Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Neurotox Res ; 40(6): 2016-2026, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36550222

ABSTRACT

Acrylamide (AM) is a potent neurotoxin and carcinogen that is mainly formed by the Maillard reaction of asparagine with starch at high temperatures. However, the toxicity mechanism underlying AM has not been investigated from a proteomic perspective, and the regulation of protein expression by AM remains poorly understood. This research was the first to utilize proteomics to explore the mechanism of AM exposure-induced neuroinflammation. Target proteins were obtained by differential protein analysis, functional annotation, and enrichment analysis of proteomics. Then, molecular biology methods, including Western blot, qPCR, and immunofluorescence, were used to verify the results and explore possible mechanisms. We identified 100 key differential metabolites by proteomic analysis, which was involved in the occurrence of various biological functions. Among them, the KEGG pathway enrichment analysis showed that the differential proteins were enriched in the P53 pathway, sulfur metabolism pathway, and ferroptosis. Finally, the differential target protein we locked was LARP7. Molecular biological verification found that AM exposure inhibited the expression of LARP7 and induced the burst of inflammation, while SRT1720 agonist treatment showed no effect on LARP7, but significant changes in inflammatory factors and NF-κB. Taken together, these findings suggested that AM may activate NF-κB to induce neuroinflammation by inhibiting the LARP7-SIRT1 pathway. And our study provided a direction for AM-induced neurotoxicity through proteomics and multiple biological analysis methods.


Subject(s)
NF-kappa B , Sirtuin 1 , Humans , NF-kappa B/metabolism , Sirtuin 1/metabolism , Neuroinflammatory Diseases , Microglia , Acrylamide/toxicity , Acrylamide/metabolism , Proteomics , Ribonucleoproteins/metabolism , Ribonucleoproteins/pharmacology
2.
Oxid Med Cell Longev ; 2021: 9360339, 2021.
Article in English | MEDLINE | ID: mdl-34504644

ABSTRACT

Although chronic intermittent hypoxia- (IH-) induced myocardial apoptosis is an established pathophysiological process resulting in a poor prognosis for patients with obstructive sleep apnea syndrome, its underlying mechanism remains unclear. This study is aimed at exploring the role of makorin ring finger protein 1 (MKRN1) in IH-induced myocardial apoptosis and elucidating its molecular activity. First, the GSE2271 dataset was downloaded from the Gene Expression Omnibus database to identify the differentially expressed genes. Then, an SD rat model of IH, together with rat cardiomyocyte H9C2 and human cardiomyocyte AC16 IH models, was constructed. TUNEL, Western blot, and immunohistochemistry assays were used to detect cell apoptosis. Dihydroethidium staining was conducted to analyze the concentration of reactive oxygen species. In addition, RT-qPCR, Western blot, and immunohistochemistry were performed to measure the expression levels of MKRN1 and p21. The direct interaction between MKRN1 and p21 was determined using coimmunoprecipitation and ubiquitination analysis. MKRN1 expression was found to be downregulated in IH rat myocardial tissues as well as in H9C2 and AC16 cells. Upregulated expression of MKRN1 in H9C2 and AC16 cells alleviated the IH-induced reactive oxygen species production and cell apoptosis. Mechanistically, MKRN1 promoted p21 protein ubiquitination and the proteasome pathway degradation to negatively regulate p21 expression. Thus, MKRN1 regulates p21 ubiquitination to prevent IH-induced myocardial apoptosis.


Subject(s)
Nerve Tissue Proteins/pharmacology , Ribonucleoproteins/pharmacology , rho GTP-Binding Proteins/metabolism , Animals , Apoptosis/physiology , Cell Hypoxia/physiology , Humans , Mice , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Ubiquitination
3.
ACS Appl Mater Interfaces ; 12(19): 21386-21397, 2020 May 13.
Article in English | MEDLINE | ID: mdl-32315156

ABSTRACT

A preassembled Cas9/single-guide RNA complex (Cas9 ribonucleoprotein; Cas9 RNP) induces genome editing efficiently, with small off-target effects compared with the conventional techniques, such as plasmid DNA and mRNA systems. However, penetration of Cas9 RNP through the cell membrane is low. In particular, the incorporation of Cas9 RNP into neurons and the brain is challenging. In the present study, we have reported the use of a dendrimer (generation 3; G3)/glucuronylglucosyl-ß-cyclodextrin conjugate (GUG-ß-CDE (G3)) as a carrier of Cas9 RNP and evaluated genome editing activity in the neuron and the brain. A Cas9 RNP ternary complex with GUG-ß-CDE (G3) was prepared by only mixing the components. The resulting complex exhibited higher genome editing activity than the complex with the dendrimer (G3), Lipofectamine 3000 or Lipofectamine CRISPRMAX in SH-SY5Y cells, a human neuroblastoma cell line. In addition, GUG-ß-CDE (G3) enhanced the genome editing activity of Cas9 RNP in the whole mouse brain after a single intraventricular administration. Thus, GUG-ß-CDE (G3) is a useful Cas9 RNP carrier that can induce genome editing in the neuron and brain.


