitor showed a high degree of selectivity over other deubiquitinases. USP28 inhibitor treatment resulted in a dramatic decrease in c-MYC, c-JUN, and Δp63 proteins levels and consequently induced substantial regression of autochthonous murine LSCC tumours and human LSCC xenografts, thereby phenocopying the effect observed by genetic deletion. Thus, USP28 may represent a promising therapeutic target for the treatment of squamous cell lung carcinoma.


Subject(s)
DNA-Binding Proteins/genetics , Gene Deletion , Lung Neoplasms/genetics , Neoplasms, Squamous Cell/genetics , Transcription Factors/genetics , Ubiquitin Thiolesterase/genetics , Animals , DNA-Binding Proteins/metabolism , Disease Models, Animal , Humans , Mice , Transcription Factors/metabolism , Ubiquitin Thiolesterase/metabolism
4.
JCI Insight ; 6(13)2021 07 08.
Article in English | MEDLINE | ID: mdl-34236045

ABSTRACT

The AP-1 transcription factor c-Jun is required for Ras-driven tumorigenesis in many tissues and is considered as a classical proto-oncogene. To determine the requirement for c-Jun in a mouse model of K-RasG12D-induced lung adenocarcinoma, we inducibly deleted c-Jun in the adult lung. Surprisingly, we found that inactivation of c-Jun, or mutation of its JNK phosphorylation sites, actually increased lung tumor burden. Mechanistically, we found that protein levels of the Jun family member JunD were increased in the absence of c-Jun. In c-Jun-deficient cells, JunD phosphorylation was increased, and expression of a dominant-active JNKK2-JNK1 transgene further increased lung tumor formation. Strikingly, deletion of JunD completely abolished Ras-driven lung tumorigenesis. This work identifies JunD, not c-Jun, as the crucial substrate of JNK signaling and oncogene required for Ras-induced lung cancer.


Subject(s)
Adenocarcinoma of Lung , Carcinogenesis , Lung Neoplasms , Proto-Oncogene Proteins c-jun/metabolism , ras Proteins/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Genes, jun/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/metabolism , MAP Kinase Signaling System , Mice , Phosphorylation , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/metabolism
5.
Curr Cancer Drug Targets ; 21(7): 558-574, 2021.
Article in English | MEDLINE | ID: mdl-33949933

ABSTRACT

Gliomas are the most common type of malignant brain tumors. Despite significant medical advances, gliomas remain incurable and are associated with high mortality. Although numerous biomarkers of diagnostic value have been identified and significant progress in the prognosis of the outcome has been made, the treatment has not been parallelly improved during the last three decades. This review summarizes and discusses three aspects of recent discoveries related to glioma, with the objective to highlight the advantages of glioma-specific drugs targeting the cell of origin, microenvironment, and metabolism. Given the heterogeneous nature of gliomas, various cell populations have been implicated as likely sources of the tumor. Depending on the mutation(s) acquired by the cells, it is believed that neural stem/progenitor cells, oligodendrocyte progenitor cells, mature neurons, and glial cells can initiate cell transformation into a malignant phenotype. The level of tumorigenicity appears to be inversely correlated with the maturation of a given cell population. The microenvironment of gliomas includes non-cancer cells such as immune cells, fibroblasts, and cells of blood vessels, as well as secreted molecules and the extracellular matrix, and all these components play a vital role during tumor initiation and progression. We will discuss in detail how the tumor microenvironment can stimulate and drive the transformation of non-tumor cell populations into tumor-supporting cells or glioma cells. Metabolic reprogramming is a key feature of gliomas and is thought to reflect the adaptation to the increased nutritional requirements of tumor cell proliferation, growth, and survival. Mutations in the IDH gene can shape metabolic reprogramming and may generate some vulnerabilities in glioma cells, such as abnormal lipid metabolism and sensitivity to endoplasmic reticulum stress (ERS). We will analyze the prominent metabolic features of malignant gliomas and the key pathways regulating glioma metabolism. This review is intended to provide a conceptual background for the development of glioma therapies based on the properties of tumor cell populations, microenvironment, and metabolism.


