Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
ACS Appl Mater Interfaces ; 16(17): 21383-21399, 2024 May 01.
Article En | MEDLINE | ID: mdl-38626424

Osteoarthritis (OA) progression is highly associated with chondrocyte mitochondrial dysfunction and disorders of catabolism and anabolism of the extracellular matrix (ECM) in the articular cartilage. The mitochondrial unfolded protein response (UPRmt), which is an integral component of the mitochondrial quality control (MQC) system, is essential for maintaining chondrocyte homeostasis. We successfully validated the pivotal role of activating transcription factor 5 (ATF5) in upregulating the UPRmt, mitigating IL-1ß-induced inflammation and mitochondrial dysfunction, and promoting balanced metabolism in articular cartilage ECM, proving its potential as a promising therapeutic target for OA. Modified mRNAs (modRNAs) have emerged as novel and efficient gene delivery vectors for nucleic acid therapeutic approaches. In this study, we combined Atf5-modRNA (modAtf5) with engineered exosomes derived from bone mesenchymal stem cells (ExmodAtf5) to exert cytoprotective effects on chondrocytes in articular cartilage via Atf5. However, the rapid localized metabolization of ExmodAtf5 limits its application. PLGA-PEG-PLGA (Gel), an injectable thermosensitive hydrogel, was used as a carrier of ExmodAtf5 (Gel@ExmodAtf5) to achieve a sustained release of ExmodAtf5. In vitro and in vivo, the use of Gel@ExmodAtf5 was shown to be a highly effective strategy for OA treatment. The in vivo therapeutic effect of Gel@ExmodAtf5 was evidenced by the preservation of the intact cartilage surface, low OARSI scores, fewer osteophytes, and mild subchondral bone sclerosis and cystic degeneration. Consequently, the combination of ExmodAtf5 and PLGA-PEG-PLGA could significantly enhance the therapeutic efficacy and prolong the exosome release. In addition, the mitochondrial protease ClpP enhanced chondrocyte autophagy by modulating the mTOR/Ulk1 pathway. As a result of our research, Gel@ExmodAtf5 can be considered to be effective at alleviating the progression of OA.


Activating Transcription Factors , Chondrocytes , Exosomes , Mitochondria , Osteoarthritis , RNA, Messenger , Unfolded Protein Response , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/therapy , Exosomes/metabolism , Exosomes/chemistry , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Unfolded Protein Response/drug effects , Activating Transcription Factors/metabolism , Activating Transcription Factors/chemistry , Activating Transcription Factors/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Chondrocytes/metabolism , Chondrocytes/drug effects , Humans , Mesenchymal Stem Cells/metabolism , Hydrogels/chemistry , Male , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/drug effects
2.
J Biol Chem ; 293(8): 2939-2948, 2018 02 23.
Article En | MEDLINE | ID: mdl-29326161

Activating transcription factor 5 (ATF5) is a member of the ATF/cAMP response element-binding protein family of transcription factors. ATF5 regulates stress responses and cell survival, proliferation, and differentiation and also plays a role in viral infections, cancer, diabetes, schizophrenia, and the olfactory system. Moreover, it was found to also have a critical cell cycle-dependent structural function at the centrosome. However, the mechanism that controls the localization of ATF5 at the centrosome is unclear. Here we report that ATF5 is small ubiquitin-like modifier (SUMO) 2/3-modified at a conserved SUMO-targeting consensus site in various types of mammalian cells. We found that SUMOylation of ATF5 is elevated in the G1 phase of the cell cycle and diminished in the G2/M phase. ATF5 SUMOylation disrupted the interaction of ATF5 with several centrosomal proteins and dislodged ATF5 from the centrosome at the end of the M phase. Of note, blockade of ATF5 SUMOylation deregulated the centrosome cycle, impeded ATF5 translocation from the centrosome, and caused genomic instability and G2/M arrest in HeLa cells. Our results indicate that ATF5 SUMOylation is an essential mechanism that regulates ATF5 localization and function at the centrosome.


