Network


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

Hotspot


Dive into the research topics where Michael N. Lombardo is active.

Publication


Featured researches published by Michael N. Lombardo.


Journal of Molecular Biology | 2009

Crystal structures of wild-type and mutant methicillin-resistant Staphylococcus aureus dihydrofolate reductase reveal an alternate conformation of NADPH that may be linked to trimethoprim resistance.

Kathleen M. Frey; Jieying Liu; Michael N. Lombardo; David B. Bolstad; Dennis L. Wright; Amy C. Anderson

Both hospital- and community-acquired Staphylococcus aureus infections have become major health concerns in terms of morbidity, suffering and cost. Trimethoprim-sulfamethoxazole (TMP-SMZ) is an alternative treatment for methicillin-resistant S. aureus (MRSA) infections. However, TMP-resistant strains have arisen with point mutations in dihydrofolate reductase (DHFR), the target for TMP. A single point mutation, F98Y, has been shown biochemically to confer the majority of this resistance to TMP. Using a structure-based approach, we have designed a series of novel propargyl-linked DHFR inhibitors that are active against several trimethoprim-resistant enzymes. We screened this series against wild-type and mutant (F98Y) S. aureus DHFR and found that several are active against both enzymes and specifically that the meta-biphenyl class of these inhibitors is the most potent. In order to understand the structural basis of this potency, we determined eight high-resolution crystal structures: four each of the wild-type and mutant DHFR enzymes bound to various propargyl-linked DHFR inhibitors. In addition to explaining the structure-activity relationships, several of the structures reveal a novel conformation for the cofactor, NADPH. In this new conformation that is predominantly associated with the mutant enzyme, the nicotinamide ring is displaced from its conserved location and three water molecules complete a network of hydrogen bonds between the nicotinamide ring and the protein. In this new position, NADPH has reduced interactions with the inhibitor. An equilibrium between the two conformations of NADPH, implied by their occupancies in the eight crystal structures, is influenced both by the ligand and the F98Y mutation. The mutation induced equilibrium between two NADPH-binding conformations may contribute to decrease TMP binding and thus may be responsible for TMP resistance.


Journal of Structural Biology | 2010

Towards the Understanding of Resistance Mechanisms in Clinically Isolated Trimethoprim-resistant, Methicillin-resistant Staphylococcus aureus Dihydrofolate Reductase

Kathleen M. Frey; Michael N. Lombardo; Dennis L. Wright; Amy C. Anderson

Resistance to therapeutics such as trimethoprim-sulfamethoxazole has become an increasing problem in strains of methicillin-resistant Staphylococcus aureus (MRSA). Clinically isolated trimethoprim-resistant strains reveal a double mutation, H30N/F98Y, in dihydrofolate reductase (DHFR). In order to develop novel and effective therapeutics against these resistant strains, we evaluated a series of propargyl-linked antifolate lead compounds for inhibition of the mutant enzyme. For the propargyl-linked antifolates, the F98Y mutation generates minimal (between 1.2- and 6-fold) losses of affinity and the H30N mutation generates greater losses (between 2.4- and 48-fold). Conversely, trimethoprim affinity is largely diminished by the F98Y mutation (36-fold) and is not affected by the H30N mutation. In order to elucidate a mechanism of resistance, we determined a crystal structure of a complex of this double mutant with a lead propargyl-linked antifolate. This structure suggests a resistance mechanism consistent both for the propargyl-linked class of antifolates and for trimethoprim that is based on the loss of a conserved water-mediated hydrogen bond.


Journal of Medicinal Chemistry | 2014

Propargyl-linked antifolates are dual inhibitors of Candida albicans and Candida glabrata.

Narendran G-Dayanandan; Janet L. Paulsen; Kishore Viswanathan; Santosh Keshipeddy; Michael N. Lombardo; Wangda Zhou; Kristen M. Lamb; Adrienne E. Sochia; Jeremy B. Alverson; Nigel D. Priestley; Dennis L. Wright; Amy C. Anderson

Species of Candida, primarily C. albicans and with increasing prevalence, C. glabrata, are responsible for the majority of fungal bloodstream infections that cause morbidity, especially among immune compromised patients. While the development of new antifungal agents that target the essential enzyme, dihydrofolate reductase (DHFR), in both Candida species would be ideal, previous attempts have resulted in antifolates that exhibit inconsistencies between enzyme inhibition and antifungal properties. In this article, we describe the evaluation of pairs of propargyl-linked antifolates that possess similar physicochemical properties but different shapes. All of these compounds are effective at inhibiting the fungal enzymes and the growth of C. glabrata; however, the inhibition of the growth of C. albicans is shape-dependent with extended para-linked compounds proving more effective than compact, meta-linked compounds. Using crystal structures of DHFR from C. albicans and C. glabrata bound to lead compounds, 13 new para-linked compounds designed to inhibit both species were synthesized. Eight of these compounds potently inhibit the growth of both fungal species with three compounds displaying dual MIC values less than 1 μg/mL. Analysis of the active compounds shows that shape and distribution of polar functionality is critical in achieving dual antifungal activity.