Subject(s)
Brain/metabolism , CRISPR-Associated Protein 9/pharmacology , Dendrimers/chemistry , Drug Carriers/chemistry , Gene Editing/methods , Ribonucleoproteins/pharmacology , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Gene Transfer Techniques , Humans , Mice, Inbred BALB C
4.
Nat Commun ; 11(1): 1880, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32312982

ABSTRACT

PI3K/AKT signaling is known to regulate cancer metabolism, but whether metabolic feedback regulates the PI3K/AKT pathway is unclear. Here, we demonstrate the important reciprocal crosstalk between the PI3K/AKT signal and pentose phosphate pathway (PPP) branching metabolic pathways. PI3K/AKT activation stabilizes G6PD, the rate-limiting enzyme of the PPP, by inhibiting the newly identified E3 ligase TIRM21 and promotes the PPP. PPP metabolites, in turn, reinforce AKT activation and further promote cancer metabolic reprogramming by blocking the expression of the AKT inhibitor PHLDA3. Knockout of TRIM21 or PHLDA3 promotes crosstalk and cell proliferation. Importantly, PTEN null human cancer cells and in vivo murine models are sensitive to anti-PPP treatments, suggesting the importance of the PPP in maintaining AKT activation even in the presence of a constitutively activated PI3K pathway. Our study suggests that blockade of this reciprocal crosstalk mechanism may have a therapeutic benefit for cancers with PTEN loss or PI3K/AKT activation.


Subject(s)
Nuclear Proteins/metabolism , Pentose Phosphate Pathway/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribonucleoproteins/metabolism , A549 Cells , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Models, Animal , Gene Knockout Techniques , Glucosephosphate Dehydrogenase , Humans , Male , Mice , Neoplasm Transplantation , Neoplasms/metabolism , Nuclear Proteins/pharmacology , PTEN Phosphohydrolase/metabolism , Pentose Phosphate Pathway/physiology , Ribonucleoproteins/pharmacology , Signal Transduction
5.
J Invest Dermatol ; 140(10): 1985-1993.e5, 2020 10.
Article in English | MEDLINE | ID: mdl-32142798

ABSTRACT

End-joining‒based gene editing is frequently used for efficient reframing and knockout of target genes. However, the associated random, unpredictable, and often heterogeneous repair outcomes limit its applicability for therapeutic approaches. This study revealed more precise and predictable outcomes simply on the basis of the sequence context at the CRISPR/Cas9 target site. The severe dystrophic form of the blistering skin disease epidermolysis bullosa (DEB) represents a suitable model platform to test these recent developments for the disruption and reframing of dominant and recessive alleles, respectively, both frequently seen in DEB. We delivered a CRISPR/Cas9 nuclease as ribonucleoprotein into primary wild-type and recessive DEB keratinocytes to introduce a precise predictable single adenine sense-strand insertion at the target site. We achieved type VII collagen knockout in more than 40% of ribonucleoprotein-treated primary wild-type keratinocytes and type VII collagen restoration in more than 70% of ribonucleoprotein-treated recessive DEB keratinocytes. Next-generation sequencing of the on-target site revealed the presence of the precise adenine insertion upstream of the pathogenic mutation in at least 17% of all analyzed COL7A1 alleles. This demonstrates that COL7A1 editing based on precise end-joining‒mediated DNA repair is an efficient strategy to revert the disease-associated nature of DEB regardless of the mutational inheritance.


Subject(s)
CRISPR-Cas Systems , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Gene Editing , Cells, Cultured , DNA End-Joining Repair , Female , High-Throughput Nucleotide Sequencing , Humans , Keratinocytes/metabolism , Mutation , Ribonucleoproteins/pharmacology
6.
J Gen Virol ; 101(1): 21-32, 2020 01.
Article in English | MEDLINE | ID: mdl-31702536

ABSTRACT

Peptide-based inhibitors hold promising potential in the development of antiviral therapy. Here, we investigated the antiviral potential of fragmented viral proteins derived from ribonucleoprotein (RNP) components of the human respiratory syncytial virus (HRSV). Based on a mimicking approach that targets the functional domains of viral proteins, we designed various fragments of nucleoprotein (N), matrix protein M2-1 and phosphoprotein (P) and tested the antiviral activity in an RSV mini-genome system. We found that the fragment comprising residues 130-180 and 212-241 in the C-terminal region of P (81 amino acid length), denoted as P Fr, significantly inhibited the polymerase activity through competitive binding to the full-length P. Further deletion analysis of P Fr suggested that three functional domains in P Fr (oligomerization, L-binding and nucleocapsid binding) are required for maximum inhibitory activity. More importantly, a purified recombinant P Fr displayed significant antiviral activity at low nanomolar range in RSV-infected HEp-2 cells. These results highlight P as an important target for the development of antiviral compounds against RSV and other paramyxoviruses.