Subject(s)
Brain Neoplasms , Glioma , Brain Neoplasms/drug therapy , Cell Transformation, Neoplastic , Glioma/drug therapy , Humans , Prognosis , Tumor Microenvironment
6.
Onco Targets Ther ; 12: 3467-3480, 2019.
Article in English | MEDLINE | ID: mdl-31123407

ABSTRACT

Introduction: Brain glioma is the most common type of primary malignancy in the central nervous system (CNS), with high recurrence and mortality rate, especially glioblastoma (GBM). Recent evidence suggests a role for many long noncoding RNAs (lncRNAs) in the pathogenesis, proliferation, apoptosis, metastasis, and chemotherapeutic resistance of cancer cells. Although the functions of some lncRNAs in the occurrence and development of gliomas have been confirmed, detailed mechanisms of action are lacking. Furthermore, the biological roles of many other lncRNAs in glioma have not been reported at all. Methods: In this study, we identified a novel lncRNA, UBE2R2-AS1, which was dramatically downregulated in glioma compared with normal tissue, by performing microarray detection of six pairs of glioma samples and adjacent normal tissues. In vitro experiments demonstrated that UBE2R2-AS1 regulated glioma cell proliferation, apoptosis, and migration. Results: UBE2R2-AS1 acted as a competing endogenous RNA (ceRNA) to target Toll-like receptor 4 (TLR4) mRNA by binding to miR-877-3p. Furthermore, lncRNA UBE2R2-AS1 suppressed glioblastoma cell growth, migration, and invasion, as well as promoting cell apoptosis by targeting miR-877-3p/TLR4 directly. Conclusion: This information regarding UBE2R2-AS1 and its glioma-related molecular mechanisms will aid the future identification of new lncRNA-directed diagnostics and drug-targeting therapies.

7.
Blood ; 128(16): 2017-2021, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27581360

ABSTRACT

The ataxia telangiectasia mutated (ATM)-interacting protein ATMIN mediates noncanonical ATM signaling in response to oxidative and replicative stress conditions. Like ATM, ATMIN can function as a tumor suppressor in the hematopoietic system: deletion of Atmin under the control of CD19-Cre results in B-cell lymphomas in aging mice. ATM signaling is essential for lymphopoiesis and hematopoietic stem cell (HSC) function; however, little is known about the role of ATMIN in hematopoiesis. We thus sought to investigate whether the absence of ATMIN would affect primitive hematopoietic cells in an ATM-dependent or -independent manner. Apart from its role in B-cell development, we show that ATMIN has an ATM-independent function in the common myeloid progenitors (CMPs) by deletion of Atmin in the entire hematopoietic system using Vav-Cre. Despite the lack of lymphoma formation, ATMIN-deficient mice developed chronic leukopenia as a result of high levels of apoptosis in B cells and CMPs and induced a compensatory mechanism in which HSCs displayed enhanced cycling. Consequently, ATMIN-deficient HSCs showed impaired regeneration ability with the induction of the DNA oxidative stress response, especially when aged. ATMIN, therefore, has multiple roles in different cell types, and its absence results in perturbed hematopoiesis, especially during stress conditions and aging.


Subject(s)
Aging , Apoptosis/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells , Oxidative Stress/genetics , Transcription Factors , Aging/genetics , Aging/metabolism , Aging/pathology , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Chronic Disease , Gene Deletion , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukopenia/genetics , Leukopenia/metabolism , Leukopenia/pathology , Mice , Mice, Knockout , Transcription Factors/genetics , Transcription Factors/metabolism
8.
J Cell Biol ; 198(1): 127-41, 2012 Jul 09.
Article in English | MEDLINE | ID: mdl-22753894

ABSTRACT

The AP-1 transcription factor c-Jun is a master regulator of the axonal response in neurons. c-Jun also functions as a negative regulator of myelination in Schwann cells (SCs) and is strongly reactivated in SCs upon axonal injury. We demonstrate here that, after injury, the absence of c-Jun specifically in SCs caused impaired axonal regeneration and severely increased neuronal cell death. c-Jun deficiency resulted in decreased expression of several neurotrophic factors, and GDNF and Artemin, both of which encode ligands for the Ret receptor tyrosine kinase, were identified as novel direct c-Jun target genes. Genetic inactivation of Ret specifically in neurons resulted in regeneration defects without affecting motoneuron survival and, conversely, administration of recombinant GDNF and Artemin protein substantially ameliorated impaired regeneration caused by c-Jun deficiency. These results reveal an unexpected function for c-Jun in SCs in response to axonal injury, and identify paracrine Ret signaling as an important mediator of c-Jun function in SCs during regeneration.