Activating Transcription Factors/metabolism , Centrosome/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitins/metabolism , Activating Transcription Factors/chemistry , Activating Transcription Factors/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Centrosome/enzymology , Consensus Sequence , Conserved Sequence , Gene Deletion , Genomic Instability , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Transport , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/antagonists & inhibitors , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/genetics , Ubiquitins/antagonists & inhibitors , Ubiquitins/chemistry , Ubiquitins/genetics
3.
Clin Cancer Res ; 22(18): 4698-711, 2016 Sep 15.
Article En | MEDLINE | ID: mdl-27126996

PURPOSE: Despite significant progress in cancer research, many tumor entities still have an unfavorable prognosis. Activating transcription factor 5 (ATF5) is upregulated in various malignancies and promotes apoptotic resistance. We evaluated the efficacy and mechanisms of the first described synthetic cell-penetrating inhibitor of ATF5 function, CP-d/n-ATF5-S1. EXPERIMENTAL DESIGN: Preclinical drug testing was performed in various treatment-resistant cancer cells and in vivo xenograft models. RESULTS: CP-d/n-ATF5-S1 reduced the transcript levels of several known direct ATF5 targets. It depleted endogenous ATF5 and induced apoptosis across a broad panel of treatment-refractory cancer cell lines, sparing non-neoplastic cells. CP-d/n-ATF5-S1 promoted tumor cell apoptotic susceptibility in part by reducing expression of the deubiquitinase Usp9X and led to diminished levels of antiapoptotic Bcl-2 family members Mcl-1 and Bcl-2. In line with this, CP-d/n-ATF5-S1 synergistically enhanced tumor cell apoptosis induced by the BH3-mimetic ABT263 and the death ligand TRAIL. In vivo, CP-d/n-ATF5-S1 attenuated tumor growth as a single compound in glioblastoma, melanoma, prostate cancer, and triple receptor-negative breast cancer xenograft models. Finally, the combination treatment of CP-d/n-ATF5-S1 and ABT263 significantly reduced tumor growth in vivo more efficiently than each reagent on its own. CONCLUSIONS: Our data support the idea that CP-d/n-ATF5-S1, administered as a single reagent or in combination with other drugs, holds promise as an innovative, safe, and efficient antineoplastic agent against treatment-resistant cancers. Clin Cancer Res; 22(18); 4698-711. ©2016 AACR.


Activating Transcription Factors/chemistry , Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Drug Resistance, Neoplasm/drug effects , Peptides/pharmacology , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell-Penetrating Peptides/chemical synthesis , Disease Models, Animal , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Peptides/chemical synthesis , Sulfonamides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Methods Mol Biol ; 1372: 49-59, 2016.
Article En | MEDLINE | ID: mdl-26530914

Molecular therapy using small interfering RNA (siRNA) shows great promise in the development of novel therapeutics for cancer. Although various approaches have been developed for in vivo delivery of siRNAs into tumors, stability of siRNA in blood circulation, and low efficiency of siRNA delivery into tumor cells are the major obstacles for further translation into cancer therapeutics. In this protocol, we describe methods of the production of shRNA expressing DNA nanocassettes by PCR amplification of double-stranded DNA fragments containing a U6 promoter and a shRNA gene. Those DNA nanocassettes can be conjugated to the polymer coating of nanoparticles that are targeted to cellular receptors highly expressed in tumor cells, such as urokinase plasminogen activator receptor (uPAR), for targeted delivery and receptor mediated internalization of shRNA expressing DNA nanocassettes. Methods for in vitro and in vivo evaluation of target specificity and gene-knockdown effect are also provided.


Gene Expression , Gene Transfer Techniques , Molecular Imaging/methods , Nanoparticles , Neoplasms/genetics , RNA, Small Interfering/genetics , Activating Transcription Factors/chemistry , Activating Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cloning, Molecular , Disease Models, Animal , Gene Silencing , Humans , Luminescent Measurements/methods , Mice , Nanoparticles/chemistry , Neoplasms/diagnosis , Neoplasms/therapy , Plasmids/administration & dosage , Plasmids/chemistry , Plasmids/genetics , RNA Interference , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Xenograft Model Antitumor Assays
5.
Fish Shellfish Immunol ; 54: 144-52, 2016 Jul.
Article En | MEDLINE | ID: mdl-26481519

