Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Nature ; 618(7965): 590-597, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37258672

ABSTRACT

Rapidly evolving influenza A viruses (IAVs) and influenza B viruses (IBVs) are major causes of recurrent lower respiratory tract infections. Current influenza vaccines elicit antibodies predominantly to the highly variable head region of haemagglutinin and their effectiveness is limited by viral drift1 and suboptimal immune responses2. Here we describe a neuraminidase-targeting monoclonal antibody, FNI9, that potently inhibits the enzymatic activity of all group 1 and group 2 IAVs, as well as Victoria/2/87-like, Yamagata/16/88-like and ancestral IBVs. FNI9 broadly neutralizes seasonal IAVs and IBVs, including the immune-evading H3N2 strains bearing an N-glycan at position 245, and shows synergistic activity when combined with anti-haemagglutinin stem-directed antibodies. Structural analysis reveals that D107 in the FNI9 heavy chain complementarity-determinant region 3 mimics the interaction of the sialic acid carboxyl group with the three highly conserved arginine residues (R118, R292 and R371) of the neuraminidase catalytic site. FNI9 demonstrates potent prophylactic activity against lethal IAV and IBV infections in mice. The unprecedented breadth and potency of the FNI9 monoclonal antibody supports its development for the prevention of influenza illness by seasonal and pandemic viruses.


Subject(s)
Antibodies, Viral , Antibody Specificity , Influenza A virus , Influenza B virus , Influenza Vaccines , Influenza, Human , Molecular Mimicry , Neuraminidase , Animals , Humans , Mice , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Antibody Specificity/immunology , Arginine/chemistry , Catalytic Domain , Hemagglutinins, Viral/immunology , Influenza A virus/classification , Influenza A virus/enzymology , Influenza A virus/immunology , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/classification , Influenza B virus/enzymology , Influenza B virus/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Influenza, Human/prevention & control , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Neuraminidase/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Seasons , Sialic Acids/chemistry
2.
Proc Natl Acad Sci U S A ; 120(11): e2220677120, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36888659

ABSTRACT

Control over transition metal redox state is essential for metalloprotein function and can be achieved via coordination chemistry and/or sequestration from bulk solvent. Human methylmalonyl-Coenzyme A (CoA) mutase (MCM) catalyzes the isomerization of methylmalonyl-CoA to succinyl-CoA using 5'-deoxyadenosylcobalamin (AdoCbl) as a metallocofactor. During catalysis, the occasional escape of the 5'-deoxyadenosine (dAdo) moiety leaves the cob(II)alamin intermediate stranded and prone to hyperoxidation to hydroxocobalamin, which is recalcitrant to repair. In this study, we have identified the use of bivalent molecular mimicry by ADP, coopting the 5'-deoxyadenosine and diphosphate moieties in the cofactor and substrate, respectively, to protect against cob(II)alamin overoxidation on MCM. Crystallographic and electron paramagnetic resonance (EPR) data reveal that ADP exerts control over the metal oxidation state by inducing a conformational change that seals off solvent access, rather than by switching five-coordinate cob(II)alamin to the more air stable four-coordinate state. Subsequent binding of methylmalonyl-CoA (or CoA) promotes cob(II)alamin off-loading from MCM to adenosyltransferase for repair. This study identifies an unconventional strategy for controlling metal redox state by an abundant metabolite to plug active site access, which is key to preserving and recycling a rare, but essential, metal cofactor.


Subject(s)
Molecular Mimicry , Vitamin B 12 , Humans , Oxidation-Reduction , Adenosine Diphosphate/metabolism , Vitamin B 12/metabolism , Methylmalonyl-CoA Mutase/chemistry , Methylmalonyl-CoA Mutase/metabolism
3.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884459

ABSTRACT

Urinary tract infections (UTIs) represent a health problem of the first magnitude since they affect large segments of the population, cause increased mortality and comorbidity, and have a high incidence of relapse. Therefore, UTIs cause a major socioeconomic concern. Current antibiotic treatments have various limitations such as the appearance of resistance to antibiotics, nephrotoxicity, and side effects such as gastrointestinal problems including microbiota alterations that contribute to increasing antibiotic resistance. In this context, Itxasol© has emerged, approved as an adjuvant for the treatment of UTIs. Designed with biomimetic principles, it is composed of arbutin, umbelliferon, and N-acetyl cysteine. In this work, we review the activities of these three compounds concerning the changes they produce in the expression of bacterial genes and those related to inflammation as well as assess how they are capable of affecting the DNA of bacteria and fungi.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Bacterial Proteins/genetics , Urinary Tract Infections/microbiology , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Anti-Bacterial Agents/therapeutic use , Arbutin/pharmacology , Arbutin/therapeutic use , Bacteria/drug effects , Drug Combinations , Gene Expression Regulation, Bacterial/drug effects , Humans , Molecular Mimicry , Umbelliferones/pharmacology , Umbelliferones/therapeutic use , Urinary Tract Infections/drug therapy
4.
Sci Rep ; 11(1): 21848, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750393

