Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
EBioMedicine ; 90: 104500, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36893587

ABSTRACT

BACKGROUND: Despite the advent of improved therapeutic options for advanced prostate cancer, the durability of clinical benefits is limited due to inevitable development of resistance. By constitutively sustaining androgen receptor (AR) signaling, expression of ligand-binding domain truncated AR variants (AR-V(ΔLBD)) accounts for the major mechanism underlying the resistance to anti-androgen drugs. Strategies to target AR and its LBD truncated variants are needed to prevent the emergence or overcome drug resistance. METHODS: We utilize Proteolysis Targeting Chimeras (PROTAC) technology to achieve induced degradation of both full-length AR (AR-FL) and AR-V(ΔLBD) proteins. In the ITRI-PROTAC design, an AR N-terminal domain (NTD) binding moiety is appended to von-Hippel-Lindau (VHL) or Cereblon (CRBN) E3 ligase binding ligand with linker. FINDINGS: In vitro studies demonstrate that ITRI-PROTAC compounds mechanistically degrade AR-FL and AR-V(ΔLBD) proteins via ubiquitin-proteasome system, leading to impaired AR transactivation on target gene expression, and inhibited cell proliferation accompanied by apoptosis activation. The compounds also significantly inhibit enzalutamide-resistant growth of castration resistant prostate cancer (CRPC) cells. In castration-, enzalutamide-resistant CWR22Rv1 xenograft model without hormone ablation, ITRI-90 displays a pharmacokinetic profile with decent oral bioavailability and strong antitumor efficacy. INTERPRETATION: AR NTD that governs the transcriptional activities of all active variants has been considered attractive therapeutic target to block AR signaling in prostate cancer cells. We demonstrated that utilizing PROTAC for induced AR protein degradation via NTD represents an efficient alternative therapeutic strategy for CRPC to overcome anti-androgen resistance. FUNDING: The funding detail can be found in the Acknowledgements section.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Proteolysis Targeting Chimera , Ligands , Nitriles/therapeutic use , Cell Line, Tumor , Proteolysis
2.
Invest Ophthalmol Vis Sci ; 62(13): 12, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34661609

ABSTRACT

Purpose: The purpose of this study was to develop a preclinical compound, ITRI-E-(S)4046, a dual synergistic inhibitor of myosin light chain kinase 4 (MYLK4) and Rho-related protein kinase (ROCK), for reducing intraocular pressure (IOP). Methods: ITRI-E-(S)4046 is an amino-pyrazole derivative with physical and chemical properties suitable for ophthalmic formulation. In vitro kinase inhibition was evaluated using the Kinase-Glo Luminescent Kinase Assays. A comprehensive kinase selectivity analysis of ITRI-E-(S)4046 was performed using the KINOMEscan assay from DiscoverRx. The IOP reduction and tolerability of ITRI-E-(S)4046 were assessed in ocular normotensive rabbits, ocular normotensive non-human primates, and ocular hypertensive rabbits. In vivo studies were conducted to assess drug concentrations in ocular tissue. The adverse ocular effects of rabbit eyes were evaluated following the OECD405 guidelines. Results: ITRI-E-(S)4046 showed highly selective kinase inhibitory activity against ROCK1/2, MYLK4, and mitogen-activated protein kinase kinase kinase 19 (MAP3K19), with high specificity against protein kinase A, G, and C families. In ocular normotensive rabbits and non-human primates, the mean IOP reductions of 0.1% ITRI-E-(S)4046 eye drops were 29.8% and 28.5%, respectively. In hypertonic saline-induced and magnetic beads-induced ocular hypertensive rabbits, the mean IOP reductions of ITRI-E-(S)4046 0.1% eye drops were 46.9% and 22.0%, respectively. ITRI-E-(S)4046 was well tolerated with only temporary and minor signs of hyperemia. Conclusions: ITRI-E-(S)4046 is a novel type of highly specific ROCK1/2 and MYLK4 inhibitor that can reduce IOP in normotensive and hypertensive animal models. It has the potential to become an effective and well-tolerated treatment for glaucoma.