A mitochondrial specific stress response termed mitochondrial unfolded protein response (UPR(mt)) is activated in responding to disturbance of protein homeostasis in mitochondria. The activating transcription factor associated with stress-1 (designated as ATFS-1) is the key regulator of UPR(mt). To investigating the roles of ATFS-1 (LvATFS-1) in Litopenaeus vannamei mitochondrial stress remission and immunity, it's full length cDNA was cloned. The open reading frame of LvATFS-1 was 1, 557 bp in length, deducing to a 268 amino acids protein. LvATFS-1 was highly expressed in muscle, hemocytes and eyestalk. Subcellular location assays showed that N-terminal of LvATFS-1 contained a mitochondrial targeting sequence, which could directed the fused EGFP located to mitochondria. And the C-terminal of LvATFS-1, which had a nuclear localization signal, expressed in nucleus. The in vitro experiments verified that LvATFS-1 could reduced the level of intracellular reactive oxygen species (ROS). And results of real-time RT-PCR indicated that LvATFS-1 might scavenge excess ROS via ROS-eliminating genes regulation. Reporter gene assays showed that LvATFS-1 could upregulated the expression of the antimicrobial peptide genes in Drosophila Schneider 2 cells. Results of real-time RT-PCR showed that Vibrio alginolyticus or white spot syndrome virus (WSSV) infection induced the expression of LvATFS-1. And knocked-down LvATFS-1 by RNAi resulted in a higher cumulative mortality of L. vannamei upon V. alginolyticus or WSSV infection. These results suggested that LvATFS-1 not only rolled in mitochondrial specific stress responding, but also important for L. vannamei immunologic defence.


Activating Transcription Factors/genetics , Penaeidae/physiology , Activating Transcription Factors/chemistry , Activating Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Regulation , Organ Specificity , Penaeidae/genetics , Penaeidae/immunology , Penaeidae/microbiology , Reactive Oxygen Species/metabolism , Unfolded Protein Response , Vibrio alginolyticus/physiology , White spot syndrome virus 1/physiology
6.
PLoS One ; 10(4): e0122980, 2015.
Article En | MEDLINE | ID: mdl-25849628

Evidence suggests that stimulating apoptosis in malignant cells without inflicting collateral damage to the host's normal tissues is a promising cancer therapy. Chemo- and radiation therapies that, especially if combined, induce apoptosis in tumor cells have been used for treating cancer patients for decades. These treatments, however, are limited in their ability to discriminate between malignant and non-malignant cells and, therefore, produce substantial healthy tissue damage and subsequent toxic side-effects. In addition, as a result of these therapies, many tumor types acquire an apoptosis-resistant phenotype and become more aggressive and metastatic. Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL) has been considered a promising and reliable selective inducer of apoptosis in cancerous cells. TRAIL, however, is not uniformly effective in cancer and multiple cancer cell types are considered resistant to natural TRAIL. To overcome this deficiency of TRAIL, we have earlier constructed a yeast-human hybrid leucine zipper-TRAIL in which the yeast GCN4-pII leucine zipper was fused to human TRAIL (GCN4-TRAIL). This construct exhibited a significantly improved anti-tumor apoptotic activity and safety, but is potentially immunogenic in humans. Here, we report a novel, potent, and fully human ATF7 leucine zipper-TRAIL (ATF7-TRAIL) fusion construct that is expected to have substantially lower immunogenicity. In solution, ATF7-TRAIL exists solely as a trimer with a Tm of 80°C and is active against cancer cells both in vitro and in vivo, in a mouse tumor xenograft model. Our data suggest that our re-engineered TRAIL is a promising candidate for further evaluation as an antitumor agent.


Apoptosis/drug effects , Leucine Zippers/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Activating Transcription Factors/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Female , Humans , Mice , Models, Molecular , Protein Multimerization , Protein Structure, Quaternary , Recombinant Fusion Proteins/chemistry , TNF-Related Apoptosis-Inducing Ligand/chemistry , Xenograft Model Antitumor Assays
7.
J Inorg Biochem ; 148: 2-10, 2015 Jul.
Article En | MEDLINE | ID: mdl-25798989