Subject(s)
Antiviral Agents/metabolism , Respiratory Syncytial Virus, Human/metabolism , Ribonucleoproteins/metabolism , Ribonucleoproteins/pharmacology , Viral Proteins/metabolism , Viral Proteins/pharmacology , Virus Diseases/drug therapy , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Humans , Nucleocapsid/metabolism , Nucleoproteins/metabolism , Phosphoproteins/metabolism , Protein Transport/physiology
7.
Acc Chem Res ; 52(6): 1555-1564, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31099553

ABSTRACT

The discovery of CRISPR/Cas has revolutionized the field of genome editing. CRIPSR/Cas components are part of the bacterial immune system and are able to induce double-strand DNA breaks in the genome, which are resolved by endogenous DNA repair mechanisms. The most relevant of these are the error-prone nonhomologous end joining and homology directed repair pathways. The former can lead to gene knockout by introduction of insertions and deletions at the cut site, while the latter can be used for gene correction based on a provided repair template. In this Account, we focus on the delivery aspects of CRISPR/Cas for therapeutic applications in vivo. Safe and effective delivery of the CRISPR/Cas components into the nucleus of affected cells is essential for therapeutic gene editing. These components can be delivered in several formats, such as pDNA, viral vectors, or ribonuclear complexes. In the ideal case, the delivery system should address the current limitations of CRISPR gene editing, which are (1) lack of targeting specific tissues or cells, (2) the inability to enter cells, (3) activation of the immune system, and (4) off-target events. To circumvent most of these problems, initial therapeutic applications of CRISPR/Cas were performed on cells ex vivo via classical methods (e.g., microinjection or electroporation) and novel methods (e.g., TRIAMF and iTOP). Ideal candidates for such methods are, for example, hematopoietic cells, but not all tissue types are suited for ex vivo manipulation. For direct in vivo application, however, delivery systems are needed that can target the CRISPR/Cas components to specific tissues or cells in the human body, without causing immune activation or causing high frequencies of off-target effects. Viral systems have been used as a first resort to transduce cells in vivo. These systems suffer from problems related to packaging constraints, immunogenicity, and longevity of Cas expression, which favors off-target events. Viral vectors are as such not the best choice for direct in vivo delivery of CRISPR/Cas. Synthetic vectors can deliver nucleic acids as well, without the innate disadvantages of viral vectors. They can be classed into lipid, polymeric, and inorganic particles, all of which have been reported in the literature. The advantage of synthetic systems is that they can deliver the CRISPR/Cas system also as a preformed ribonucleoprotein complex. The transient nature of this approach favors low frequencies of off-target events and minimizes the window of immune activation. Moreover, from a pharmaceutical perspective, synthetic delivery systems are much easier to scale up for clinical use compared to viral vectors and can be chemically functionalized with ligands to obtain target cell specificity. The first preclinical results with lipid nanoparticles delivering CRISPR/Cas either as mRNA or ribonucleoproteins are very promising. The goal is translating these CRISPR/Cas therapeutics to a clinical setting as well. Taken together, these current trends seem to favor the use of sgRNA/Cas ribonucleoprotein complexes delivered in vivo by synthetic particles.


Subject(s)
CRISPR-Associated Protein 9/pharmacology , CRISPR-Cas Systems/genetics , Drug Carriers/chemistry , Gene Editing/methods , Metal Nanoparticles/chemistry , Animals , CRISPR-Associated Protein 9/genetics , Gene Transfer Techniques , Humans , Mice , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/pharmacology , Ribonucleoproteins/genetics , Ribonucleoproteins/pharmacology
8.
Antiviral Res ; 127: 32-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26777733

ABSTRACT

The tripartite motif protein 21 (TRIM21) is a ubiquitously expressed E3 ubiquitin ligase and an intracellular antibody receptor. TRIM21 mediates antibody-dependent intracellular neutralization (ADIN) in cytosol and provides an intracellular immune response to protect host defense against pathogen infection. In this study, swine TRIM21 (sTRIM21) was cloned and its role in ADIN was investigated. The expression of sTRIM21 is induced by type I interferon in PK-15 cells. sTRIM21 restricts FMDV infection in the presence of FMDV specific antibodies. Furthermore, sTRIM21 interacts with Fc fragment of swine immunoglobulin G (sFc) fused VP1 of FMDV and thereby causing its degradation. Both the RING and SPRY domains are essential for sTRIM21 to degrade sFc-fused VP1. These results suggest that the intracellular neutralization features of FMDV contribute to the antiviral activity of sTRIM21. sTRIM21 provide another intracellular mechanism to inhibit FMDV infection in infected cells.