Subject(s)
Axons/physiology , Motor Neurons/physiology , Nerve Regeneration/physiology , Paracrine Communication/physiology , Proto-Oncogene Proteins c-jun/physiology , Schwann Cells/physiology , Animals , Cell Survival , Down-Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mice , Nerve Tissue Proteins/physiology
9.
J Neurochem ; 121(4): 607-18, 2012 May.
Article in English | MEDLINE | ID: mdl-22372722

ABSTRACT

Although neural c-Jun is essential for successful peripheral nerve regeneration, the cellular basis of this effect and the impact of c-Jun activation are incompletely understood. In the current study, we explored the effects of neuron-selective c-Jun deletion, substitution of serine 63 and 73 phosphoacceptor sites with non-phosphorylatable alanine, and deletion of Jun N-terminal kinases 1, 2 and 3 in mouse facial nerve regeneration. Removal of the floxed c-jun gene in facial motoneurons using cre recombinase under control of a neuron-specific synapsin promoter (junΔS) abolished basal and injury-induced neuronal c-Jun immunoreactivity, as well as most of the molecular responses following facial axotomy. Absence of neuronal Jun reduced the speed of axonal regeneration following crush, and prevented most cut axons from reconnecting to their target, significantly reducing functional recovery. Despite blocking cell death, this was associated with a large number of shrunken neurons. Finally, junΔS mutants also had diminished astrocyte and microglial activation and T-cell influx, suggesting that these non-neuronal responses depend on the release of Jun-dependent signals from neighboring injured motoneurons. The effects of substituting serine 63 and 73 phosphoacceptor sites (junAA), or of global deletion of individual kinases responsible for N-terminal c-Jun phosphorylation were mild. junAA mutants showed decrease in neuronal cell size, a moderate reduction in post-axotomy CD44 levels and slightly increased astrogliosis. Deletion of Jun N-terminal kinase (JNK)1 or JNK3 showed delayed functional recovery; deletion of JNK3 also interfered with T-cell influx, and reduced CD44 levels. Deletion of JNK2 had no effect. Thus, neuronal c-Jun is needed in regeneration, but JNK phosphorylation of the N-terminus mostly appears to not be required for its function.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Neurons/physiology , Proto-Oncogene Proteins c-jun/physiology , Animals , Atrophy , Axons/ultrastructure , Cell Death , Female , Hyaluronan Receptors/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/physiology , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/physiology , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/physiology , Motor Neurons/physiology , Nerve Regeneration/genetics , Neurons/ultrastructure , Phosphorylation , Point Mutation/physiology , Proto-Oncogene Proteins c-jun/genetics
10.
Dev Biol ; 358(1): 201-12, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21827743

ABSTRACT

The F-box protein Fbw7 (also known as Fbxw7, hCdc4 and Sel-10) functions as a substrate recognition component of a SCF-type E3 ubiquitin ligase. SCF(Fbw7) facilitates polyubiquitination and subsequent degradation of various proteins such as Notch, cyclin E, c-Myc and c-Jun. Fbw7 is highly expressed in the nervous system and controls neural stem cell differentiation and apoptosis via Notch and c-Jun during embryonic development (Hoeck et al., 2010). Fbw7 deletion in the neural lineage is perinatal lethal and thus prohibits studying the role of Fbw7 in the adult nervous system. fbw7 mRNA is highly expressed in the postnatal brain and to gain insights into the function of Fbw7 in postnatal neurogenesis we analysed Fbw7 function in the cerebellum. We generated conditional Fbw7-knockout mice (fbw7(∆Cb)) by inactivating Fbw7 specifically in the cerebellar anlage. This resulted in decreased cerebellar size, reduced Purkinje cell number and defects in axonal arborisation. Moreover, Fbw7-deficient cerebella showed supranumeral fissures and aberrant progenitor cell migration. Protein levels of the Fbw7 substrates Notch1 and N-terminally phosphorylated c-Jun were upregulated in fbw7(∆Cb) mice. Concomitant deletion of c-Jun, and also the junAA knock-in mutation which specifically abrogates c-Jun N-terminal phosphorylation, rescued Purkinje cell numbers and arborisation in the fbw7(∆Cb) background. Taken together these data demonstrate that Fbw7 is essential during cerebellar development, and identify N-terminally phosphorylated c-Jun as an important substrate of SCF(Fbw7) during neurogenesis.