Subject(s)
Benzoates/pharmacology , Calcium-Binding Proteins/antagonists & inhibitors , Intraocular Pressure/drug effects , Isoquinolines/pharmacology , Myosin-Light-Chain Kinase/antagonists & inhibitors , Ocular Hypertension/drug therapy , Sulfonamides/pharmacology , beta-Alanine/analogs & derivatives , Animals , Disease Models, Animal , Humans , Macaca , Male , Ocular Hypertension/physiopathology , Rabbits , Tonometry, Ocular , beta-Alanine/pharmacology , rho-Associated Kinases/antagonists & inhibitors
3.
Biomedicines ; 9(2)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557143

ABSTRACT

Prostate cancer (PCa) is one of the most commonly diagnosed cancers in men and usually becomes refractory because of recurrence and metastasis. CD44, a transmembrane glycoprotein, serves as a receptor for hyaluronic acid (HA). It has been found to be abundantly expressed in cancer stem cells (CSCs) that often exhibit a radioresistant phenotype. Cytolethal distending toxin (CDT), produced by Campylobacter jejuni, is a tripartite genotoxin composed of CdtA, CdtB, and CdtC subunits. Among the three, CdtB acts as a type I deoxyribonuclease (DNase I), which creates DNA double-strand breaks (DSBs). Nanoparticles loaded with antitumor drugs and specific ligands that recognize cancerous cell receptors are promising methods to overcome the therapeutic challenges. In this study, HA-decorated nanoparticle-encapsulated CdtB (HA-CdtB-NPs) were prepared and their targeted therapeutic activity in radioresistant PCa cells was evaluated. Our results showed that HA-CdtB-NPs sensitized radioresistant PCa cells by enhancing DSB and causing G2/M cell-cycle arrest, without affecting the normal prostate epithelial cells. HA-CdtB-NPs possess maximum target specificity and delivery efficiency of CdtB into the nucleus and enhance the effect of radiation in radioresistant PCa cells. These findings demonstrate that HA-CdtB-NPs exert target specificity accompanied with radiomimetic activity and can be developed as an effective strategy against radioresistant PCa.

4.
Sci Rep ; 9(1): 4205, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862888

ABSTRACT

Helicobacter pylori infection is associated with high incidence of gastric diseases. The extensive therapy of H. pylori infection with antibiotics has increased its resistance rates worldwide. Ovatodiolide, a pure constituent isolated from Anisomeles indica, has been demonstrated to possess bactericidal activity against H. pylori. In this study, ovatodiolide inhibited the growth of both H. pylori reference strain and clinical multidrug-resistant isolates. Docking analysis revealed that ovatodiolide fits into the hydrophobic pocket of a ribosomal protein, RpsB. Furthermore, ovatodiolide inhibited bacterial growth by reducing levels of RpsB, which plays a crucial role in protein translation. Our results demonstrate that ovatodiolide binds to a ribosomal protein and interferes with protein synthesis. This study provides evidence that ovatodiolide has the potential to be developed into a potent therapeutic agent for treating H. pylori infection.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins/chemistry , Diterpenes , Drug Resistance, Multiple, Bacterial/drug effects , Helicobacter pylori , Lamiaceae/chemistry , Molecular Docking Simulation , Ribosomal Proteins/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Diterpenes/chemistry , Diterpenes/pharmacology , Helicobacter pylori/chemistry , Helicobacter pylori/growth & development , Humans
5.
Nat Commun ; 8: 15874, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28639619