Subject(s)
Antibodies, Neutralizing/immunology , Cytosol/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Ribonucleoproteins/immunology , Ribonucleoproteins/pharmacology , Animals , Antibody Specificity/immunology , Antiviral Agents , Cloning, Molecular , Cytoplasm , Cytosol/metabolism , Cytosol/virology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/virology , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Neutralization Tests , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/genetics , Sequence Analysis, DNA , Swine
9.
Assay Drug Dev Technol ; 13(8): 488-506, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26461433

ABSTRACT

Novel antiviral drugs, which are less prone to resistance development, are desirable alternatives to the currently approved drugs for the treatment of potentially serious influenza virus infections. The viral polymerase is highly conserved and serves as an attractive target for antiviral drugs since potent inhibitors would directly stop viral replication at an early stage. Recent structural studies on the functional domains of the heterotrimeric influenza polymerase, which comprises subunits PA, PB1, and PB2, opened the way to a structure-based approach for optimizing inhibitors of viral replication. These strategies, however, are limited by the use of isolated protein fragments instead of employing the entire ribonucleoprotein complex (RNP), which represents the functional form of the influenza polymerase in infected cells. In this study, we have established a screening assay for efficient and reliable analysis of potential influenza polymerase inhibitors of various molecular targets such as monoselective polymerase inhibitors targeting the endonuclease site, the cap-binding domain, and the polymerase active site, respectively. By utilizing whole viral RNPs and a radioactivity-free endpoint detection with the capability for efficient compound screening while offering high-content information on potential inhibitors to drive medicinal chemistry program in a reliable manner, this biochemical assay provides significant advantages over the currently available conventional assays. We propose that this assay can eventually be adapted for coinstantaneous analysis and subsequent optimization of two or more different chemical scaffold classes targeting multiple active sites within the polymerase complex, thus enabling the evaluation of drug combinations and characterization of molecules with dual functionality.


Subject(s)
Antiviral Agents/analysis , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/analysis , Influenza A virus/enzymology , Ribonucleoproteins/analysis , Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/genetics , Drug Evaluation, Preclinical/methods , Humans , Influenza A virus/drug effects , Ribonucleoproteins/genetics , Ribonucleoproteins/pharmacology , Virus Replication/drug effects , Virus Replication/physiology
10.
Mitochondrion ; 11(4): 564-74, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21406250

ABSTRACT

Mutations in mitochondrial DNA (mtDNA) generate multi-system disorders due to failure of ATP production. A cybrid containing a 1.9-kb mtDNA deletion from a patient with Kearns Sayre Syndrome is respiration-defective and grows glycolytically. When treated with a ribonucleoprotein (RNP) complex of polycistronic RNA 1 (pcRNA1) containing mtDNA-encoded genes and a multi-subunit carrier complex R8, full-length pcRNA1 was transported to mitochondria. Translation of the pcRNA1-encoded mRNAs was observed in mitochondria from RNP-treated cells. Respiration of the cybrid was rescued to approximately 90% of normal within hours, switching the cells to aerobic growth. These findings have implications for the development of effective mitochondrial gene therapy.


Subject(s)
DNA, Mitochondrial/metabolism , Gene Transfer Techniques , Kearns-Sayre Syndrome/genetics , Mitochondria/physiology , Ribonucleoproteins/pharmacology , Adenosine Triphosphate/biosynthesis , Base Sequence , Cell Line , Cell Respiration , Chromosome Mapping , DNA, Mitochondrial/genetics , Electron Transport Complex IV/metabolism , Gene Expression Profiling , Genetic Therapy , Humans , Kearns-Sayre Syndrome/therapy , Membrane Potential, Mitochondrial , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Oxygen/metabolism , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , RNA, Transfer/metabolism , Ribonucleoproteins/metabolism , Sequence Deletion
11.
J Immunol ; 175(12): 8409-14, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16339583