ABSTRACT

Although mindfulness-based stress reduction (MBSR) improves cognitive function, the mechanism is not clear. In this study, people aged 65 years and older were recruited from elderly communities in Chitose City, Japan, and assigned to a non-MBSR group or a MBSR group. Before and after the intervention, the Japanese version of the Montreal Cognitive Assessment (MoCA-J) was administered, and blood samples were collected. Then, neuron-derived extracellular vesicles (NDEVs) were isolated from blood samples, and microRNAs, as well as the target mRNAs, were evaluated in NDEVs. A linear mixed model analysis showed significant effects of the MBSR x time interaction on the MoCA-J scores, the expression of miRNA(miR)-29c, DNA methyltransferase 3 alpha (DNMT3A), and DNMT3B in NDEVs. These results indicate that MBSR can improve cognitive function by increasing the expression of miR-29c and decreasing the expression of DNMT3A, as well as DNMT3B, in neurons. It was also found that intracerebroventricular injection of miR-29c mimic into 5xFAD mice prevented cognitive decline, as well as neuronal loss in the subiculum area, by down-regulating Dnmt3a  and Dnmt3b  in the hippocampus. The present study suggests that MBSR can prevent neuronal loss and cognitive impairment by increasing the neuronal expression of miR-29c.


Subject(s)
Cognition , Mindfulness/methods , Aged , Aged, 80 and over , Animals , Cognition/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/therapy , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A/genetics , DNA Methyltransferase 3A/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Female , Hippocampus/metabolism , Humans , Japan , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Mimicry , Neurons/metabolism , Up-Regulation , DNA Methyltransferase 3B
5.
Molecules ; 26(11)2021 May 24.
Article in English | MEDLINE | ID: mdl-34073781

ABSTRACT

Estrogenic molecules have been reported to regulate glucose homeostasis and may be beneficial for diabetes management. Here, we investigated the estrogenic effect of ß-sitosterol-3-O-D-glucopyranoside (BSD), isolated from the fruits of Cupressus sempervirens and monitored its ability to regulate glucose utilization in skeletal muscle cells. BSD stimulated ERE-mediated luciferase activity in both ERα and ERß-ERE luc expression system with greater response through ERß in HEK-293T cells, and induced the expression of estrogen-regulated genes in estrogen responsive MCF-7 cells. In silico docking and molecular interaction studies revealed the affinity and interaction of BSD with ERß through hydrophobic interaction and hydrogen bond pairing. Furthermore, prolonged exposure of L6-GLUT4myc myotubes to BSD raised the glucose uptake under basal conditions without affecting the insulin-stimulated glucose uptake, the effect associated with enhanced translocation of GLUT4 to the cell periphery. The BSD-mediated biological response to increase GLUT4 translocation was obliterated by PI-3-K inhibitor wortmannin, and BSD significantly increased the phosphorylation of AKT (Ser-473). Moreover, BSD-induced GLUT4 translocation was prevented in the presence of fulvestrant. Our findings reveal the estrogenic activity of BSD to stimulate glucose utilization in skeletal muscle cells via PI-3K/AKT-dependent mechanism.


Subject(s)
Glucose/metabolism , Molecular Mimicry , Muscle, Skeletal/metabolism , Phytoestrogens/pharmacology , Sitosterols/pharmacology , Glucose Transporter Type 4/metabolism , HEK293 Cells , Humans , MCF-7 Cells , Magnetic Resonance Spectroscopy , Mass Spectrometry , Muscle, Skeletal/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sitosterols/chemistry
6.
J Inorg Biochem ; 219: 111431, 2021 06.
Article in English | MEDLINE | ID: mdl-33798828