ABSTRACT

Long noncoding RNAs (lncRNAs) have been implicated in hypoxia/HIF-1-associated cancer progression through largely unknown mechanisms. Here we identify MIR31HG as a hypoxia-inducible lncRNA and therefore we name it LncHIFCAR (long noncoding HIF-1α co-activating RNA); we describe its oncogenic role as a HIF-1α co-activator that regulates the HIF-1 transcriptional network, crucial for cancer development. Extensive analyses of clinical data indicate LncHIFCAR level is substantially upregulated in oral carcinoma, significantly associated with poor clinical outcomes and representing an independent prognostic predictor. Overexpression of LncHIFCAR induces pseudo-hypoxic gene signature, whereas knockdown of LncHIFCAR impairs the hypoxia-induced HIF-1α transactivation, sphere-forming ability, metabolic shift and metastatic potential in vitro and in vivo. Mechanistically, LncHIFCAR forms a complex with HIF-1α via direct binding and facilitates the recruitment of HIF-1α and p300 cofactor to the target promoters. Our results uncover an lncRNA-mediated mechanism for HIF-1 activation and establish the clinical values of LncHIFCAR in prognosis and potential therapeutic strategy for oral carcinoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mouth Neoplasms/genetics , RNA, Long Noncoding/genetics , Animals , Biomarkers, Tumor/genetics , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Nude , Middle Aged , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Prognosis , Proportional Hazards Models , Survival Analysis , Tumor Hypoxia/genetics , Xenograft Model Antitumor Assays
6.
Cell Rep ; 16(11): 3016-3027, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27626669

ABSTRACT

The histone lysine demethylase KDM4A/JMJD2A has been implicated in prostate carcinogenesis through its role in transcriptional regulation. Here, we describe KDM4A as a E2F1 coactivator and demonstrate a functional role for the E2F1-KDM4A complex in the control of tumor metabolism. KDM4A associates with E2F1 on target gene promoters and enhances E2F1 chromatin binding and transcriptional activity, thereby modulating the transcriptional profile essential for cancer cell proliferation and survival. The pyruvate dehydrogenase kinases (PDKs) PDK1 and PDK3 are direct targets of KDM4A and E2F1 and modulate the switch between glycolytic metabolism and mitochondrial oxidation. Downregulation of KDM4A leads to elevated activity of pyruvate dehydrogenase and mitochondrial oxidation, resulting in excessive accumulation of reactive oxygen species. The altered metabolic phenotypes can be partially rescued by ectopic expression of PDK1 and PDK3, indicating a KDM4A-dependent tumor metabolic regulation via PDK. Our results suggest that KDM4A is a key regulator of tumor metabolism and a potential therapeutic target for prostate cancer.


Subject(s)
E2F1 Transcription Factor/metabolism , Glycolysis , Jumonji Domain-Containing Histone Demethylases/metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Chromatin/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice, Nude , Oxidation-Reduction , Phenotype , Prostatic Neoplasms/genetics , Protein Binding , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Transcription, Genetic
7.
Int J Mol Sci ; 16(12): 28943-78, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26690121

ABSTRACT

Hormone-refractory prostate cancer frequently relapses from therapy and inevitably progresses to a bone-metastatic status with no cure. Understanding of the molecular mechanisms conferring resistance to androgen deprivation therapy has the potential to lead to the discovery of novel therapeutic targets for type of prostate cancer with poor prognosis. Progression to castration-resistant prostate cancer (CRPC) is characterized by aberrant androgen receptor (AR) expression and persistent AR signaling activity. Alterations in metabolic activity regulated by oncogenic pathways, such as c-Myc, were found to promote prostate cancer growth during the development of CRPC. Non-coding RNAs represent a diverse family of regulatory transcripts that drive tumorigenesis of prostate cancer and various other cancers by their hyperactivity or diminished function. A number of studies have examined differentially expressed non-coding RNAs in each stage of prostate cancer. Herein, we highlight the emerging impacts of microRNAs and long non-coding RNAs linked to reactivation of the AR signaling axis and reprogramming of the cellular metabolism in prostate cancer. The translational implications of non-coding RNA research for developing new biomarkers and therapeutic strategies for CRPC are also discussed.