ABSTRACT

Sjögren's syndrome is a poorly understood autoimmune inflammatory illness that affects the salivary and lacrimal glands as well as other organ systems. We undertook the present study to determine whether mice immunized with short peptides from the 60-kDa Ro (or SSA) Ag, which is a common target of the autoimmunity of Sjögren's syndrome, develop an illness similar to Sjögren's syndrome. BALB/c mice were immunized with one of two short peptides from 60-kDa Ro that are know to induce epitope spreading. The animals were analyzed for the presence of anti-Ro and anti-La (or SSB) in the sera by immunoblot and ELISA. Salivary glands were collected and examined by histology after H&E staining. Salivary lymphocytes were purified and studied for cell surface makers by fluorescence-activated cell sorting. Timed stimulated salivary flow was measured. As reported previously, BALB/c mice immunized with 60-kDa Ro peptides developed an immune response directed against the entire Ro/La ribonucleoprotein particle that was similar to that found in humans with lupus or Sjögren's syndrome. Functional studies showed a statistical decrease in salivary flow in immunized mice compared with controls. Furthermore, there were lymphocytic infiltrates in the salivary glands of immunized animals that were not present in controls. The infiltrates consisted of both CD4- and CD8+ T lymphocytes as well as B lymphocytes. BALB/c mice immunized with 60-kDa Ro peptides develop anti-Ro, salivary gland lymphocyte infiltrates, and salivary dysfunction that is highly reminiscent of human Sjögren's syndrome.


Subject(s)
Peptide Fragments/pharmacology , Ribonucleoproteins/pharmacology , Salivary Glands/physiopathology , Sjogren's Syndrome/etiology , Animals , Antibodies/blood , Chemotaxis, Leukocyte , Immunization , Immunophenotyping , Lymphocytes , Mice , Mice, Inbred BALB C , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Ribonucleoproteins/administration & dosage , Ribonucleoproteins/immunology , Salivary Glands/immunology , Sjogren's Syndrome/pathology
12.
J Virol ; 79(20): 13105-15, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16189012

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of respiratory illness in infants, immunocompromised patients, and the elderly. New antiviral agents would be important tools in the treatment of acute RSV disease. RSV encodes its own RNA-dependent RNA polymerase that is responsible for the synthesis of both genomic RNA and subgenomic mRNAs. The viral polymerase also cotranscriptionally caps and polyadenylates the RSV mRNAs at their 5' and 3' ends, respectively. We have previously reported the discovery of the first nonnucleoside transcriptase inhibitor of RSV polymerase through high-throughput screening. Here we report the design of inhibitors that have improved potency both in vitro and in antiviral assays and that also exhibit activity in a mouse model of RSV infection. We have isolated virus with reduced susceptibility to this class of inhibitors. The mutations conferring resistance mapped to a novel motif within the RSV L gene, which encodes the catalytic subunit of RSV polymerase. This motif is distinct from the catalytic region of the L protein and bears some similarity to the nucleotide binding domain within nucleoside diphosphate kinases. These findings lead to the hypothesis that this class of inhibitors may block synthesis of RSV mRNAs by inhibiting guanylylation of viral transcripts. We show that short transcripts produced in the presence of inhibitor in vitro do not contain a 5' cap but, instead, are triphosphorylated, confirming this hypothesis. These inhibitors constitute useful tools for elucidating the molecular mechanism of RSV capping and represent valid leads for the development of novel anti-RSV therapeutics.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , RNA, Messenger/metabolism , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/enzymology , Ribonucleoproteins/pharmacology , Administration, Intranasal , Amino Acid Sequence , Animals , Catalytic Domain/genetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA Caps/biosynthesis , RNA Caps/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/genetics , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Ribonucleoproteins/administration & dosage , Ribonucleoproteins/chemistry , Sequence Alignment , Virus Replication/drug effects
13.
Genes Cells ; 10(10): 953-62, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16164596

ABSTRACT

Fragile X syndrome is caused by expansion of a d(CGG) triplet repeat in the 5'-untranslated region of the first exon of the FMR1 gene resulting in silencing of the gene. The d(CGG) repeat has been reported to form hairpin and quadruplex structures in vitro, and formation of these higher structures could be responsible for its unstable expansion in the syndrome, although molecular mechanisms underlying the repeat expansion still remain elusive. We have previously proved that UP1, a proteolytic product of hnRNP A1, unfolds the intramolecular quadruplex structures of d(GGCAG)5 and d(TTAGGG)4 and abrogates the arrest of DNA synthesis at d(GGG)n sites. Here, we demonstrate that the d(CGG) repeat forms a peculiar DNA structure, which deviates from the canonical B-form structure. In addition, UP1 was demonstrated by CD spectrum analysis to unfold this characteristic higher structure of the d(CGG) repeat and to abrogate the arrest of DNA synthesis at the site. This ability of UP1 suggests that unfolding of unusual DNA structures of a triplet repeat is required for DNA synthesis processes.