ABSTRACT

Oxidative stress that results from an imbalance between the concentrations of reactive species (RS) and antioxidant defenses is associated with many pathologies. Superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase are among the key enzymes that maintain the low nanomolar physiological concentrations of superoxide and hydrogen peroxide. The increase in the levels of these species and their progeny could have deleterious effects. In this context, chemists have developed SOD and CAT mimics to supplement them when cells are overwhelmed with oxidative stress. However, the beneficial activity of such molecules in cells depends not only on their intrinsic catalytic activities but also on their stability in biological context, their cell penetration and their cellular localization. We have employed cellular assays to characterize several compounds that possess SOD and CAT activities and have been frequently used in cellular and animal models. We used cellular assays that address SOD and CAT activities of the compounds. Finally, we determined the effect of compounds on the suppression of the inflammation in HT29-MD2 cells challenged by lipopolysaccharide. When the assay requires penetration inside cells, the SOD mimics Mn(III) meso-tetrakis(N-(2'-n-butoxyethyl)pyridinium-2-yl)porphyrin (MnTnBuOE-2-PyP5+) and Mn(II) dichloro[(4aR,13aR,17aR,21aR)-1,2,3,4,4a,5,6,12,13,13a,14,15,16,17,17a,18,19,20,21,21a-eicosahydro-11,7-nitrilo-7Hdibenzo[b,h] [1,4, 7,10] tetraazacycloheptadecine-κN5,κN13,κN18,κN21,κN22] (Imisopasem manganese, M40403, CG4419) were found efficacious at 10 µM, while Mn(II) chloro N-(phenolato)-N,N'-bis[2-(N-methyl-imidazolyl)methyl]-ethane-1,2-diamine (Mn1) requires an incubation at 100 µM. This study thus demonstrates that MnTnBuOE-2-PyP5+, M40403 and Mn1 were efficacious in suppressing inflammatory response in HT29-MD2 cells and such action appears to be related to their ability to enter the cells and modulate reactive oxygen species (ROS) levels.


Subject(s)
Catalase/metabolism , Manganese/metabolism , Organometallic Compounds/metabolism , Superoxide Dismutase/metabolism , Animals , Antioxidants/metabolism , Cell Line , Glutathione Peroxidase/metabolism , Humans , Hydrogen Peroxide/metabolism , Metalloporphyrins/metabolism , Molecular Mimicry , Oxidation-Reduction , Oxidative Stress , Porphyrins/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism
7.
Eur J Pharmacol ; 900: 174038, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33737008

ABSTRACT

Subarachnoid hemorrhage (SAH) due to rupture of an intracranial aneurysm leads to vasospasm resulting in delayed cerebral ischemia. Therapeutic options are currently limited to hemodynamic optimization and nimodipine, which have marginal clinical efficacy. Nitric oxide (NO) modulates cerebral blood flow through activation of the cGMP-Protein Kinase G (PKG) pathway. Our hypothesis is that SAH results in downregulation of signaling components in the NO-PKG pathway which could explain why treatments for vasospasm targeting this pathway lack efficacy and that treatment with a cell permeant phosphopeptide mimetic of downstream effector prevents delayed vasospasm after SAH. Using a rat endovascular perforation model, reduced levels of NO-PKG pathway molecules were confirmed. Additionally, it was determined that expression and phosphorylation of a PKG substrate: Vasodilator-stimulated phosphoprotein (VASP) was downregulated. A family of cell permeant phosphomimetic of VASP (VP) was wasdesigned and shown to have vasorelaxing property that is synergistic with nimodipine in intact vascular tissuesex vivo. Hence, treatment targeting the downstream effector of the NO signaling pathway, VASP, may bypass receptors and signaling elements leading to vasorelaxation and that treatment with VP can be explored as a therapeutic strategy for SAH induced vasospasm and ameliorate neurological deficits.


Subject(s)
Phosphopeptides/therapeutic use , Subarachnoid Hemorrhage/drug therapy , Vasodilator Agents/therapeutic use , Vasospasm, Intracranial/drug therapy , Animals , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cyclic GMP-Dependent Protein Kinases/drug effects , Down-Regulation , Drug Design , Drug Synergism , Microfilament Proteins/drug effects , Microfilament Proteins/metabolism , Molecular Mimicry , Nimodipine/pharmacology , Nitric Oxide/metabolism , Phosphopeptides/pharmacokinetics , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Subarachnoid Hemorrhage/metabolism , Swine , Vasodilator Agents/pharmacokinetics
8.
Int J Biol Macromol ; 159: 886-895, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32413470

ABSTRACT

The genus Cnidoscolus (Euphorbiaceae) is widely distributed in tropical areas. In the Northeast of Brazil, the species C. quercifolius is endemic and has been used in traditional medicine. In this study, a novel protein was isolated from C. quercifolius seeds and characterized by its molecular weight, primary structure, isoelectric point (pI), and carbohydrate content. The hypoglycemic activity of this protein was investigated by in vitro assay with the RIN-5F glucose-responsive cell line and in vivo test using alloxan-induced diabetic mice models. In addition, safe use of the protein was also investigated by cytotoxicity, hemagglutinating, and immunogenicity assays. The protein which was named Cq-IMP (Cnidoscolus quercifolius - Insulin Mimetic Protein) showed a single 11.18 KDa glycopolypeptide chain (16.4% of carbohydrates, m/m), pI of 8.0 and N-terminal sequence (TKDPELKQcKKQQKKqQQYDDDDKK) with similarity around 46-62% to sucrose binding protein-like and vicilin-like protein that was confirmed by mass spectrometry tryptic peptides analysis. Besides that, Cq-IMP presented anti-insulin antibody cross-reactivity as hypoglycemic activity in both in vitro and in vivo models. Additionally, it did not present any toxicity by methods tested. In conclusion, Cq-IMP is an insulin-mimetic protein, with a potent hypoglycemic activity and no toxicity showing great potential for therapeutic applications and drug development.