Subject(s)
Prostate/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Untranslated/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Animals , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Male , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Untranslated/genetics , Receptors, Androgen/genetics
8.
Mol Microbiol ; 97(6): 1128-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26082024

ABSTRACT

Helicobacter pylori inhabits the gastric mucosa where it senses and responds to various stresses via a two-component systems (TCSs) that enable its persistent colonization. The aim of this study was to investigate whether any of the three paired TCSs (ArsRS, FleRS and CrdRS) in H. pylori respond to nitrosative stress. The results showed that the expression of crdS was significantly increased upon exposure to nitric oxide (NO). crdS-knockout (ΔcrdS) and crdR/crdS-knockout (ΔcrdRS) H. pylori, but not arsS-knockout (ΔarsS) or fleS-knockout (ΔfleS) H. pylori, showed a significant loss of viability upon exposure to NO compared with wild-type strain. Knockin crdS (ΔcrdS-in) significantly restored viability in the presence of NO. Global transcriptional profiling analysis of wild-type and ΔcrdS H. pylori in the presence or absence of NO showed that 101 genes were differentially expressed, including copper resistance determinant A (crdA), transport, binding and envelope proteins. The CrdR binding motifs were investigated by competitive electrophoretic mobility shift assay, which revealed that the two AC-rich regions in the crdA promoter region are required for binding. These results demonstrate that CrdR-crdA interaction enables H. pylori to survive under nitrosative stress.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Helicobacter pylori/metabolism , Nitric Oxide/metabolism , Stress, Physiological , Base Sequence , Copper/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Knockout Techniques , Helicobacter pylori/genetics , Molecular Sequence Data , Promoter Regions, Genetic
9.
Proc Natl Acad Sci U S A ; 111(52): 18697-702, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25512540

ABSTRACT

Long noncoding RNAs (lncRNAs) have been implicated in a variety of physiological and pathological processes, including cancer. In prostate cancer, prostate cancer gene expression marker 1 (PCGEM1) is an androgen-induced prostate-specific lncRNA whose overexpression is highly associated with prostate tumors. PCGEM1's tumorigenic potential has been recently shown to be in part due to its ability to activate androgen receptor (AR). Here, we report a novel function of PCGEM1 that provides growth advantages for cancer cells by regulating tumor metabolism via c-Myc activation. PCGEM1 promotes glucose uptake for aerobic glycolysis, coupling with the pentose phosphate shunt to facilitate biosynthesis of nucleotide and lipid, and generates NADPH for redox homeostasis. We show that PCGEM1 regulates metabolism at a transcriptional level that affects multiple metabolic pathways, including glucose and glutamine metabolism, the pentose phosphate pathway, nucleotide and fatty acid biosynthesis, and the tricarboxylic acid cycle. The PCGEM1-mediated gene regulation takes place in part through AR activation, but predominantly through c-Myc activation, regardless of hormone or AR status. Significantly, PCGEM1 binds directly to target promoters, physically interacts with c-Myc, promotes chromatin recruitment of c-Myc, and enhances its transactivation activity. We also identified a c-Myc binding domain on PCGEM1 that contributes to the PCGEM1-dependent c-Myc activation and target induction. Together, our data uncover PCGEM1 as a key transcriptional regulator of central metabolic pathways in prostate cancer cells. By being a coactivator for both c-Myc and AR, PCGEM1 reprograms the androgen network and the central metabolism in a tumor-specific way, making it a promising target for therapeutic intervention.


Subject(s)
Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Aerobiosis/genetics , Cell Line, Tumor , Glycolysis/genetics , HEK293 Cells , Humans , Male , NADP/genetics , NADP/metabolism , Pentose Phosphate Pathway/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
10.
J Med Chem ; 57(14): 5975-85, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24971742