Subject(s)
Nucleic Acid Conformation , Ribonucleoproteins/metabolism , Thymus Hormones/metabolism , Trinucleotide Repeats/drug effects , Circular Dichroism , DNA/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Humans , Kinetics , Potassium Chloride/metabolism , Recombinant Proteins/metabolism , Ribonucleoproteins/pharmacology , Thymus Hormones/pharmacology , Trinucleotide Repeats/genetics
14.
Protein Expr Purif ; 39(2): 288-95, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15642481

ABSTRACT

According to codon preference of Escherichia coli, the optimized coding sequence of human vasostatin120-180aa (VAS) was obtained by chemical synthesis and molecular cloning methods. Using PCR and enzyme digestion, the full encoding sequence for VAS was cloned into the E. coli expression vector pALEX and expressed as a GST fusion protein in BL21 (DE3) strain. GST-VAS protein approximately accounted for 45% of the total bacterial proteins. Most of target protein existed in inclusion body. To improve the solubility of GST-VAS, the contribution of low temperature and molecular chaperone co-expression to the solubility of GST-VAS was tested. The results showed that co-expression with chaperons, TF and GroES/GroEL, and low expression temperature cooperatively improved the solubility of GST-VAS from 10 to 85%, and the yield of soluble GST-VAS was sixfold increased. When purified by GST affinity chromatography, 50 mg GST-VAS was obtained with purity over 85% from 1 L culture. Intact VAS was released by enterokinase digestion and further purified by Sephadex G50 gel filtration chromatography. About 7.2 mg intact homogeneous VAS protein was finally produced from 1L bacterial culture. The identity of GST-VAS and VAS was validated by Western blotting analysis. Recombinant VAS protein displayed distinct inhibition of endothelial cell proliferation and anti-angiogenic activity by chick embryo chorioallantoic membrane assay.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Calcium-Binding Proteins/isolation & purification , Calcium-Binding Proteins/metabolism , Neovascularization, Physiologic/drug effects , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Ribonucleoproteins/isolation & purification , Ribonucleoproteins/metabolism , Animals , Blotting, Western , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/pharmacology , Calreticulin , Cell Proliferation/drug effects , Cells, Cultured , Chaperonin 10/genetics , Chaperonin 10/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Chick Embryo , Chorioallantoic Membrane/drug effects , Cloning, Molecular , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Fibroblasts/metabolism , Gene Expression , Glutathione Transferase/metabolism , Humans , Isopropyl Thiogalactoside/pharmacology , Mice , NIH 3T3 Cells , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptidylprolyl Isomerase/metabolism , Plasmids , Promoter Regions, Genetic , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ribonucleoproteins/genetics , Ribonucleoproteins/pharmacology , Solubility , Temperature , Umbilical Veins/cytology
15.
J Virol ; 78(7): 3763-76, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15016896

ABSTRACT

La, a 52-kDa autoantigen in patients with systemic lupus erythematosus, was one of the first cellular proteins identified to interact with viral internal ribosome entry site (IRES) elements and stimulate poliovirus (PV) and hepatitis C virus (HCV) IRES-mediated translation. Previous results from our laboratory have shown that a small, yeast RNA (IRNA) could selectively inhibit PV and HCV IRES-mediated translation by sequestering the La protein. Here we have identified an 18-amino-acid-long sequence from the N-terminal "La motif" which is required for efficient interaction of La with IRNA and viral 5' untranslated region (5'-UTR) elements. A synthetic peptide (called LAP, for La peptide) corresponding to this sequence (amino acids 11 to 28) of La was found to efficiently inhibit viral IRES-mediated translation in vitro. The LAP efficiently enters Huh-7 cells and preferentially inhibits HCV IRES-mediated translation programmed by a bicistronic RNA in vivo. The LAP does not bind RNA directly but appears to block La binding to IRNA and PV 5'-UTR. Competition UV cross-link and translation rescue experiments suggested that LAP inhibits IRES-mediated translation by interacting with proteins rather than RNA. Mutagenesis of LAP demonstrates that single amino acid changes in a highly conserved sequence within LAP are sufficient to eliminate the translation-inhibitory activity of LAP. When one of these mutations (Y23Q) is introduced into full-length La, the mutant protein is severely defective in interacting with the PV IRES element and consequently unable to stimulate IRES-mediated translation. However, the La protein with a mutation of the next tyrosine moiety (Y24Q) could still interact with PV 5'-UTR and stimulate viral IRES-mediated translation significantly. These results underscore the importance of the La N-terminal amino acids in RNA binding and viral RNA translation. The possible role of the LAP sequence in La-RNA binding and stimulation of viral IRES-mediated translation is discussed.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Hepacivirus/drug effects , Peptide Fragments/pharmacology , Poliovirus/drug effects , Protein Biosynthesis/drug effects , RNA, Viral/metabolism , Ribonucleoproteins/antagonists & inhibitors , Ribonucleoproteins/pharmacology , 5' Untranslated Regions/genetics , 5' Untranslated Regions/metabolism , 5' Untranslated Regions/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Autoantigens/chemistry , Autoantigens/metabolism , Autoantigens/pharmacology , Cell Extracts/pharmacology , Cell Line, Tumor , Cell Membrane Permeability , Cell-Free System , HeLa Cells , Hepacivirus/genetics , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Poliovirus/genetics , RNA Caps/genetics , RNA Caps/metabolism , RNA, Viral/genetics , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Sequence Deletion/genetics , Tyrosine/genetics , Tyrosine/metabolism , SS-B Antigen
16.
Gen Comp Endocrinol ; 136(2): 217-24, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15028525