Subject(s)
Euphorbiaceae/chemistry , Glycoproteins/chemistry , Hypoglycemic Agents/chemistry , Insulin/chemistry , Molecular Mimicry , Plant Proteins/chemistry , Seeds/chemistry , Administration, Oral , Animals , Chromatography, Liquid , Glucose Tolerance Test , Glycoproteins/administration & dosage , Glycoproteins/isolation & purification , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/isolation & purification , Mice , Molecular Structure , Molecular Weight , Plant Proteins/administration & dosage , Plant Proteins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spectrum Analysis , alpha-Amylases/antagonists & inhibitors , alpha-Amylases/chemistry
9.
Microb Pathog ; 138: 103818, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31676366

ABSTRACT

Currently, the world health sector faces a big problem due to the increase of bacterial strains resistant to antibiotics. In 2017, the World Health Organization reported a list of resistant bacteria, among which Pseudomonas aeruginosa was present. This opportunistic pathogen is associated to nosocomial infections, and no effective vaccines against this bacterium have been found. Larrea divaricata Cav. (jarilla) is a shrub highly distributed in America and widely used in folk medicine. In our laboratory, cross-reactivity of antibodies obtained from the recognition of jarilla proteins against proteins from gram-negative bacteria has been demonstrated. The objective of this study was to study the cross-reactivity of anti-L. divaricata antibodies with P. aeruginosa extracellular proteins in order to find an innocuous prophylactic therapy against this nosocomial pathogen. We observed that antibodies generated by proteins from jarilla crude extract recognized antigenic determinants present in extracellular proteins of P. aeruginosa. However, further studies are needed to investigate the neutralizing capacity of these antibodies on the specific enzymatic proteins involved in the pathogenicity of this bacterium.


Subject(s)
Cross Reactions/immunology , Larrea/chemistry , Larrea/immunology , Molecular Mimicry , Plant Extracts/immunology , Plant Proteins/immunology , Pseudomonas aeruginosa/immunology , Animals , Antibodies/immunology , Enzyme-Linked Immunosorbent Assay , Immunity, Humoral , Immunoglobulin G/immunology , Larrea/metabolism , Mice , Plant Extracts/chemistry , Plant Proteins/chemistry , Plant Proteins/isolation & purification
10.
J Biochem Mol Toxicol ; 33(12): e22405, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31593333

ABSTRACT

Ankylosing spondylitis (AS) is a high disability and greatly destructive disease. In this study, we preliminarily studied the function and mechanism of bilobalide (BIL) on interleukin (IL)-17-induced inflammatory injury in ATDC5 cells. CCK-8 and migration assays were used to detect the functions of IL-7, BIL, and microRNA (miR)-125a on cell viability and migration. The miR-125a level was changed by transfection, and tested by real-time quantitative polymerase chain reaction. Additionally, Western blot tested the levels of inflammatory factors (IL-6 and tumor necrosis factor-α), matrix metalloproteinases (MMPs), and pathway-related proteins. Moreover, the enzyme-linked immunosorbent assay also was used to detect inflammatory factor levels. IL-7 was used to construct an inflammatory injury model in ATDC5 cells. Based on this, BIL inhibited IL-17-induced cell viability, migration, and expressions of inflammatory factors and MMPs. Furthermore, we found BIL negatively regulated miR-125a, and the miR-125a mimic could partly reverse the effects of BIL on IL-17-injury. Finally, we showed that BIL inhibited the c-Jun N-terminal kinase (JNK) and nuclear factor kappa B (NF-κB) pathways, and the miR-125a mimic had the opposite effect. BIL inhibited IL-17-induced inflammatory injury in ATDC5 cells by downregulation of miR-125a via JNK and NF-κB signaling pathways.


Subject(s)
Cyclopentanes/pharmacology , Down-Regulation/drug effects , Furans/pharmacology , Ginkgolides/pharmacology , Inflammation/chemically induced , Interleukin-17/pharmacology , MicroRNAs/metabolism , Plant Extracts/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Ginkgo biloba , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Matrix Metalloproteinases/metabolism , Mice , MicroRNAs/chemistry , MicroRNAs/genetics , Molecular Mimicry , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Spondylitis, Ankylosing/metabolism , Transfection
11.
Curr Protoc Immunol ; 126(1): e78, 2019 09.
Article in English | MEDLINE | ID: mdl-31483103

ABSTRACT

MicroRNAs (miRNAs) are short (19- to 25-nucleotide) noncoding RNA molecules that target mRNAs to repress gene expression and that play important roles in regulating many fundamental biological functions including cell differentiation, development, growth, and metabolism. They are well conserved in eukaryotic cells and are considered essential ancient elements of gene regulation. miRNA genes are transcribed by RNA polymerase II to generate primary miRNAs (pri-miRNAs), which are cleaved by microprocessor complex in the nucleus to generate stem-loop structures known as pre-miRNAs. Pre-miRNAs are translocated to the cytoplasm and cleaved by Dicer to form the mature miRNAs, which mediate mRNA degradation through their loading to the RNA-induced silencing complex (RISC) and binding to complementary sequences within target mRNAs to repress their translation by mRNA degradation and/or translation inhibition. Because ∼1900 miRNA genes are reported in the human genome, many associated with disease, appropriate methods to study miRNA expression and regulation under physiological and pathological conditions have become increasingly important to the study of many aspects of human biology, including immune regulation. As with small interfering RNA (siRNA), the mechanism of miRNA-mediated targeting has been used to develop miRNA-based therapeutics. For a complete and systematic analysis, it is critical to utilize a variety of different tools to analyze the expression of pri-mRNAs, pre-miRNAs, and mature miRNAs and characterize their targets both in vitro and in vivo. Such studies will facilitate future novel drug design and development. This unit provides six basic protocols for miRNA analysis, covering next-generation sequencing, quantitative real-time PCR (qRT-PCR), and digoxigenin-based expression analysis of pri-mRNAs, pre-miRNAs, and mature miRNAs; mapping of pri-miRNA and their cleavage sites by rapid amplification of cDNA ends (RACE); electrophoretic mobility shift assays (EMSAs) or biotin-based nonradioactive detection of miRNA-protein complexes (miRNPs); and functional analysis of miRNAs using miRNA mimics and inhibitors. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Biomimetics/methods , MicroRNAs/genetics , RNA, Messenger/genetics , Electrophoretic Mobility Shift Assay , Gene Expression , Gene Expression Regulation , Gene Silencing , High-Throughput Nucleotide Sequencing , Humans , Molecular Mimicry
12.
J Virol ; 93(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31092576

ABSTRACT

A key step in the Ebola virus (EBOV) replication cycle involves conformational changes in viral glycoprotein 2 (GP2) which facilitate host-viral membrane fusion and subsequent release of the viral genome. Ebola GP2 plays a critical role in virus entry and has similarities in mechanism and structure to the HIV gp41 protein for which inhibitors have been successfully developed. In this work, a putative binding pocket for the C-terminal heptad repeat in the N-terminal heptad repeat trimer was targeted for identification of small molecules that arrest EBOV-host membrane fusion. Two computational structure-based virtual screens of ∼1.7 M compounds were performed (DOCK program) against a GP2 five-helix bundle, resulting in 165 commercially available compounds purchased for experimental testing. Based on assessment of inhibitory activity, cytotoxicity, and target specificity, four promising candidates emerged with 50% inhibitory concentration values in the 3 to 26 µM range. Molecular dynamics simulations of the two most potent candidates in their DOCK-predicted binding poses indicate that the majority of favorable interactions involve seven highly conserved residues that can be used to guide further inhibitor development and refinement targeting EBOV.IMPORTANCE The most recent Ebola virus disease outbreak, from 2014 to 2016, resulted in approximately 28,000 individuals becoming infected, which led to over 12,000 causalities worldwide. The particularly high pathogenicity of the virus makes paramount the identification and development of promising lead compounds to serve as inhibitors of Ebola infection. To limit viral load, the virus-host membrane fusion event can be targeted through the inhibition of the class I fusion glycoprotein of Ebolavirus In the current work, several promising small-molecule inhibitors that target the glycoprotein GP2 were identified through systematic application of structure-based computational and experimental drug design procedures.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Molecular Mimicry , Viral Envelope Proteins/antagonists & inhibitors , Virus Internalization/drug effects , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cell Line , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding
14.
Eur J Med Chem ; 163: 28-36, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30503941

ABSTRACT

Phytoestrogens are class of natural compounds that shares structural similarity with estrogen and has the capacity to alter the fertilization in mammals. Till early 1990s, the natural phytoestrogens as well as their synthetic analogues were explored for their fertility modulating activity. During late 1990s, two findings renewed the interest on phytoestrogens as means to control hormone induced cancer: (i) revelation of overexpression of CYP1B1 in breast & ovarian cancer and (ii) protection offered by alphanapthoflavone (ANF) against hormone induced cancer. The objective of the review is to summarize the CYP1B1 inhibitory activity of phytoestrogens and their synthetic analogues reported till date. This review is an attempt to classify phytoestrogens and their synthetic analogues on their chemical architecture rather than simply by their chemical class (flavones, stilbenes etc.). This provides a broader sense to cluster many chemical classes under a particular chemical architecture/framework. Accordingly, we divided the phytoestrogen into three different classes based on two aryl groups (Ar) separated by linker (X), which may be either cyclic (c) or linear (l). The number in subscript to X denotes number of atoms: (i) Ar-cX4-Ar, (ii) Ar-lX3-Ar and (iii) Ar-lX2-Ar. This provides an opportunity to cluster flavones, quinolines and quinazolinones under Ar-cX4-Ar class, while biphenyl ureas and chalcones under Ar-lX3-Ar class. We believe in coming years many chemical scaffolds may be clustered under this framework.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Neoplasms/drug therapy , Phytoestrogens/chemistry , Animals , Antineoplastic Agents, Phytogenic/classification , Classification , Cluster Analysis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/classification , Humans , Molecular Mimicry , Neoplasms/chemically induced
15.
Leuk Lymphoma ; 60(4): 1062-1070, 2019 04.
Article in English | MEDLINE | ID: mdl-30277117

ABSTRACT

The interaction between CD40, and its ligand, CD154, is essential for the development of humoral and cellular immune responses. The selective inhibition or activation of this pathway forms the basis for the development of new therapeutics against immunologically based diseases and malignancies. We are developing a gene fusion of Salmonella typhi OmpC protein expressing the CD154 Tyr140-Ser-149 amino acid strand. This OmpC-CD154 binds CD40 and activates B cells. In this study, we demonstrate that OmpC-CD154p treatment inhibits cell growth, proliferation and induced apoptosis in the B-NHL cell lines Raji and Ramos. The Bcl-2 family proteins were regulated and the Bcl-6 and YY1 oncoproteins were inhibited. p38 MAPK activation is an important mechanism underlying the effect on proliferation and apoptosis mediated by this fusion protein. This study establishes a basis for the possible use of fusion protein OmpC-CD154 as an alternative treatment for B-NHL.


Subject(s)
Caspases/metabolism , Peptides/pharmacology , Proto-Oncogene Proteins c-bcl-6/metabolism , YY1 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CD40 Antigens/metabolism , CD40 Ligand/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Molecular Mimicry , Peptides/chemistry , Porins/chemistry
16.
Zhonghua Nan Ke Xue ; 24(6): 533-539, 2018 06.
Article in Chinese | MEDLINE | ID: mdl-30173460

ABSTRACT

Objective: To investigate the effect of Qilan Capsules (QLC) on the expressions of the related proteins HIF-1α, VEGF-α, EphA2 and MMP-1 in the formation of vasculogenic mimicry (VM) in prostate cancer. METHODS: Prostate cancer PC-3 cells were cultured, transfected with siRNA, and divided into eight groups, blank control, HIF-1α siRNA, VEGF-α siRNA, EphA2 siRNA, QLC intervention, QLC + HIF-1α siRNA, QLC + VEGF-α siRNA, and QLC + EphA2 siRNA. The expressions of the HIF-1α, VEGF-α and EphA2 proteins in the pathway of VEGF were determined by Western blot. RESULTS: Compared with the blank control group, the expression of HIF-1α was evidently decreased in the HIF-lα siRNA and QLC + HIF-lα siRNA groups (0.624 7 ± 0.042 8 vs 0.032 8 ± 0.002 5 and 0.036 8 ± 0.018 1, P < 0.05), so were that of VEGF-α in the VEGF-α siRNA and QLC + VEGF-α siRNA groups (0.068 9 ± 0.005 1 vs 0.016 9 ± 0.000 7 and 0.010 9 ± 0.000 8, P < 0.05), that of EphA2 in the EphA2 siRNA and QLC + EphA2 siRNA groups though with no statistically significant difference (0.1684 ± 0.0126 vs 0.134 5 ± 0.028 6 and 0.165 4 ± 0.039 8, P > 0.05), and that of MMP-1 in the HIF-lα siRNA, VEGF-α siRNA and EphA2 siRNA groups (1.696 1 ± 0.152 7 vs 0.435 9 ± 0.036 9, 0.198 7 ± 0.009 0 and 0.0218 ± 0.000 7, P < 0.05). CONCLUSIONS: Qilan Capsules can suppress VM formation in prostate cancer by inhibiting the expressions of HIF-1α, VEGF-α and MMP-1, which plays a role in the clinical treatment of prostate cancer by checking the growth and development of the blood supply system in the tumor tissue.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Matrix Metalloproteinase 1/metabolism , Prostatic Neoplasms/metabolism , Receptor, EphA2/metabolism , Vascular Endothelial Growth Factor A/metabolism , Capsules , Humans , Male , Molecular Mimicry , RNA, Small Interfering/metabolism , Transfection
17.
J Cell Biochem ; 119(10): 8623-8635, 2018 11.
Article in English | MEDLINE | ID: mdl-30058740

ABSTRACT

Non-small-cell lung cancer (NSCLC) appears to be a significant threat to public health worldwide. MicroRNAs have been identified as significant regulators for the development of NSCLC. Previous reports have suggested that hsa-mir-485-5p is dysregulated in various cancers. RXRα, as a kind of nuclear receptor, is an effective target of cancer treatment. Cancer stem cells (CSCs) are recognized as the main cause for tumor metastasis, recurrence, and chemotherapy resistance. However, the mechanism by which hsa-mir-485-5p and RXRα modulate CSCs in NSCLC remains unknown. Here, we found that hsa-mir-485-5p was decreased in serum samples from patients with NSCLC and NSCLC cells. Meanwhile, epigallocatechin-3-gallate (EGCG), an effective anticancer compound extracted from green tea, can enhance hsa-mir-485-5p expression. Hsa-mir-485-5p mimics markedly inhibited NSCLC cell growth and induced cell apoptosis. However, inhibition of hsa-mir-485-5p significantly enriched CSC-like traits. Moreover, bioinformatics analysis predicted the binding correlation between hsa-mir-485-5p and RXRα, which was confirmed by a dual-luciferase reporter assay. We observed that RXRα was increased in NSCLC and EGCG could inhibit RXRα levels dose dependently. In addition, RXRα upregulation or activation expanded the CSC-like properties of NSCLC cells, whereas RXRα inhibition or inactivation could exert a reverse phenomenon. Consistently, in vivo experiments also validated that EGCG could repress the CSC-like characteristics by modulating the hsa-mir-485-5p/RXRα axis. Our findings may reveal a novel molecular mechanism for the treatment of NSCLC.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Catechin/analogs & derivatives , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Retinoid X Receptor alpha/metabolism , A549 Cells , Analysis of Variance , Animals , Catechin/therapeutic use , Down-Regulation , Gene Knockdown Techniques , Gene Silencing , HEK293 Cells , Humans , Mice , Mice, Nude , MicroRNAs/chemistry , MicroRNAs/genetics , Molecular Mimicry/genetics , Retinoid X Receptor alpha/genetics , Signal Transduction/drug effects , Transfection , Up-Regulation , Xenograft Model Antitumor Assays
18.
Proc Natl Acad Sci U S A ; 115(12): E2819-E2828, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29507247

ABSTRACT

Treatment of bacterial infections is becoming a serious clinical challenge due to the global dissemination of multidrug antibiotic resistance, necessitating the search for alternative treatments to disarm the virulence mechanisms underlying these infections. Uropathogenic Escherichia coli (UPEC) employs multiple chaperone-usher pathway pili tipped with adhesins with diverse receptor specificities to colonize various host tissues and habitats. For example, UPEC F9 pili specifically bind galactose or N-acetylgalactosamine epitopes on the kidney and inflamed bladder. Using X-ray structure-guided methods, virtual screening, and multiplex ELISA arrays, we rationally designed aryl galactosides and N-acetylgalactosaminosides that inhibit the F9 pilus adhesin FmlH. The lead compound, 29ß-NAc, is a biphenyl N-acetyl-ß-galactosaminoside with a Ki of ∼90 nM, representing a major advancement in potency relative to the characteristically weak nature of most carbohydrate-lectin interactions. 29ß-NAc binds tightly to FmlH by engaging the residues Y46 through edge-to-face π-stacking with its A-phenyl ring, R142 in a salt-bridge interaction with its carboxylate group, and K132 through water-mediated hydrogen bonding with its N-acetyl group. Administration of 29ß-NAc in a mouse urinary tract infection (UTI) model significantly reduced bladder and kidney bacterial burdens, and coadministration of 29ß-NAc and mannoside 4Z269, which targets the type 1 pilus adhesin FimH, resulted in greater elimination of bacteria from the urinary tract than either compound alone. Moreover, FmlH specifically binds healthy human kidney tissue in a 29ß-NAc-inhibitable manner, suggesting a key role for F9 pili in human kidney colonization. Thus, these glycoside antagonists of FmlH represent a rational antivirulence strategy for UPEC-mediated UTI treatment.


Subject(s)
Adhesins, Escherichia coli/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Urinary Tract Infections/microbiology , Adhesins, Escherichia coli/metabolism , Animals , Crystallography, X-Ray , Drug Evaluation, Preclinical/methods , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Female , Galactosides/chemical synthesis , Galactosides/chemistry , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/microbiology , Ligands , Mice, Inbred C3H , Molecular Docking Simulation , Molecular Mimicry , Urinary Tract Infections/drug therapy , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/pathogenicity
19.
J Bioinform Comput Biol ; 16(2): 1840007, 2018 04.
Article in English | MEDLINE | ID: mdl-29439644

ABSTRACT

An integrated computational approach to in silico drug design was used to identify novel HIV-1 fusion inhibitor scaffolds mimicking broadly neutralizing antibody (bNab) 10E8 targeting the membrane proximal external region (MPER) of the HIV-1 gp41 protein. This computer-based approach included (i) generation of pharmacophore models representing 3D-arrangements of chemical functionalities that make bNAb 10E8 active towards the gp41 MPER segment, (ii) shape and pharmacophore-based identification of the 10E8-mimetic candidates by a web-oriented virtual screening platform pepMMsMIMIC, (iii) high-throughput docking of the identified compounds with the gp41 MPER peptide, and (iv) molecular dynamics simulations of the docked structures followed by binding free energy calculations. As a result, eight hits-able to mimic pharmacophore properties of bNAb 10E8 by specific and effective interactions with the MPER region of the HIV-1 protein gp41 were selected as the most probable 10E8-mimetic candidates. Similar to 10E8, the predicted compounds target the critically important residues of a highly conserved hinge region of the MPER peptide that provides a conformational flexibility necessary for its functioning in cell-virus membrane fusion process. In light of the data obtained, the identified small molecules may present promising HIV-1 fusion inhibitor scaffolds for the design of novel potent antiviral drugs.


Subject(s)
Antibodies, Neutralizing/chemistry , Computational Biology/methods , HIV Envelope Protein gp41/immunology , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/pharmacology , Computer Simulation , Drug Evaluation, Preclinical/methods , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/metabolism , HIV Fusion Inhibitors/metabolism , Hydrogen Bonding , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Mimicry , Tryptophan/chemistry
20.
National Journal of Andrology ; (12): 533-539, 2018.
Article in Chinese | WPRIM | ID: wpr-689695

ABSTRACT

<p><b>Objective</b>To investigate the effect of Qilan Capsules (QLC) on the expressions of the related proteins HIF-1α, VEGF-α, EphA2 and MMP-1 in the formation of vasculogenic mimicry (VM) in prostate cancer.</p><p><b>METHODS</b>Prostate cancer PC-3 cells were cultured, transfected with siRNA, and divided into eight groups, blank control, HIF-1α siRNA, VEGF-α siRNA, EphA2 siRNA, QLC intervention, QLC + HIF-1α siRNA, QLC + VEGF-α siRNA, and QLC + EphA2 siRNA. The expressions of the HIF-1α, VEGF-α and EphA2 proteins in the pathway of VEGF were determined by Western blot.</p><p><b>RESULTS</b>Compared with the blank control group, the expression of HIF-1α was evidently decreased in the HIF-lα siRNA and QLC + HIF-lα siRNA groups (0.624 7 ± 0.042 8 vs 0.032 8 ± 0.002 5 and 0.036 8 ± 0.018 1, P < 0.05), so were that of VEGF-α in the VEGF-α siRNA and QLC + VEGF-α siRNA groups (0.068 9 ± 0.005 1 vs 0.016 9 ± 0.000 7 and 0.010 9 ± 0.000 8, P < 0.05), that of EphA2 in the EphA2 siRNA and QLC + EphA2 siRNA groups though with no statistically significant difference (0.1684 ± 0.0126 vs 0.134 5 ± 0.028 6 and 0.165 4 ± 0.039 8, P > 0.05), and that of MMP-1 in the HIF-lα siRNA, VEGF-α siRNA and EphA2 siRNA groups (1.696 1 ± 0.152 7 vs 0.435 9 ± 0.036 9, 0.198 7 ± 0.009 0 and 0.0218 ± 0.000 7, P < 0.05).</p><p><b>CONCLUSIONS</b>Qilan Capsules can suppress VM formation in prostate cancer by inhibiting the expressions of HIF-1α, VEGF-α and MMP-1, which plays a role in the clinical treatment of prostate cancer by checking the growth and development of the blood supply system in the tumor tissue.</p>


Subject(s)
Humans , Male , Capsules , Drugs, Chinese Herbal , Pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit , Metabolism , Matrix Metalloproteinase 1 , Metabolism , Molecular Mimicry , Prostatic Neoplasms , Metabolism , RNA, Small Interfering , Metabolism , Receptor, EphA2 , Metabolism , Transfection , Vascular Endothelial Growth Factor A , Metabolism
SELECTION OF CITATIONS
SEARCH DETAIL