Subject(s)
Cerebellum/embryology , F-Box Proteins/metabolism , Neurogenesis/physiology , Purkinje Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Southern , Cerebellum/metabolism , DNA Primers/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Immunohistochemistry , Mice , Mice, Knockout , Proto-Oncogene Proteins c-jun/metabolism , Purkinje Cells/cytology , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
11.
Mol Cell Biol ; 30(15): 3842-52, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20516211

ABSTRACT

In the nervous system, cell death by apoptosis plays a critical role during normal development and pathological neurodegeneration. Jun N-terminal kinases (JNKs) are essential regulators of neuronal apoptosis. The AP-1 transcription factor c-Jun is phosphorylated at multiple sites within its transactivation domain by the JNKs, and c-Jun phosphorylation is required for JNK-induced neurotoxicity. While the importance of c-Jun as a mediator of apoptotic JNK signaling in neurons is firmly established, the molecular mechanism underlying the requirement for c-Jun N-terminal phosphorylation is enigmatic. Here we identify the multifunctional protein Bag1-L as a coactivator of phosphorylated c-Jun. Bag1-L preferentially interacts with N-terminally phosphorylated c-Jun, and Bag1-L greatly augments transcriptional activation by phosphorylated c-Jun. Chromatin immunoprecipitation experiments revealed binding of Bag1-L to the promoters of proapoptotic AP-1 target genes, and overexpression of Bag1-L augmented cell death in primary neurons. Therefore, Bag1-L functions as a coactivator regulating neurotoxicity mediated by phosphorylated c-Jun.


Subject(s)
Apoptosis/physiology , Neurons/physiology , Animals , Apoptosis/genetics , Cell Death/genetics , DNA-Binding Proteins , MAP Kinase Signaling System/genetics , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , PC12 Cells , Phosphorylation , Rats , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors
12.
EMBO J ; 26(23): 4891-901, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-17972914

ABSTRACT

The ERK MAPK signalling pathway is a highly conserved kinase cascade linking transmembrane receptors to downstream effector mechanisms. To investigate the function of ERK in neurons, a constitutively active form of MEK1 (caMEK1) was conditionally expressed in the murine brain, which resulted in ERK activation and caused spontaneous epileptic seizures. ERK activation stimulated phosphorylation of eukaryotic translation initiation factor 4E (eIF4E) and augmented NMDA receptor 2B (NR2B) protein levels. Pharmacological inhibition of NR2B function impaired synaptic facilitation in area cornus ammonicus region 3 (CA3) in acute hippocampal slices derived from caMEK1-expressing mice and abrogated epilepsy in vivo. In addition, expression of caMEK1 caused phosphorylation of the transcription factor, cAMP response element-binding protein (CREB) and increased transcription of ephrinB2. EphrinB2 overexpression resulted in increased NR2B tyrosine phosphorylation, which was essential for caMEK1-induced epilepsy in vivo, since conditional inactivation of ephrinB2 greatly reduced seizure frequency in caMEK1 transgenic mice. Therefore, our study identifies a mechanism of epileptogenesis that links MAP kinase to Eph/Ephrin and NMDA receptor signalling.


Subject(s)
Epilepsy/etiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Animals , Cyclic AMP/metabolism , Enzyme Activation , Ephrin-B2/metabolism , Epilepsy/enzymology , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Mice , Mice, Transgenic , Models, Biological , Phosphorylation , Receptors, N-Methyl-D-Aspartate/metabolism , Transcription, Genetic
13.
Neuron ; 43(1): 57-67, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15233917

ABSTRACT

Nerve injury triggers numerous changes in the injured neurons and surrounding nonneuronal cells that ultimately result in successful target reinnervation or cell death. c-Jun is a component of the heterodimeric AP-1 transcription factor, and c-Jun is highly expressed in response to neuronal trauma. Here we have investigated the role of c-jun during axonal regeneration using mice lacking c-jun in the central nervous system. After transection of the facial nerve, the absence of c-Jun caused severe defects in several aspects of the axonal response, including perineuronal sprouting, lymphocyte recruitment, and microglial activation. c-Jun-deficient motorneurons were atrophic, resistant to axotomy-induced cell death, and showed reduced target muscle reinnervation. Expression of CD44, galanin, and alpha7beta1 integrin, molecules known to be involved in regeneration, was greatly impaired, suggesting a mechanism for c-Jun-mediated axonal growth. Taken together, our results identify c-Jun as an important regulator of axonal regeneration in the injured central nervous system.


Subject(s)
Facial Nerve Injuries/metabolism , Growth Cones/metabolism , Nerve Regeneration/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/metabolism , Animals , Atrophy/genetics , Atrophy/metabolism , Axotomy , Cell Death/genetics , Down-Regulation/genetics , Facial Nerve/cytology , Facial Nerve/growth & development , Facial Nerve/metabolism , Facial Nerve Injuries/genetics , Galanin/metabolism , Gliosis/genetics , Growth Cones/ultrastructure , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Integrins/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Microglia/cytology , Microglia/metabolism , Motor Neurons/cytology , Motor Neurons/metabolism , Muscle, Skeletal/innervation , Neuronal Plasticity/genetics , Proto-Oncogene Proteins c-jun/deficiency , Recovery of Function/genetics
14.
Science ; 303(5662): 1374-8, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14739463