ABSTRACT

Chromogranin A (CGA) N-terminal fragments corresponding to residues 1-76 and 1-113, named vasostatins for their inhibitory effects on vascular tension, have been postulated as important homeostatic regulators of the cardiovascular system. We have used an in vitro isolated working frog (Rana esculenta) heart as a bioassay to study the effects of exogenous human recombinant CGA 1-76 (VS-1) and human CGA 7-57 synthetic peptide on cardiac performance. Under basal conditions, the concentration-response curves of the two peptides exhibited a significant negative inotropism. This vasostatin response was unaffected by pretreatment with either Triton X-100 or two nitric oxide synthase inhibitors, i.e., N(G)-monomethyl-L-arginine and L-N5 (5)(1-iminoethyl) ornithine or the soluble guanylate cyclase inhibitor 1H-(1,2,4) oxadiazolo-(4,3-a) quinoxalin-1-one, indicating an endocardial endothelium-nitric oxide-cGMP-independent mechanism. The negative inotropism was also unaffected by either adrenergic (i.e., phentolamine and propranolol) or muscarinic (atropine) receptor or G proteins (pertussis toxin) inhibition. On the contrary, it was dependent from both extracellular Ca(2+) and K(+) channels, since it was abolished by pretreatment to either the Ca(2+) channel inhibitors lanthanum and diltiazem or the K(+) channel inhibitors Ba(2+), 4-aminopyridine, tetraethylammonium chloride, and glibenclamide. In conclusion, the findings that vasostatins exert an inhibitory modulation on basal cardiac performance and counteract, as previously reported, the adrenergic-mediated positive inotropism, strongly support a cardio-regulatory role for these peptides.


Subject(s)
Calcium-Binding Proteins/pharmacology , Chromogranins/chemistry , Myocardial Contraction/drug effects , Peptide Fragments/pharmacology , Rana esculenta , Ribonucleoproteins/pharmacology , Amino Acid Sequence , Animals , Calcium-Binding Proteins/chemistry , Calreticulin , Chromogranin A , Depression, Chemical , In Vitro Techniques , Molecular Sequence Data , Peptide Fragments/chemistry , Potassium/pharmacology , Receptors, Adrenergic, beta/physiology , Receptors, Muscarinic/physiology , Ribonucleoproteins/chemistry , Sequence Alignment
17.
Med Sci (Paris) ; 19(12): 1271-9, 2003 Dec.
Article in French | MEDLINE | ID: mdl-14691753

ABSTRACT

RNA-proteins interactions are involved in numerous cellular functions. These interactions are found in most cases within complex macromolecular assemblies. The recent development of tools and techniques to study RNA-protein complexes has significantly increased our knowledge in the nature of these specific interactions. The aim of this article is to present the different techniques used to study RNA-protein complexes, as well as recent data concerning the application of RNA as therapeutic molecules.


Subject(s)
Protein Binding , RNA/metabolism , Ribonucleoproteins/pharmacology , Humans
18.
J Autoimmun ; 21(1): 17-26, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12892732