ABSTRACT

The KDM4/JMJD2 Jumonji C-containing histone lysine demethylases (KDM4A-KDM4D), which selectively remove the methyl group(s) from tri/dimethylated lysine 9/36 of H3, modulate transcriptional activation and genome stability. The overexpression of KDM4A/KDM4B in prostate cancer and their association with androgen receptor suggest that KDM4A/KDM4B are potential progression factors for prostate cancer. Here, we report the crystal structure of the KDM4B·pyridine 2,4-dicarboxylic acid·H3K9me3 ternary complex, revealing the core active-site region and a selective K9/K36 site. A selective KDM4A/KDM4B inhibitor, 4, that occupies three subsites in the binding pocket is identified by virtual screening. Pharmacological and genetic inhibition of KDM4A/KDM4B significantly blocks the viability of cultured prostate cancer cells, which is accompanied by increased H3K9me3 staining and transcriptional silencing of growth-related genes. Significantly, a substantial portion of differentially expressed genes are AR-responsive, consistent with the roles of KDM4s as critical AR activators. Our results point to KDM4 as a useful therapeutic target and identify a new inhibitor scaffold.


Subject(s)
Enzyme Inhibitors/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/chemistry , Molecular Targeted Therapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Stilbenes/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Kinetics , Male , Models, Molecular , Prostatic Neoplasms/enzymology , Stilbenes/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
11.
PLoS One ; 8(12): e83322, 2013.
Article in English | MEDLINE | ID: mdl-24349489

ABSTRACT

BACKGROUND: Inherited cardiac conduction diseases (CCD) are rare but are caused by mutations in a myriad of genes. Recently, whole-exome sequencing has successfully led to the identification of causal mutations for rare monogenic Mendelian diseases. OBJECTIVE: To investigate the genetic background of a family affected by inherited CCD. METHODS AND RESULTS: We used whole-exome sequencing to study a Chinese family with multiple family members affected by CCD. Using the pedigree information, we proposed a heterozygous missense mutation (c.G695T, Gly232Val) in the lamin A/C (LMNA) gene as a candidate mutation for susceptibility to CCD in this family. The mutation is novel and is expected to affect the conformation of the coiled-coil rod domain of LMNA according to a structural model prediction. Its pathogenicity in lamina instability was further verified by expressing the mutation in a cellular model. CONCLUSIONS: Our results suggest that whole-exome sequencing is a feasible approach to identifying the candidate genes underlying inherited conduction diseases.


Subject(s)
Arrhythmias, Cardiac/genetics , Exome , Genetic Diseases, Inborn/genetics , Heart Conduction System/abnormalities , Lamin Type A/genetics , Mutation, Missense , Pedigree , Adult , Aged , Aged, 80 and over , Amino Acid Substitution , Brugada Syndrome , Cardiac Conduction System Disease , DNA Mutational Analysis , Family , Female , Humans , Male , Middle Aged , Protein Structure, Tertiary
12.
BMC Genomics ; 14: 824, 2013 Nov 23.
Article in English | MEDLINE | ID: mdl-24267727

ABSTRACT

BACKGROUND: SUMOylation, as part of the epigenetic regulation of transcription, has been intensively studied in lower eukaryotes that contain only a single SUMO protein; however, the functions of SUMOylation during mammalian epigenetic transcriptional regulation are largely uncharacterized. Mammals express three major SUMO paralogues: SUMO-1, SUMO-2, and SUMO-3 (normally referred to as SUMO-1 and SUMO-2/3). Herpesviruses, including Kaposi's sarcoma associated herpesvirus (KSHV), seem to have evolved mechanisms that directly or indirectly modulate the SUMO machinery in order to evade host immune surveillance, thus advancing their survival. Interestingly, KSHV encodes a SUMO E3 ligase, K-bZIP, with specificity toward SUMO-2/3 and is an excellent model for investigating the global functional differences between SUMO paralogues. RESULTS: We investigated the effect of experimental herpesvirus reactivation in a KSHV infected B lymphoma cell line on genomic SUMO-1 and SUMO-2/3 binding profiles together with the potential role of chromatin SUMOylation in transcription regulation. This was carried out via high-throughput sequencing analysis. Interestingly, chromatin immunoprecipitation sequencing (ChIP-seq) experiments showed that KSHV reactivation is accompanied by a significant increase in SUMO-2/3 modification around promoter regions, but SUMO-1 enrichment was absent. Expression profiling revealed that the SUMO-2/3 targeted genes are primarily highly transcribed genes that show no expression changes during viral reactivation. Gene ontology analysis further showed that these genes are involved in cellular immune responses and cytokine signaling. High-throughput annotation of SUMO occupancy of transcription factor binding sites (TFBS) pinpointed the presence of three master regulators of immune responses, IRF-1, IRF-2, and IRF-7, as potential SUMO-2/3 targeted transcriptional factors after KSHV reactivation. CONCLUSION: Our study is the first to identify differential genome-wide SUMO modifications between SUMO paralogues during herpesvirus reactivation. Our findings indicate that SUMO-2/3 modification near protein-coding gene promoters occurs in order to maintain host immune-related gene unaltered during viral reactivation.