ABSTRACT

Jun N-terminal kinases (JNKs) are essential for neuronal microtubule assembly and apoptosis. Phosphorylation of the activating protein 1 (AP1) transcription factor c-Jun, at multiple sites within its transactivation domain, is required for JNK-induced neurotoxicity. We report that in neurons the stability of c-Jun is regulated by the E3 ligase SCF(Fbw7), which ubiquitinates phosphorylated c-Jun and facilitates c-Jun degradation. Fbw7 depletion resulted in accumulation of phosphorylated c-Jun, stimulation of AP1 activity, and neuronal apoptosis. SCF(Fbw7) therefore antagonizes the apoptotic c-Jun-dependent effector arm of JNK signaling, allowing neurons to tolerate potentially neurotoxic JNK activity.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/physiology , Proto-Oncogene Proteins c-jun/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle Proteins/genetics , Cell Line , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Humans , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System , Mice , Molecular Sequence Data , PC12 Cells , Phosphorylation , RNA, Small Interfering/metabolism , Rats , Transcription Factor AP-1/metabolism , Transfection , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
15.
Clin Chim Acta ; 336(1-2): 39-44, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14500032

ABSTRACT

BACKGROUND: The cAMP response of porcine thyrocytes to the immunoglobulin (Ig) fraction from Graves' sera is increased if these fractions are prepared with higher than usual concentrations of polyethylene glycol (PEG; 22.5% rather than 13.5%), leading to the suggestion that additional factors might exist in serum which influence the ability of autoantibodies to stimulate thyroid cells. METHODS: We characterised the stimulatory activity of fractions prepared by differential PEG precipitation of Graves' sera, using heterologous eukaryotic cells expressing recombinant human thyrotrophin (TSH) receptor. RESULTS: We found no evidence that material soluble in 13.5% PEG but precipitated by 22.5% PEG was stimulatory either on its own or in combination with the immunoglobulin-containing material precipitated by 13.5% PEG. Indeed, the stimulatory effect was reproduced by simply including low concentrations of PEG (1-4%) in the diluted sera from the majority of Graves' patients (12/18 tested) added to the target cells. Intriguingly, PEG had no effect on basal levels, with normal sera or stimulation by thyrotrophin. CONCLUSIONS: The increase in stimulation reported when using immunoglobulins prepared with higher concentrations of PEG is attributable to a direct effect of the extra PEG (which had not been removed from the preparations) during the stimulation of the cells. Because the effect is observed with recombinant heterologous cells, it must be caused by interaction between PEG and the autoantibodies and/or the receptor-it cannot involve any other thyroid-specific molecule, although an involvement of other molecules widely distributed on many cell types is possible.


Subject(s)
Autoantibodies/immunology , Graves Disease/immunology , Receptors, Thyrotropin/immunology , Animals , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Humans , Polyethylene Glycols/pharmacology , Receptors, Thyrotropin/genetics , Recombinant Proteins/immunology , Stimulation, Chemical , Thyrotropin/metabolism
16.
Clin Endocrinol (Oxf) ; 59(4): 437-41, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14510905

ABSTRACT

OBJECTIVE: Identifying sites on the TSH-receptor that are involved in the pathological stimulation of the thyroid by autoantibodies in Graves' disease would aid the development of new therapies. We tested a series of monoclonal antibodies that recognize the native receptor for their ability to inhibit stimulation of the receptor in vitro. PATIENTS AND METHODS: Heterologous cells expressing the recombinant human TSH-receptor were stimulated with TSH or serum samples from 13 Graves' disease patients or the MRC Long-Acting Thyroid Stimulator standard B (LATS-B) and their cAMP responses measured. The effect on this stimulation of various doses of purified monoclonal antibodies with defined epitopes was determined. RESULTS: Antibodies against one epitope (residues 381-384) inhibited TSH-stimulated cyclic adenosine monophosphate (cAMP) production (1 microg/ml causing 50% inhibition of the response to 100 microU/ml TSH) and also inhibited cAMP production induced by sera from approximately 40% (6/14) of Graves' disease patients, including the MRC LATS-B standard. CONCLUSIONS: Residues 381-384 of the human TSH-receptor are important in the physiological and pathological stimulation of the thyroid. This opens the possibility of more specific therapy of some Graves' disease patients by agents directed against this epitope.


Subject(s)
Graves Disease/immunology , Receptors, Thyrotropin/immunology , Animals , Antibodies, Monoclonal , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Cyclic AMP/immunology , Epitopes/immunology , Graves Disease/drug therapy , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...