ABSTRACT

BACKGROUND: Antibodies to La/SSB are usually found in sera of patients with Sjogren's Syndrome (SS) and Systemic Lupus Erythematosus (SLE). Recent work from our laboratory (Mol Med 2002;8:293-305) revealed that an active idiotypic network involving antibodies to epitopes of La/SSB and their anti-idiotypes exist in human sera. The anti-idiotypic antibodies were detected using complementary peptides to B-cell epitopes of the autoantigen. The principle of the complementary peptides is based on the 'molecular recognition' theory. According to this theory, translation of two complementary RNA strands (coding and non-coding strand) into protein, generate a pair of peptides, which bind each other with specificity and high affinity. AIM: To investigate antibody production and T-cell responses in non-autoimmune-susceptible animal strains which were immunized with the epitope 289-308aa of La/SSB as well as its complementary epitope. MATERIALS AND METHODS: Balb/c mice were immunized with a peptide corresponding to epitope 289-308aa (pep) or its complementary (cpep) peptide (5 animals/group). The sera were tested for the presence of antibodies to pep and cpep as well as for epitope spreading to recombinant human La/SSB and a major B-cell epitope of La/SSB spanning the region 349-364aa. Another group of animals was sacrificed on day 10 and T-cell responses against pep and cpep were evaluated in cells from lymph nodes and spleen. RESULTS: Immunizations with either pep or cpep led to the appearance of antibodies against the immunogen peptide by day 31 which subsequently was followed by antibody production to its complementary peptide by day 55. In two out of five animals immunized with the epitope 289-308aa, a spreading of the immune response to epitope 349-364aa was observed. In the remaining three animals, negative for antibodies to pep349-364, a specific treatment of sera, using cpep349-364 revealed that anti-idiotypic antibodies masked antibodies to pep349-364. In all immunization experiments high T-cell proliferative responses to both pep and cpep peptides were detected. CONCLUSIONS: Complementary peptides to epitopes of La/SSB can be utilized as probes to study the development of an idiotypic-anti-idiotypic network towards the major autoantigen. The ability of pep and cpep peptides to induce both B-cell and T-cell responses may provide useful insights into understanding further the initiation and maintenance of autoimmune response and create new tools for therapeutic intervention.


Subject(s)
Antibodies/immunology , Epitopes/immunology , Immunoglobulin Idiotypes/immunology , Peptides/immunology , Ribonucleoproteins/immunology , Animals , Autoantigens , Female , Immunization , Mice , Mice, Inbred BALB C , Peptides/pharmacology , Ribonucleoproteins/pharmacology , Vaccines/pharmacology , SS-B Antigen
19.
Ann N Y Acad Sci ; 971: 362-5, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12438153

ABSTRACT

An in vitro isolated working frog heart (Rana esculenta) was used to study the effects of exogenous CGA(1-76) (vasostatin 1), CGA(1-113) (vasostatin 2), and the synthetic CGA(7-57) on cardiac performance. Under basal cardiac conditions, the dose-response curves of the three peptides from 10(-8) to 10(-7) M showed a significant calcium-dependent negative inotropism that involved neither the endocardial endothelium nor the adrenergic and muscarinic receptors. In addition, the CgA fragments clearly counteracted the typical positive inotropism of isoprenaline (10(-<9) M). Taken together, these results provide the first evidence for a cardio-suppressive role for the vasostatins.


Subject(s)
Calcium-Binding Proteins/pharmacology , Heart/drug effects , Peptide Fragments/pharmacology , Ribonucleoproteins/pharmacology , Animals , Calcium/metabolism , Calreticulin , Dose-Response Relationship, Drug , Endocardium/cytology , Female , Isoproterenol/pharmacology , Male , Peptides/chemistry , Perfusion , Rana esculenta
20.
Nucleic Acids Res ; 30(20): 4500-8, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12384597

ABSTRACT

Translation initiation in Coxsackievirus B3 (CVB3) occurs via ribosome binding to an internal ribosome entry site (IRES) located in the 5'-untranslated region (UTR) of the viral RNA. This unique mechanism of translation initiation requires various trans-acting factors from the host. We show that human La autoantigen (La) binds to the CVB3 5'-UTR and also demonstrate the dose-dependent effect of exogenously added La protein in stimulating CVB3 IRES-mediated translation. The requirement of La for CVB3 IRES mediated translation has been further demonstrated by inhibition of translation as a result of sequestering La and its restoration by exogenous addition of recombinant La protein. The abundance of La protein in various mouse tissue extracts has been probed using anti-La antibody. Pancreatic tissue, a target organ for CVB3 infection, was found to have a large abundance of La protein which was demonstrated to interact with the CVB3 5'-UTR. Furthermore, exogenous addition of pancreas extract to in vitro translation reactions resulted in a dose dependent stimulation of CVB3 IRES-mediated translation. These observations indicate the role of La in CVB3 IRES-mediated translation, and suggest its possible involvement in the efficient translation of the viral RNA in the pancreas.


Subject(s)
Enterovirus B, Human/genetics , Gene Expression Regulation, Viral , Protein Biosynthesis , RNA, Viral/metabolism , Ribonucleoproteins/physiology , 5' Untranslated Regions , Animals , Autoantigens , Binding Sites , Binding, Competitive , Dose-Response Relationship, Drug , Enterovirus B, Human/metabolism , HeLa Cells , Humans , Mice , Pancreas/metabolism , Protein Biosynthesis/drug effects , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , RNA, Viral/chemistry , Ribonucleoproteins/pharmacology , Ribosomes/metabolism , Tissue Distribution , SS-B Antigen
SELECTION OF CITATIONS
SEARCH DETAIL
...