Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
mSphere ; 4(4)2019 07 03.
Article in English | MEDLINE | ID: mdl-31270174

ABSTRACT

Penicillin-binding proteins (PBPs) are essential for bacterial cell wall biosynthesis, and several are clinically validated antibacterial targets of ß-lactam antibiotics. We identified mutations in the mrdA gene encoding the PBP2 protein in two Escherichia coliblaNDM-1 clinical isolates that reduce susceptibility to carbapenems and to the intrinsic antibacterial activity of a diazabicyclooctane (DBO) PBP2 and ß-lactamase inhibitor. These mutations coexisted with previously described mutations in ftsI (encoding PBP3) that reduce susceptibility to monobactams, penicillins, and cephalosporins. Clinical exposure to ß-lactams is driving the emergence of multifactorial resistance that may impact the therapeutic usefulness of existing antibacterials and novel compounds that target PBPs.IMPORTANCE Emerging antibacterial resistance is a consequence of the continued use of our current antibacterial therapies, and it is limiting their utility, especially for infections caused by multidrug-resistant isolates. ß-Lactams have enjoyed extensive clinical success, but their broad usage is linked to perhaps the most extensive and progressive example of resistance development for any antibacterial scaffold. In Gram-negative pathogens, this largely involves constant evolution of new ß-lactamases able to degrade successive generations of this scaffold. In addition, more recently, alterations in the targets of these compounds, penicillin-binding proteins (PBPs), are being described in clinical isolates, which often also have multiple ß-lactamases. This study underscores the multifactorial nature of ß-lactam resistance by uncovering alterations of PBP2 that reduce susceptibility to carbapenems in E. coli clinical isolates that also have alterations of PBP3 and express the NDM-1 ß-lactamase. The changes in PBP2 also reduced susceptibility to the intrinsic antibacterial activity of some diazabicyclooctane (DBO) compounds that can target PBP2. This may have implications for the development and use of the members of this relatively newer scaffold that are inhibitors of PBP2 in addition to their inhibition of serine-ß-lactamases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azabicyclo Compounds/pharmacology , Carbapenems/pharmacology , Escherichia coli Proteins/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Penicillin-Binding Proteins/genetics , Peptidoglycan Glycosyltransferase/genetics , Azabicyclo Compounds/chemistry , Microbial Sensitivity Tests , Mutation , beta-Lactam Resistance , beta-Lactamases/genetics , beta-Lactams/pharmacology
2.
ChemMedChem ; 12(20): 1687-1692, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28881459

ABSTRACT

Conformationally constrained tetracyclic fluoroquinolones (FQs) were synthesized and profiled for their microbiological spectrum. The installation of a seven-membered ring between the pyrrolidine substituents and the C8 position on the FQ core scaffold resulted in a remarkable enhancement of microbiological potency toward both Gram-positive and Gram-negative bacteria. Focused optimization of seven-membered ring composition, stereochemistry, and amine placement led to the discovery of the two lead compounds that were selected for further progression.


Subject(s)
Fluoroquinolones/chemical synthesis , Fluoroquinolones/pharmacology , Tetracyclines/chemical synthesis , Tetracyclines/pharmacology , Acinetobacter baumannii/drug effects , Microbial Sensitivity Tests , Molecular Structure , Pseudomonas aeruginosa/drug effects , Structure-Activity Relationship
3.
Neuro Oncol ; 9(3): 259-70, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17522330

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a proinvasive and immunosuppressive cytokine that plays a major role in the malignant phenotype of gliomas. One novel strategy of disabling TGF-beta activity in gliomas is to disrupt the signaling cascade at the level of the TGF-beta receptor I (TGF-betaRI) kinase, thus abrogating TGF-beta-mediated invasiveness and immune suppression. SX-007, an orally active, small-molecule TGF-betaRI kinase inhibitor, was evaluated for its therapeutic potential in cell culture and in an in vivo glioma model. The syngeneic, orthotopic glioma model SMA-560 was used to evaluate the efficacy of SX-007. Cells were implanted into the striatum of VM/Dk mice. Dosing began three days after implantation and continued until the end of the study. Efficacy was established by assessing survival benefit. SX-007 dosed at 20 mg/kg p.o. once daily (q.d.) modulated TGF-beta signaling in the tumor and improved the median survival. Strikingly, approximately 25% of the treated animals were disease-free at the end of the study. Increasing the dose to 40 mg/kg q.d. or 20 mg/kg twice daily did not further improve efficacy. The data suggest that SX-007 can exert a therapeutic effect by reducing TGF-beta-mediated invasion and reversing immune suppression. SX-007 modulates the TGF-beta signaling pathway and is associated with improved survival in this glioma model. Survival benefit is due to reduced tumor invasion and reversal of TGF-beta-mediated immune suppression, allowing for rejection of the tumor. Together, these results suggest that treatment with a TGF-betaRI inhibitor may be useful in the treatment of glioblastoma.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Immunologic Surveillance/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Transforming Growth Factor beta/drug effects , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/immunology , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glioma/immunology , Humans , Immunoblotting , Immunohistochemistry , Mice , Receptor, Transforming Growth Factor-beta Type I , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Xenograft Model Antitumor Assays
4.
Cancer Res ; 64(15): 5200-11, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15289325

ABSTRACT

Transforming growth factor-beta (TGF-beta) suppresses tumor formation by blocking cell cycle progression and maintaining tissue homeostasis. In pancreatic carcinomas, this tumor suppressive activity is often lost by inactivation of the TGF-beta-signaling mediator, Smad4. We found that human pancreatic carcinoma cell lines that have undergone deletion of MADH4 constitutively expressed high endogenous levels of phosphorylated receptor-associated Smad proteins (pR-Smad2 and pR-Smad3), whereas Smad4-positive lines did not. These elevated pR-Smad levels could not be attributed to a decreased dephosphorylation rate nor to increased expression of TGF-beta type I (TbetaR-I) or type II (TbetaR-II) receptors. Although minimal amounts of free bioactive TGF-beta1 and TGF-beta2 were detected in conditioned medium, treatment with a pan-specific (but not a TGF-beta3 specific) TGF-beta-neutralizing antibody and with anti-alpha(V)beta(6) integrin antibody decreased steady-state pSmad2 levels and activation of a TGF-beta-inducible reporter gene in neighboring cells, respectively. Thus, activation of TGF-beta at the cell surface was responsible for the increased autocrine endogenous and paracrine signaling. Blocking TbetaR-I activity using a selective kinase inhibitor (SD-093) strongly decreased the in vitro motility and invasiveness of the pancreatic carcinoma cells without affecting their growth characteristics, morphology, or the subcellular distribution of E-cadherin and F-actin. Moreover, exogenous TGF-beta strongly stimulated in vitro invasiveness of BxPC-3 cells, an effect that could also be blocked by SD-093. Thus, the motile and invasive properties of Smad4-deficient pancreatic cancer cells are at least partly driven by activation of endogenous TGF-beta signaling. Therefore, targeting the TbetaR-I kinase represents a potentially powerful novel therapeutic approach for the treatment of this disease.


Subject(s)
Activin Receptors, Type I/metabolism , DNA-Binding Proteins/deficiency , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Trans-Activators/deficiency , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cadherins/metabolism , Cell Movement , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Gene Deletion , Humans , Integrins/antagonists & inhibitors , Integrins/immunology , Integrins/metabolism , Pancreatic Neoplasms/pathology , Phenotype , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Smad2 Protein , Smad3 Protein , Smad4 Protein , Trans-Activators/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , Transforming Growth Factor beta3 , Tumor Cells, Cultured , Wound Healing
5.
Biochem Pharmacol ; 68(1): 41-50, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15183116

ABSTRACT

Transforming growth factor (TGFbeta) is a 25-kDa dimeric polypeptide that plays a key role in a variety of physiological processes and disease states. Blocking TGFbeta signaling represents a potentially powerful and conceptually novel approach to the treatment of disorders in which the signaling pathway is constitutively activated, such as cancer, chronic inflammation with fibrosis and select immune disorders. In this paper, we describe the biological properties of a novel series of quinazoline-derived inhibitors of the type I transforming growth factor receptor kinase (TbetaKIs) that bind to the ATP-binding site and keep the kinase in its inactive conformation. These compounds effectively inhibited TGFbeta-induced Smad2 phosphorylation in cultured cells in vitro with an IC(50) between 20 and 300 nM. Moreover, TbetaKIs were able to broadly block TGFbeta-induced reporter gene activation. Finally, TbetaKIs inhibited TGFbeta-mediated growth inhibition of normal murine mammary epithelial cells (NMuMG) and mink lung epithelial cells (Mv1Lu), and TGFbeta-induced epithelial-mesenchymal transdifferentiation (EMT) of NMuMG cells. Thus, these chemical TbetaKIs have the potential to be further developed as anti-cancer and -fibrosis agents. In addition, they represent valuable new tools for dissecting the biochemical mechanisms of TGFbeta signal transduction and understanding the role of TGFbeta signaling pathways in different physiological and disease processes.


Subject(s)
Peptides/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , DNA-Binding Proteins/metabolism , Drug Interactions , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Mice , Phosphorylation/drug effects , Signal Transduction/physiology , Smad2 Protein , Trans-Activators/metabolism , Transcriptional Activation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...