We report the development of rhenium(I) polypyridine complexes appended with a dibenzocyclooctyne (DIBO) moiety as bioorthogonal probes for azide-modified biomolecules. Three phosphorescent rhenium(I) polypyridine DIBO complexes [Re(N^N)(CO)3(py-C6-DIBO)][CF3SO3] (py-C6-DIBO=3-(N-(6-(3,4:7,8-dibenzocyclooctyne-5-oxycarbonylamino)hexyl)aminocarbonyl)pyridine; N^N=1,10-phenanthroline (phen) (1a), 3,4,7,8-tetramethyl-1,10-phenanthroline (Me4-phen) (2a), 4,7-diphenyl-1,10-phenanthroline (Ph2-phen) (3a)) and their DIBO-free counterparts [Re(N^N)(CO)3(py-C6-BOC)][CF3SO3] (py-C6-BOC=3-(N-(6-(tert-butoxycarbonylamino)hexyl)aminocarbonyl)pyridine; N^N=phen (1b), Me4-phen (2b), Ph2-phen (3b)) were synthesized and characterized. Upon photoexcitation, all the complexes displayed intense and long-lived yellow triplet metal-to-ligand charge-transfer ((3)MLCT) (dπ(Re)→π*(N^N)) emission. The DIBO complexes underwent facile reactions with benzyl azide in methanol at 298 K with second-order rate constants (k2) in the range of 0.077 to 0.091 M(-1) s(-1). As revealed from SDS-PAGE analysis, the DIBO complexes can selectively label azide-modified proteins and the resulting bioconjugates displayed strong phosphorescence upon photoexcitation. Results of inductively coupled plasma mass spectrometry (ICP-MS) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays indicated that the DIBO complexes accumulated in Chinese Hamster Ovary (CHO) cells with considerable cytotoxic activity. Upon incubation of CHO cells with these complexes, relatively weak intracellular emission was observed. In contrast, upon pretreatment of the cells with 1,3,4,6-tetra-O-acetyl-N-azidoacetyl-D-mannosamine (Ac4ManNAz), intense emission was observed from the cell membrane and some internal compartments. The results suggest that the DIBO complexes are promising candidates for imaging azide-labeled biomolecules.


Luminescence , Molecular Probes/chemistry , Organometallic Compounds/chemistry , Rhenium/chemistry , Activating Transcription Factors/chemistry , Activating Transcription Factors/metabolism , Activating Transcription Factors/pharmacology , Animals , CHO Cells , Cattle , Cell Survival/drug effects , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel , Humans , Inhibitory Concentration 50 , Microscopy, Confocal , Models, Chemical , Molecular Probes/chemical synthesis , Molecular Probes/metabolism , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Protein Binding , Rhenium/metabolism , Serum Albumin/chemistry , Serum Albumin/metabolism , Serum Albumin/pharmacology , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Serum Albumin, Bovine/pharmacology
8.
PLoS One ; 9(12): e116048, 2014.
Article En | MEDLINE | ID: mdl-25545367

Activating transcription factor 2 (ATF2) and its homolog ATF7 are phosphorylated at Thr-69/Thr-71 and at Thr-51/Thr-53, respectively, by stress-activated MAPKs regulating their transcriptional functions in G1 and S phases. However, little is known about the role of ATF2 and ATF7 in G2/M phase. Here, we show that Cdk1-cyclin B1 phosphorylates ATF2 at Thr-69/Thr-71 and ATF7 at Thr-51/Thr-53 from early prophase to anaphase in the absence of any stress stimulation. Knockdown of ATF2 or ATF7 decreases the rate of cell proliferation and the number of cells in M-phase. In particular, the knockdown of ATF7 severely inhibits cell proliferation and G2/M progression. The inducible expression of a mitotically nonphosphorylatable version of ATF7 inhibits G2/M progression despite the presence of endogenous ATF7. We also show that mitotic phosphorylation of ATF7 promotes the activation of Aurora kinases, which are key enzymes for early mitotic events. These results suggest that the Cdk1-mediated phosphorylation of ATF7 facilitates G2/M progression, at least in part, by enabling Aurora signaling.


Activating Transcription Factors/metabolism , CDC2 Protein Kinase/metabolism , Cell Division , Phosphothreonine/metabolism , Activating Transcription Factor 2/chemistry , Activating Transcription Factor 2/metabolism , Activating Transcription Factors/chemistry , Amino Acid Sequence , Anaphase , Aurora Kinases/metabolism , G2 Phase , Gene Knockdown Techniques , HeLa Cells , Humans , Models, Biological , Molecular Sequence Data , Phosphorylation , Prophase
9.
Sci China Life Sci ; 56(12): 1124-33, 2013 Dec.
Article En | MEDLINE | ID: mdl-24302293

Studies of herpes simplex virus type 1 (HSV-1) infection have shown that many known and unknown cellular molecules involved in viral proliferation are up-regulated following HSV-1 infection. In this study, using two-dimensional polyacrylamide gel electrophoresis, we found that the expression of the HSV-1 infection response repressive protein (HIRRP, GI 16552881) was up-regulated in human L02 cells infected with HSV-1. HIRRP, an unknown protein, was initially localized in the cytoplasm and then translocated into the nucleus of HSV-1-infected cells. Further analysis showed that HIRRP represses HSV-1 proliferation by inhibiting transcription of the viral genome by interacting with the cellular transcription factor, ATF5, via its N-terminal domain. ATF5 represses the transcription of many host genes but can also act as an activator of genes containing a specific motif. We found that ATF5 promotes the proliferation of HSV-1 via a potential mechanism by which ATF5 enhances the transcription of viral genes during the course of an HSV-1 infection; HIRRP then induces feedback repression of this transcription by interacting with ATF5.