Antimicrobial Agents and Chemotherapy | 2014

Crystal structures of Klebsiella pneumoniae dihydrofolate reductase bound to propargyl-linked antifolates reveal features for potency and selectivity

Kristen M. Lamb; Michael N. Lombardo; Jeremy B. Alverson; Nigel D. Priestley; Dennis L. Wright; Amy C. Anderson

ABSTRACT Resistance to the antibacterial antifolate trimethoprim (TMP) is increasing in members of the family Enterobacteriaceae, driving the design of next-generation antifolates effective against these Gram-negative pathogens. The propargyl-linked antifolates are potent inhibitors of dihydrofolate reductases (DHFR) from several TMP-sensitive and -resistant species, including Klebsiella pneumoniae. Recently, we have determined that these antifolates inhibit the growth of strains of K. pneumoniae, some with MIC values of 1 μg/ml. In order to further the design of potent and selective antifolates against members of the Enterobacteriaceae, we determined the first crystal structures of K. pneumoniae DHFR bound to two of the propargyl-linked antifolates. These structures highlight that interactions with Leu 28, Ile 50, Ile 94, and Leu 54 are necessary for potency; comparison with structures of human DHFR bound to the same inhibitors reveal differences in residues (N64E, P61G, F31L, and V115I) and loop conformations (residues 49 to 53) that may be exploited for selectivity.


ACS Medicinal Chemistry Letters | 2016

Charged Nonclassical Antifolates with Activity Against Gram-Positive and Gram-Negative Pathogens.

Eric W. Scocchera; Stephanie M. Reeve; Santosh Keshipeddy; Michael N. Lombardo; Behnoush Hajian; Adrienne E. Sochia; Jeremy B. Alverson; Nigel D. Priestley; Amy C. Anderson; Dennis L. Wright

Although classical, negatively charged antifolates such as methotrexate possess high affinity for the dihydrofolate reductase (DHFR) enzyme, they are unable to penetrate the bacterial cell wall, rendering them poor antibacterial agents. Herein, we report a new class of charged propargyl-linked antifolates that capture some of the key contacts common to the classical antifolates while maintaining the ability to passively diffuse across the bacterial cell wall. Eight synthesized compounds exhibit extraordinary potency against Gram-positive S. aureus with limited toxicity against mammalian cells and good metabolic profile. High resolution crystal structures of two of the compounds reveal extensive interactions between the carboxylate and active site residues through a highly organized water network.


Biochemistry | 2015

Measuring Propargyl-Linked Drug Populations Inside Bacterial Cells, and Their Interaction with a Dihydrofolate Reductase Target, by Raman Microscopy

Hossein Heidari-Torkabadi; Tao Che; Michael N. Lombardo; Dennis L. Wright; Amy C. Anderson; Paul R. Carey

We report the first Raman spectroscopic study of propargyl-linked dihydrofolate reductase (DHFR) inhibitors being taken up by wild type Escherichia coli, Klebsiella pneumoniae, and Staphylococcus aureus cells. A novel protocol is developed where cells are exposed to the fermentation medium containing a known amount of an inhibitor. At a chosen time point, the cells are centrifuged and washed to remove the extracellular compound, then frozen and freeze-dried. Raman difference spectra of the freeze-dried cells (cells exposed to the drug minus cells alone) provide spectra of the compounds inside the cells, where peak intensities allow us to quantify the number of inhibitors within each cell. A time course for the propargyl-linked DHFR inhibitor UCP 1038 soaking into E. coli cells showed that penetration occurs very quickly and reaches a plateau after 10 min exposure to the inhibitor. After 10 min drug exposure, the populations of two inhibitors, UCP 1038 and UCP 1089, were ~1.5 × 10(6) molecules in each E. coli cell, ~4.7 × 10(5) molecules in each K. pneumonia cell, and ~2.7 × 10(6) in each S. aureus cell. This is the first in situ comparison of inhibitor population in Gram-negative and Gram-positive bacterial cells. The positions of the Raman peaks also reveal the protonation of diaminopyrimidine ring upon binding to DHFR inside cells. The spectroscopic signature of protonation was characterized by binding an inhibitor to a single crystal of DHFR.