Subject(s)
Chromatin/metabolism , Herpesvirus 8, Human/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitins/metabolism , Virus Activation , Cell Line, Tumor , Chromatin/virology , Chromatin Immunoprecipitation , Epigenesis, Genetic/immunology , Gene Ontology , Genes, MHC Class II , HEK293 Cells , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Transcriptome
13.
PLoS One ; 8(6): e66202, 2013.
Article in English | MEDLINE | ID: mdl-23762481

ABSTRACT

Cytolethal distending toxin (CDT) produced by Campylobacter jejuni comprises a heterotrimeric complex formed by CdtA, CdtB, and CdtC. Among these toxin subunits, CdtA and CdtC function as essential proteins that mediate toxin binding to cytoplasmic membranes followed by delivery of CdtB into the nucleus. The binding of CdtA/CdtC to the cell surface is mediated by cholesterol, a major component in lipid rafts. Although the putative cholesterol recognition/interaction amino acid consensus (CRAC) domain of CDT has been reported from several bacterial pathogens, the protein regions contributing to CDT binding to cholesterol in C. jejuni remain unclear. Here, we selected a potential CRAC-like region present in the CdtC from C. jejuni for analysis. Molecular modeling showed that the predicted functional domain had the shape of a hydrophobic groove, facilitating cholesterol localization to this domain. Mutation of a tyrosine residue in the CRAC-like region decreased direct binding of CdtC to cholesterol rather than toxin intermolecular interactions and led to impaired CDT intoxication. These results provide a molecular link between C. jejuni CdtC and membrane-lipid rafts through the CRAC-like region, which contributes to toxin recognition and interaction with cholesterol.


Subject(s)
Bacterial Toxins/chemistry , Campylobacter jejuni/chemistry , Cholesterol/chemistry , Membrane Microdomains/chemistry , Protein Subunits/chemistry , Amino Acid Motifs , Animals , Bacterial Toxins/metabolism , Binding Sites , CHO Cells , Campylobacter jejuni/metabolism , Campylobacter jejuni/pathogenicity , Cholesterol/metabolism , Cricetulus , Hydrophobic and Hydrophilic Interactions , Membrane Microdomains/metabolism , Molecular Docking Simulation , Molecular Sequence Data , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Subunits/metabolism , Tyrosine/chemistry , Tyrosine/genetics
14.
Article in English | MEDLINE | ID: mdl-23431343

ABSTRACT

The bacterial pathogen Helicobacter pylori (Hp) is the leading risk factor for the development of gastric cancer. Hp virulence factor, cytotoxin-associated gene A (CagA) interacted with cholesterol-enriched microdomains and leads to induction of inflammation in gastric epithelial cells (AGS). In this study, we identified a triterpenoid methylantcinate B (MAB) from the medicinal mushroom Antrodia camphorata which inhibited the translocation and phosphorylation of CagA and caused a reduction in hummingbird phenotype in HP-infected AGS cells. Additionally, MAB suppressed the Hp-induced inflammatory response by attenuation of NF-κB activation, translocation of p65 NF-κB, and phosphorylation of IκB-α, indicating that MAB modulates CagA-mediated signaling pathway. Additionally, MAB also suppressed the IL-8 luciferase activity and its secretion in HP-infected AGS cells. On the other hand, molecular structure simulations revealed that MAB interacts with CagA similarly to that of cholesterol. Moreover, binding of cholesterol to the immobilized CagA was inhibited by increased levels of MAB. Our results demonstrate that MAB is the first natural triterpenoid which competes with cholesterol bound to CagA leading to attenuation of Hp-induced pathogenesis of epithelial cells. Thus, this study indicates that MAB may have a scope to develop as a therapeutic candidate against Hp CagA-induced inflammation.

15.
J Biol Chem ; 284(39): 26708-15, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19640850

ABSTRACT

Laminaripentaose-producing beta-1,3-glucanase (LPHase), a member of glycoside hydrolase family 64, cleaves a long-chain polysaccharide beta-1,3-glucan into specific pentasaccharide oligomers. The crystal structure of LPHase from Streptomyces matensis DIC-108 was solved to 1.62 A resolution using multiple-wavelength anomalous dispersion methods. The LPHase structure reveals a novel crescent-like fold; it consists of a barrel domain and a mixed (alpha/beta) domain, forming a wide-open groove between the two domains. The liganded crystal structure was also solved to 1.80 A, showing limited conformational changes. Within the wide groove, a laminaritetraose molecule is found to sit in an electronegatively charged central region and is proximal to several conserved residues including two carboxylates (Glu(154) and Asp(170)) and four other sugar-binding residues (Thr(156), Asn(158), Trp(163), and Thr(167)). Molecular modeling using a laminarihexaose as a substrate suggests roles for Glu(154) and Asp(170) as acid and base catalysts, respectively, whereas the side chains of Thr(156), Asn(158), and Trp(163) demarcate subsite +5. Site-directed mutagenesis of Glu(154) and Asp(170) confirms that both carboxylates are essential for catalysis. Together, our results suggest that LPHase uses a direct displacement mechanism involving Glu(154) and Asp(170) to cleave a beta-1,3-glucan into specific alpha-pentasaccharide oligomers.


Subject(s)
Bacterial Proteins/metabolism , Glucan 1,3-beta-Glucosidase/metabolism , Oligosaccharides/metabolism , Streptomyces/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Catalysis , Crystallization , Crystallography, X-Ray , Glucan 1,3-beta-Glucosidase/chemistry , Glucan 1,3-beta-Glucosidase/genetics , Models, Molecular , Mutagenesis, Site-Directed , Oligosaccharides/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Streptomyces/genetics , Substrate Specificity
16.
J Biol Chem ; 282(16): 12220-9, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17307742

ABSTRACT

Helicobacter pylori AmiF formamidase that hydrolyzes formamide to produce formic acid and ammonia belongs to a member of the nitrilase superfamily. The crystal structure of AmiF was solved to 1.75A resolution using single-wavelength anomalous dispersion methods. The structure consists of a homohexamer related by 3-fold symmetry in which each subunit has an alpha-beta-beta-alpha four-layer architecture characteristic of the nitrilase superfamily. One exterior alpha layer faces the solvent, whereas the other one associates with that of the neighbor subunit, forming a tight alpha-beta-beta-alpha-alpha-beta-beta-alpha dimer. The apo and liganded crystal structures of an inactive mutant C166S were also determined to 2.50 and 2.30 A, respectively. These structures reveal a small formamide-binding pocket that includes Cys(166), Glu(60), and Lys(133) catalytic residues, in which Cys(166) acts as a nucleophile. Analysis of the liganded AmiF and N-carbamoyl d-amino acid amidohydrolase binding pockets reveals a common Cys-Glu-Lys triad, another conserved glutamate, and different subsets of ligand-binding residues. Molecular dynamic simulations show that the conserved triad has minimal fluctuations, catalyzing the hydrolysis of a specific nitrile or amide in the nitrilase superfamily efficiently.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/physiology , Cysteine/chemistry , Glutamic Acid/chemistry , Helicobacter pylori/enzymology , Lysine/chemistry , Amidohydrolases/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Crystallography, X-Ray , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...