Carrier Proteins/physiology , Herpesvirus 1, Human/physiology , Viral Proteins/physiology , Activating Transcription Factors/chemistry , Activating Transcription Factors/genetics , Activating Transcription Factors/physiology , Animals , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytoplasm/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Electrophoresis, Gel, Two-Dimensional , Gene Knockdown Techniques , Genome, Viral , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Molecular Sequence Data , Protein Interaction Domains and Motifs , Up-Regulation , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/genetics , Virus Replication/physiology
10.
Hum Mol Genet ; 22(16): 3187-94, 2013 Aug 15.
Article En | MEDLINE | ID: mdl-23591993

How the transcription repressing complex Polycomb interacts with transcriptional regulators at housekeeping genes in somatic cells is not well understood. By exploiting a CpG island (CGI) point mutation causing a Mendelian disease, we show that DNA binding of activating transcription factor (TF) determines histone acetylation and nucleosomal depletion commensurate with Polycomb exclusion from the target promoter. Lack of TF binding leads to reversible transcriptional repression imposed by nucleosomal compaction and consolidated by Polycomb recruitment and establishment of bivalent chromatin status. Thus, within a functional hierarchy of transcriptional regulators, TF binding is the main determinant of Polycomb recruitment to the CGI of a housekeeping gene in somatic cells.


Activating Transcription Factors/chemistry , B-Lymphocytes/chemistry , B-Lymphocytes/metabolism , CpG Islands/genetics , Glycosylphosphatidylinositols/deficiency , Hemoglobinuria, Paroxysmal/genetics , Mannosyltransferases/genetics , Polycomb-Group Proteins/metabolism , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Base Sequence , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Essential , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Hemoglobinuria, Paroxysmal/metabolism , Histones/chemistry , Histones/metabolism , Humans , Mannosyltransferases/metabolism , Molecular Sequence Data , Nucleosomes/metabolism , Point Mutation , Polycomb-Group Proteins/chemistry , Polycomb-Group Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Seizures
11.
Mol Pharm ; 9(11): 3190-9, 2012 Nov 05.
Article En | MEDLINE | ID: mdl-23067245

Protein aggregation is a major problem for biopharmaceuticals. While the control of aggregation is critically important for the future of protein pharmaceuticals, mechanisms of aggregate assembly, particularly the role that structure plays, are still poorly understood. Increasing evidence indicates that partially folded intermediates critically influence the aggregation pathway. We have previously reported the use of the basic leucine zipper (bZIP) domain of activating transcription factor 5 (ATF5) as a partially folded model system to investigate protein aggregation. This domain contains three regions with differing structural propensity: a N-terminal polybasic region, a central helical leucine zipper region, and a C-terminal extended valine zipper region. Additionally, a centrally positioned cysteine residue readily forms an intermolecular disulfide bond that reduces aggregation. Computational analysis of ATF5 predicts that the valine zipper region facilitates self-association. Here we test this hypothesis using a truncated mutant lacking the C-terminal valine zipper region. We compare the structure and aggregation of this mutant to the wild-type (WT) form under both reducing and nonreducing conditions. Our data indicate that removal of this region results in a loss of α-helical structure in the leucine zipper and a change in the mechanism of self-association. The mutant form displays increased association at low temperature but improved resistance to thermally induced aggregation.


Activating Transcription Factors/chemistry , Activating Transcription Factors/metabolism , Protein Multimerization , Valine/chemistry , Activating Transcription Factors/genetics , Benzothiazoles , Circular Dichroism , Fluorescence , Humans , Leucine Zippers , Mutation/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Protein Structure, Tertiary , Spectroscopy, Fourier Transform Infrared , Thiazoles/chemistry
12.
EMBO J ; 29(1): 196-208, 2010 Jan 06.
Article En | MEDLINE | ID: mdl-19893493

Many symptoms induced by isolation rearing of rodents may be relevant to neuropsychiatric disorders, including depression. However, identities of transcription factors that regulate gene expression in response to chronic social isolation stress remain elusive. The transcription factor ATF-7 is structurally related to ATF-2, which is activated by various stresses, including inflammatory cytokines. Here, we report that Atf-7-deficient mice exhibit abnormal behaviours and increased 5-HT receptor 5B (Htr5b) mRNA levels in the dorsal raphe nuclei. ATF-7 silences the transcription of Htr5B by directly binding to its 5'-regulatory region, and mediates histone H3-K9 trimethylation via interaction with the ESET histone methyltransferase. Isolation-reared wild-type (WT) mice exhibit abnormal behaviours that resemble those of Atf-7-deficient mice. Upon social isolation stress, ATF-7 in the dorsal raphe nucleus is phosphorylated via p38 and is released from the Htr5b promoter, leading to the upregulation of Htr5b. Thus, ATF-7 may have a critical role in gene expression induced by social isolation stress.


Activating Transcription Factors/metabolism , Receptors, Serotonin/genetics , Social Isolation , Activating Transcription Factor 2/metabolism , Activating Transcription Factors/chemistry , Activating Transcription Factors/deficiency , Activating Transcription Factors/genetics , Animals , Base Sequence , Gene Expression , Gene Silencing , Histones/metabolism , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raphe Nuclei/metabolism , Social Behavior , Stress, Psychological
13.
Fungal Genet Biol ; 46(12): 887-97, 2009 Dec.
Article En | MEDLINE | ID: mdl-19770065

We compared atfA and atfB, the genes encoding the respective ATF/CREB-type transcription factors in Aspergillus oryzae. The germination ratio of DeltaatfA conidia was low without any stress, unlike that of DeltaatfB conidia. The DeltaatfA conidia were more sensitive to oxidative stress than the DeltaatfB conidia, which are also sensitive to oxidative stress. We compared the gene expressions of these strains by using a DNA microarray, GeneChip. Almost all the genes regulated by atfB were also regulated by atfA, but atfA also regulated many genes that were not regulated by atfB, including some genes putatively involved in oxidative stress resistance. The level of glutamate, the major amino acid in A. oryzae conidia, was significantly low only in the DeltaatfA conidia, and the glycerol accumulation during germination was not observed only in the DeltaatfA strain. We therefore concluded that atfA is involved in germination via carbon and nitrogen source metabolism.


Activating Transcription Factors/genetics , Aspergillus oryzae/physiology , Fungal Proteins/genetics , Spores, Fungal/physiology , Activating Transcription Factors/chemistry , Activating Transcription Factors/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Aspergillus oryzae/genetics , Catalase/genetics , Catalase/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Fungal , Genes, Fungal , Glycerol/metabolism , Molecular Sequence Data , Mutation , Oligonucleotide Array Sequence Analysis , Oxidative Stress , Promoter Regions, Genetic , Sequence Alignment , Spores, Fungal/genetics , Stress, Physiological , Trehalose/metabolism
14.
Mol Pharm ; 6(4): 1205-15, 2009.
Article En | MEDLINE | ID: mdl-19435374

Amorphous aggregation is a major problem for protein biopharmaceuticals, and aggregate formation in a drug formulation can have serious health implications for the patient. In many cases, an immunogenic response is generated from the administration of a drug product containing aggregated protein. This becomes especially significant when the patient requires long-term or repeated administration of the drug, because the likelihood of a severe immune response increases. While the prevention of protein aggregation is critically important for the future of protein pharmaceuticals, the mechanism of amorphous aggregation is still poorly understood. The lack of understanding regarding nonfibrillar aggregation is largely due to the fact that assembly is difficult to study. In particular the role that various structural features (i.e., alpha-helix, beta-structure, disulfide bonds) play in the aggregation process varies with the amino acid sequence and is dependent upon tertiary structure and solution conditions. Well-structured proteins do not readily aggregate in solution, whereas partially unfolded proteins tend to aggregate rapidly and often become insoluble. Here, we present a unique and simple system for studying amorphous protein aggregation. We have previously reported the isolation of the basic leucine zipper (bZIP) domain of activating transcription factor 5 (ATF5), a protein notable for its potential as a pharmaceutical target for treatment of glioblastoma multiforme. This domain consists of a single alpha-helix and possesses a single cysteine residue. It is only partially structured and displays marginal stability in solution under physiological conditions. We have modulated solution conditions that affect backbone solubility and the oxidation state of the thiol to successfully investigate the role that alpha-helical structure and disulfide bond formation play in protein stability. Our data indicate that covalent cross-linking helps to retain ATF5's helicity, which inhibits the formation of large aggregates. These studies have led to the identification of stabilizing conditions for ATF5, which will enable further study of the protein as a pharmaceutical target. Moreover, this work has general implications for analyzing stability of helical proteins in vitro as well as the specific atomic-level interactions in ATF5 that contribute to instability and self-association.


Activating Transcription Factors/chemistry , Activating Transcription Factors/metabolism , Disulfides/chemistry , Protein Folding , Protein Multimerization , Circular Dichroism , Humans , Protein Conformation , Static Electricity
15.
Protein Expr Purif ; 62(2): 235-43, 2008 Dec.
Article En | MEDLINE | ID: mdl-18718539

Activating transcription factor 5 (ATF5) recently has been demonstrated to play a critical role in promoting the survival of human glioblastoma cells. Interference with the function of ATF5 in an in vivo rat model caused glioma cell death in primary tumors but did not affect the status of normal cells surrounding the tumor, suggesting ATF5 may prove an ideal target for anti-cancer therapy. In order to examine ATF5 as a pharmaceutical target, the protein must be produced and purified to sufficient quantity to begin analyses. Here, a procedure for expressing and refolding the bZIP domain of ATF5 in sufficient yield and final concentration to permit assay development and structural characterization of this target using solution NMR is reported. Two-dimensional NMR and circular dichroism analyses indicate the protein exists in the partially alpha-helical, monomeric x-form conformation with only a small fraction of ATF5 participating in formation of higher-order structure, presumably coiled-coil homodimerization. Despite the persistence of monomers in solution even at high concentration, an electrophoretic mobility shift assay showed that ATF5 is able to bind to the cAMP response element (CRE) DNA motif. Polyacrylamide gel electrophoresis and mass spectrometry were used to confirm that ATF5 can participate in homodimer formation and that this dimerization is mediated by disulfide bond formation.


Activating Transcription Factors/chemistry , Activating Transcription Factors/metabolism , Escherichia coli/metabolism , Leucine Zippers , Protein Folding , Activating Transcription Factors/genetics , Activating Transcription Factors/isolation & purification , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/isolation & purification , Glutathione Transferase/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Response Elements/genetics
16.
Nucleic Acids Res ; 36(9): 2838-51, 2008 May.
Article En | MEDLINE | ID: mdl-18375981

The Atf1 protein of Schizosaccharomyces pombe contains a bZIP (DNA-binding/protein dimerization) domain characteristic of ATF/CREB proteins, but no other functional domains or clear homologs have been reported. Atf1-containing, bZIP protein dimers bind to CRE-like DNA sites, regulate numerous stress responses, and activate meiotic recombination at hotspots like ade6-M26. We defined systematically the organization of Atf1 and its heterodimer partner Pcr1, which is required for a subset of Atf1-dependent functions. Surprisingly, only the bZIP domain of Pcr1 is required for hotspot activity and tethering of Atf1 to ade6 promotes recombination in the absence of its bZIP domain and the Pcr1 protein. Therefore the recombination-activation domain of Atf1-Pcr1 heterodimer resides exclusively in Atf1, and Pcr1 confers DNA-binding site specificity in vivo. Atf1 has a modular organization in which distinct regions affect differentially the osmotic stress response (OSA) and meiotic recombination (HRA, HRR). The HRA and HRR regions are necessary and sufficient to activate and repress recombination, respectively. Moreover, Atf1 defines a family of conserved proteins with discrete sequence motifs in the functional domains (OSA, HRA, HRR, bZIP). These findings reveal the functional organization of Atf1 and Pcr1, and illustrate several mechanisms by which bZIP proteins can regulate multiple, seemingly disparate activities.


Activating Transcription Factor 1/chemistry , Activating Transcription Factors/chemistry , Phosphoproteins/chemistry , Recombination, Genetic , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/genetics , Activating Transcription Factor 1/genetics , Activating Transcription Factor 1/metabolism , Activating Transcription Factors/genetics , Activating Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Chromosomes, Fungal , Conserved Sequence , Dimerization , Meiosis , Molecular Sequence Data , Mutagenesis , Osmotic Pressure , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Structure, Tertiary , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Sequence Homology, Amino Acid
...