ACS Infectious Diseases | 2016

Crystal Structures of Trimethoprim-Resistant DfrA1 Rationalize Potent Inhibition by Propargyl-Linked Antifolates

Michael N. Lombardo; Narendran G-Dayanandan; Dennis L. Wright; Amy C. Anderson

Multidrug-resistant Enterobacteriaceae, notably Escherichia coli and Klebsiella pneumoniae, have become major health concerns worldwide. Resistance to effective therapeutics is often carried by class I and II integrons that can confer insensitivity to carbapenems, extended spectrum β-lactamases, the antifolate trimethoprim, fluoroquinolones, and aminoglycosides. Specifically of interest to the study here, a prevalent gene (dfrA1) coding for an insensitive dihydrofolate reductase (DHFR) confers 190- or 1000-fold resistance to trimethoprim for K. pneumoniae and E. coli, respectively. Attaining inhibition of both the wild-type and resistant forms of the enzyme is critical for new antifolates. For several years, we have been developing the propargyl-linked antifolates (PLAs) as effective inhibitors against trimethoprim-resistant DHFR enzymes. Here, we show that the PLAs are active against both the wild-type and DfrA1 DHFR proteins. We report two high-resolution crystal structures of DfrA1 bound to potent PLAs. The structure-activity relationships and crystal structures will be critical in driving the design of broadly active inhibitors against wild-type and resistant DHFR.


Chemical Science | 2017

Characterization of trimethoprim resistant E. coli dihydrofolate reductase mutants by mass spectrometry and inhibition by propargyl-linked antifolates

Michael B. Cammarata; Ross Thyer; Michael N. Lombardo; Amy C. Anderson; Dennis L. Wright; Andrew D. Ellington; Jennifer S. Brodbelt

Pathogenic Escherichia coli, one of the primary causes of urinary tract infections, has shown significant resistance to the most popular antibiotic, trimethoprim (TMP), which inhibits dihydrofolate reductase (DHFR). The resistance is modulated by single point mutations of DHFR. The impact of two clinically relevant mutations, P21L and W30R, on the activity of DHFR was evaluated via measurement of Michaelis–Menten and inhibitory kinetics, and structural characterization was undertaken by native mass spectrometry with ultraviolet photodissociation (UVPD). Compared to WT-DHFR, both P21L and W30R mutants produced less stable complexes with TMP in the presence of co-factor NADPH as evidenced by the relative abundances of complexes observed in ESI mass spectra. Moreover, based on variations in the fragmentation patterns obtained by UVPD mass spectrometry of binary and ternary DHFR complexes, notable structural changes were localized to the substrate binding pocket for W30R and to the M20 loop region as well as the C-terminal portion containing the essential G–H functional loop for the P21L mutant. The results suggest that the mutations confer resistance through distinctive mechanisms. A novel propargyl-linked antifolate compound 1038 was shown to be a reasonably effective inhibitor of the P21L mutant.


Future Microbiology | 2015

Understanding the structural mechanisms of antibiotic resistance sets the platform for new discovery

Stephanie M. Reeve; Michael N. Lombardo; Amy C. Anderson

Understanding the structural basis of antibacterial resistance may enable rational design principles that avoid and subvert that resistance, thus leading to the discovery of more effective antibiotics. In this review, we explore the use of crystal structures to guide new discovery of antibiotics that are effective against resistant organisms. Structures of efflux pumps bound to substrates and inhibitors have aided the design of compounds with lower affinity for the pump or inhibitors that more effectively block the pump. Structures of β-lactamase enzymes have revealed the mechanisms of action toward key carbapenems and structures of gyrase have aided the design of compounds that are less susceptible to point mutations.


Journal of Physical Chemistry B | 2018

Measuring Drug-Induced Changes in Metabolite Populations of Live Bacteria: Real Time Analysis by Raman Spectroscopy

Paul R. Carey; Grant R. Whitmer; Michael J. Yoon; Michael N. Lombardo; Marianne Pusztai-Carey; Hossein Heidari-Torkabadi; Tao Che

Raman difference spectroscopy is shown to provide a wealth of molecular detail on changes within bacterial cells caused by infusion of antibiotics or hydrogen peroxide. Escherichia coli strains paired with chloramphenicol, dihydrofolate reductase propargyl-based inhibitors, meropenem, or hydrogen peroxide provide details of the depletion of protein and nucleic acid populations in real time. Additionally, other reproducible Raman features appear and are attributed to changes in cell metabolite populations. An initial candidate for one of the metabolites involves population increases of citrate, an intermediate within the tricarboxyclic acid cycle. This is supported by the observation that a strain of E. coli without the ability to synthesize citrate, gltA, lacks an intense feature in the Raman difference spectrum that has been ascribed to citrate. The methodology for obtaining the Raman data involves infusing the drug into live cells, then washing, freezing, and finally lyophilizing the cells. The freeze-dried cells are then examined under a Raman microscope. The difference spectra [cells treated with drug] - [cells without treatment] are time-dependent and can yield population kinetics for intracellular species in vivo. There is a strong resemblance between the Raman difference spectra of E. coli cells treated with meropenem and those treated with hydrogen peroxide.

Collaboration


Dive into the Michael N. Lombardo's collaboration.

Top Co-Authors

Avatar

Amy C. Anderson

University of Connecticut

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge