Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Karen Dorso is active.

Publication


Featured researches published by Karen Dorso.


Nature | 2006

Platensimycin is a selective FabF inhibitor with potent antibiotic properties

Jun Wang; Stephen M. Soisson; Katherine Young; Wesley L. Shoop; Srinivas Kodali; Andrew Galgoci; Ronald E. Painter; Gopalakrishnan Parthasarathy; Yui S. Tang; Richard D. Cummings; Sookhee Ha; Karen Dorso; Mary Motyl; Hiranthi Jayasuriya; John G. Ondeyka; Kithsiri Herath; Chaowei Zhang; Lorraine D. Hernandez; John J. Allocco; Angela Basilio; José R. Tormo; Olga Genilloud; Francisca Vicente; Fernando Pelaez; Lawrence F. Colwell; Sang Ho Lee; Bruce Michael; Thomas J. Felcetto; Charles Gill; Lynn L. Silver

Bacterial infection remains a serious threat to human lives because of emerging resistance to existing antibiotics. Although the scientific community has avidly pursued the discovery of new antibiotics that interact with new targets, these efforts have met with limited success since the early 1960s. Here we report the discovery of platensimycin, a previously unknown class of antibiotics produced by Streptomyces platensis. Platensimycin demonstrates strong, broad-spectrum Gram-positive antibacterial activity by selectively inhibiting cellular lipid biosynthesis. We show that this anti-bacterial effect is exerted through the selective targeting of β-ketoacyl-(acyl-carrier-protein (ACP)) synthase I/II (FabF/B) in the synthetic pathway of fatty acids. Direct binding assays show that platensimycin interacts specifically with the acyl-enzyme intermediate of the target protein, and X-ray crystallographic studies reveal that a specific conformational change that occurs on acylation must take place before the inhibitor can bind. Treatment with platensimycin eradicates Staphylococcus aureus infection in mice. Because of its unique mode of action, platensimycin shows no cross-resistance to other key antibiotic-resistant strains tested, including methicillin-resistant S. aureus, vancomycin-intermediate S. aureus and vancomycin-resistant enterococci. Platensimycin is the most potent inhibitor reported for the FabF/B condensing enzymes, and is the only inhibitor of these targets that shows broad-spectrum activity, in vivo efficacy and no observed toxicity.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Discovery of platencin, a dual FabF and FabH inhibitor with in vivo antibiotic properties.

Jun Wang; Srinivas Kodali; Sang Ho Lee; Andrew Galgoci; Ronald E. Painter; Karen Dorso; Fred Racine; Mary Motyl; Lorraine D. Hernandez; Elizabeth Tinney; Steven L. Colletti; Kithsiri Herath; Richard D. Cummings; Oscar Salazar; Ignacio González; Angela Basilio; Francisca Vicente; Olga Genilloud; Fernando Pelaez; Hiranthi Jayasuriya; Katherine Young; Doris F. Cully; Sheo B. Singh

Emergence of bacterial resistance is a major issue for all classes of antibiotics; therefore, the identification of new classes is critically needed. Recently we reported the discovery of platensimycin by screening natural product extracts using a target-based whole-cell strategy with antisense silencing technology in concert with cell free biochemical validations. Continued screening efforts led to the discovery of platencin, a novel natural product that is chemically and biologically related but different from platensimycin. Platencin exhibits a broad-spectrum Gram-positive antibacterial activity through inhibition of fatty acid biosynthesis. It does not exhibit cross-resistance to key antibiotic resistant strains tested, including methicillin-resistant Staphylococcus aureus, vancomycin-intermediate S. aureus, and vancomycin-resistant Enterococci. Platencin shows potent in vivo efficacy without any observed toxicity. It targets two essential proteins, β-ketoacyl-[acyl carrier protein (ACP)] synthase II (FabF) and III (FabH) with IC50 values of 1.95 and 3.91 μg/ml, respectively, whereas platensimycin targets only FabF (IC50 = 0.13 μg/ml) in S. aureus, emphasizing the fact that more antibiotics with novel structures and new modes of action can be discovered by using this antisense differential sensitivity whole-cell screening paradigm.


Journal of Biological Chemistry | 2003

Discovery of a small molecule that inhibits cell division by blocking FtsZ, a novel therapeutic target of antibiotics

Jun Wang; Andrew Galgoci; Srinivas Kodali; Kithsiri Herath; Hiranthi Jayasuriya; Karen Dorso; Francisca Vicente; Antonio Gonzalez; Doris F. Cully; David Bramhill; Sheo B. Singh

The emergence of bacterial resistance to antibiotics is a major health problem and, therefore, it is critical to develop new antibiotics with novel modes of action. FtsZ, a tubulin-like GTPase, plays an essential role in bacterial cell division, and its homologs are present in almost all eubacteria and archaea. During cell division, FtsZ forms polymers in the presence of GTP that recruit other division proteins to make the cell division apparatus. Therefore, inhibition of FtsZ polymerization will prevent cells from dividing, leading to cell death. Using a fluorescent FtsZ polymerization assay, the screening of >100,000 extracts of microbial fermentation broths and plants followed by fractionation led to the identification of viriditoxin, which blocked FtsZ polymerization with an IC50 of 8.2 μg/ml and concomitant GTPase inhibition with an IC50 of 7.0 μg/ml. That the mode of antibacterial action of viriditoxin is via inhibition of FtsZ was confirmed by the observation of its effects on cell morphology, macromolecular synthesis, DNA-damage response, and increased minimum inhibitory concentration as a result of an increase in the expression of the FtsZ protein. Viriditoxin exhibited broad-spectrum antibacterial activity against clinically relevant Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococci, without affecting the viability of eukaryotic cells.


Chemistry & Biology | 2009

Chemical genetic identification of peptidoglycan inhibitors potentiating carbapenem activity against methicillin-resistant Staphylococcus aureus.

Joann Huber; Robert G.K. Donald; Sang Ho Lee; Lisa Wang Jarantow; Michael J. Salvatore; Xin Meng; Ronald E. Painter; Russell Onishi; James Occi; Karen Dorso; Katherine Young; Young Whan Park; Stephen Skwish; Michael J. Szymonifka; Tim S. Waddell; Lynn Miesel; John W. Phillips; Terry Roemer

Methicillin-resistant Staphylococcus aureus (MRSA) is a major nosocomial and community-acquired pathogen for which few existing antibiotics are efficacious. Here we describe two structurally related synthetic compounds that potentiate beta-lactam activity against MRSA. Genetic studies indicate that these agents target SAV1754 based on the following observations: (i) it has a unique chemical hypersensitivity profile, (ii) overexpression or point mutations are sufficient to confer resistance, and (iii) genetic inactivation phenocopies the potentiating effect of these agents in combination with beta-lactams. Further, we demonstrate these agents inhibit peptidoglycan synthesis. Because SAV1754 is essential for growth and structurally related to the recently reported peptidoglycan flippase of Escherichia coli, we speculate it performs an analogous function in S. aureus. These results suggest that SAV1754 inhibitors might possess therapeutic potential alone, or in combination with beta-lactams to restore MRSA efficacy.


Journal of the American Chemical Society | 2008

Isolation, structure, and antibacterial activity of philipimycin, a thiazolyl peptide discovered from Actinoplanes philippinensis MA7347

Chaowei Zhang; James Occi; Prakash S. Masurekar; John F. Barrett; Deborah L. Zink; Scott K. Smith; Russell Onishi; Sookhee Ha; Oscar Salazar; Olga Genilloud; Angela Basilio; Francisca Vicente; Charles Gill; Emily Hickey; Karen Dorso; Mary Motyl; Sheo B. Singh

Bacterial resistance to antibiotics, particularly to multiple drug resistant antibiotics, is becoming cause for significant concern. The only really viable course of action is to discover new antibiotics with novel mode of actions. Thiazolyl peptides are a class of natural products that are architecturally complex potent antibiotics but generally suffer from poor solubility and pharmaceutical properties. To discover new thiazolyl peptides potentially with better desired properties, we designed a highly specific assay with a pair of thiazomycin sensitive and resistant strains of Staphylococcus aureus, which led to the discovery of philipimycin, a new thiazolyl peptide glycoside. It was isolated along with an acid-catalyzed degradation product by bioassay-guided fractionation. Structure of both compounds was elucidated by extensive application of 2D NMR, 1D TOCSY, and HRESIFT-MS/MS. Both compounds showed strong antibacterial activities against gram-positive bacteria including MRSA and exhibited MIC values ranging from 0.015 to 1 microg/mL. Philipimycin was significantly more potent than the degradation product. Both compounds showed selective inhibition of protein synthesis, indicating that they targeted the ribosome. Philipimycin was effective in vivo in a mouse model of S. aureus infection exhibiting an ED50 value of 8.4 mg/kg. The docking studies of philipimycin suggested that a part of the molecule interacts with the ribosome and another part with Pro23, Pro22, and Pro26 of L11 protein, which helped in explaining the differential of activities between the sensitive and resistant strains. The design and execution of the bioassay, the isolation, structure, in vitro and in vivo antibacterial activity, and docking studies of philipimycin and its degradation product are described.


The Journal of Antibiotics | 2007

Antibacterial evaluations of thiazomycin- a potent thiazolyl peptide antibiotic from Amycolatopsis fastidiosa.

Sheo B. Singh; James Occi; Hiranthi Jayasuriya; Kithsiri Herath; Mary Motyl; Karen Dorso; Charles Gill; Emily Hickey; Karen M. Overbye; John F. Barrett; Prakash S. Masurekar

Thiazomycin is a novel thiazolyl peptide closely related to nocathiacin I. It was isolated from Amycolatopsis fastidiosa by chemical and biological screening. Thiazomycin showed highly potent bactericidal activity against Gram-positive pathogens (MIC range 0.002~0.064 μg/ml) and did not show cross-resistance to clinically relevant antibiotic classes such as β-lactams, vancomycin, oxazolidinone and quinolones. It was highly efficacious against Staphylococcus aureus infection in mice exhibiting an ED99 value of 0.15 mg/kg by subcutaneous administration. It inhibited bacterial growth by selective inhibition of protein synthesis and it was thought to interact with L11 protein and 23S rRNA of the 50S ribosome. Structurally, it possesses an oxazolidine ring in the amino-sugar residue that provides further opportunities for selective chemical modifications that are not feasible with other thiazolyl peptides. More importantly such a modification can potentially lead to semi-synthetic compounds that overcome problems that have hampered clinical development of this class of compounds. Despite its positive attributes, emergence of an unacceptable frequency of resistance poses significant challenges for further development of thiazomycin and this class of molecules for therapeutic use.


The Journal of Antibiotics | 2009

Discovery of okilactomycin and congeners from Streptomyces scabrisporus by antisense differential sensitivity assay targeting ribosomal protein S4.

Chaowei Zhang; John G. Ondeyka; Deborah L. Zink; Angela Basilio; Francisca Vicente; Oscar Salazar; Olga Genilloud; Karen Dorso; Mary Motyl; Kevin M. Byrne; Sheo B. Singh

Protein synthesis inhibition is a highly successful target for developing clinically effective and safe antibiotics. There are several targets within the ribosomal machinery, and small ribosomal protein S4 (RPSD) is one of the newer targets. Screening of microbial extracts using antisense-sensitized rpsD Staphylococcus aureus strain led to isolation of okilactomycin and four new congeners from Streptomyces scabrisporus. The major compound, okilactomycin, was the most active, with a minimum detection concentration of 3–12 μg ml−1 against antisense assay, and showed an MIC of 4–16 μg ml−1 against Gram-positive bacteria, including S. aureus. The congeners were significantly less active in all assays, and all compounds showed a slight preferential inhibition of RNA synthesis over DNA and protein synthesis. Antisense technology, due to increased sensitivity, continues to yield new, even though weakly active, antibiotics.


Journal of Natural Products | 2009

Isolation, Structure, and Antibacterial Activities of Lucensimycins D−G, Discovered from Streptomyces lucensis MA7349 Using an Antisense Strategy⊥

Sheo B. Singh; Deborah L. Zink; Karen Dorso; Mary Motyl; Oscar Salazar; Angela Basilio; Francisca Vicente; Kevin M. Byrne; Sookhee Ha; Olga Genilloud

Bacterial resistance to existing antibiotics continues to grow, necessitating the discovery of new compounds of this type. Antisense-based whole-cell target-based screening is a new and highly sensitive antibiotic discovery approach that has led to a number of new natural product antibiotics. Screening with a rpsD-sensitized strain led to the discovery of a number of natural product polyketides from Streptomyces lucensis. Complete workup of the fermentation extract of this strain allowed for the isolation of seven new compounds, lucensimycins A-G (1-3, 4a, 5-7), with varying degrees of antibacterial activities. Lucensimycin E (5) exhibited the best activity and showed MIC values of 32 microg/mL against Staphylococcus aureus and 8 microg/mL against Streptococcus pneumoniae. The isolation, structure elucidation, and antibacterial activities of four new members, lucensimycins D-G, are described. Lucensimycins D (4a) and E (5) are N-acetyl-l-cysteine adducts of lucensimycin A (1). Semisynthesis of lucensimycins D and E from lucensimycin A has also been described. Lucensimycins F and G are myo-inositolyl-alpha-2-amino-2-deoxy-l-idosyl amide derivatives of lucensimycins D and E, respectively. The relative configuration of these compounds was determined, in part, by molecular dynamics simulations.


Bioorganic & Medicinal Chemistry Letters | 1999

Synthesis and activity of 2-(sulfonamido)methyl-carbapenems: Discovery of a novel, anti-MRSA 1,8-naphthosultam pharmacophore

Robert R. Wilkening; Ronald W. Ratcliffe; Kenneth J. Wildonger; Lovji D. Cama; Kevin D. Dykstra; Frank P. DiNinno; Timothy A. Blizzard; Milton L. Hammond; James V. Heck; Karen Dorso; E.St. Rose; Joyce Kohler; Gail G. Hammond

A series of 1beta-methyl carbapenems substituted at the 2-position with lipophilic, acyclic and cyclic (sulfonamido)methyl groups was prepared and evaluated for activity against resistant gram-positive bacteria. From these studies, the 1,8-naphthosultamyl group emerged as a novel, PBP2a-binding, anti-MRSA pharmacophore worthy of further exploration.


Bioorganic & Medicinal Chemistry | 2009

Isolation, structure and antibacterial activity of pleosporone from a pleosporalean ascomycete discovered by using antisense strategy.

Chaowei Zhang; John G. Ondeyka; Deborah L. Zink; Angela Basilio; Francisca Vicente; Javier Collado; Gonzalo Platas; Joann Huber; Karen Dorso; Mary Motyl; Kevin M. Byrne; Sheo B. Singh

Protein synthesis is one of the best antibacterial targets that have led to the development of a number of highly successful clinical drugs. Protein synthesis is catalyzed by ribosome, which is comprised of a number of ribosomal proteins that help the catalysis process. Ribosomal protein S4 (RPSD) is one of the proteins that is a part of the ribosomal machinery and is a potential new target for the discovery of antibacterial agents. Screening of microbial extracts using antisense-sensitized rpsD Staphylococcus aureus strain led to the isolation of pleosporone, a new compound, with modest antibacterial activities with MIC ranging from 1 to 64 microg/mL. This compound showed the highest sensitivity for Streptococcus pneumoniae and Haemophilus influenzae, and exhibited MICs of 4 and 1 microg/mL, respectively. Pleosporone showed modest selectivity for the inhibition of RNA synthesis compared to DNA and protein synthesis, and showed activity against HeLa cells. Isolation, structure elucidation, and biological activity of pleosporone have been described.

Collaboration


Dive into the Karen Dorso's collaboration.

Researchain Logo
Decentralizing Knowledge