Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 44
1.
ACS Infect Dis ; 9(10): 2016-2024, 2023 Oct 13.
Article En | MEDLINE | ID: mdl-37655755

Menaquinone (MK) is an essential component in the oxidative phosphorylation pathway of Gram-positive bacteria. Drugs targeting enzymes involved in MK biosynthesis can prevent electron transfer, which leads to ATP starvation and thereby death of microorganisms. Previously, we reported a series of MenA inhibitors and demonstrated their antimicrobial activity against Gram-positive bacteria, including Methicillin-resistant Staphylococcus aureus (MRSA) and mycobacteria. These inhibitors were developed by mimicking demethylmenaquinone, a product of MenA enzymatic reaction in MK biosynthesis. In this study, compound NM4, MK biosynthesis inhibitor, inhibited the formation of MRSA biofilm and it was screened against 1952 transposon mutants to elucidate mechanisms of action; however, no resistant mutants were found. Also, compound NM4 induced the production of reactive oxygen species (ROS) by blocking electron transfer in the oxidative phosphorylation pathway as observed by MRSA growth recovery using various ROS scavengers. An oxygen consumption assay also showed that NM4 blocks the oxygen consumption by MRSA, but the addition of menaquinone (MK) restores growth of MRSA. The NM4-treated MRSA induced the expression of catalase by more than 25%, as quantified by the native gel. A pulmonary murine model exhibited that NM4 significantly reduced bacterial lung load in mice without toxicity. An NM4-resistant USA300 strain was developed to attempt to identify the targets participating in the mechanism of resistance. Our results support that respiration and oxidative phosphorylation are potential targets for developing antimicrobial agents against MRSA. Altogether, our findings suggest the potential use of MK biosynthesis inhibitors as an effective antimicrobial agent against MRSA.

2.
Mol Pharm ; 20(8): 4058-4070, 2023 08 07.
Article En | MEDLINE | ID: mdl-37471668

There is a major need for the development of new therapeutics to combat antibiotic-resistant Staphylococcus aureus. Recently, gallium (Ga)-based complexes have shown promising antimicrobial effects against various bacteria, including multidrug-resistant organisms, by targeting multiple heme/iron-dependent metabolic pathways. Among these, Ga protoporphyrin (GaPP) inhibits bacterial growth by targeting heme pathways, including aerobic respiration. Ga(NO3)3, an iron mimetic, disrupts elemental iron pathways. Here, we demonstrate the enhanced antimicrobial activity of the combination of GaPP and Ga(NO3)3 against methicillin-resistant S. aureus (MRSA) under iron-limited conditions, including small colony variants (SCV). This therapy demonstrated significant antimicrobial activity without inducing slow-growing SCV. We also observed that the combination of GaPP and Ga(NO3)3 inhibited the MRSA catalase but not above that seen with Ga(NO3)3 alone. Neither GaPP nor Ga(NO3)3 alone or their combination inhibited the dominant superoxide dismutase expressed (SodA) under the iron-limited conditions examined. Intranasal administration of the combination of the two compounds improved drug biodistribution in the lungs compared to intraperitoneal administration. In a murine MRSA lung infection model, we observed a significant increase in survival and decrease in MRSA lung CFUs in mice that received combination therapy with intranasal GaPP and Ga(NO3)3 compared to untreated control or mice receiving GaPP or Ga(NO3)3 alone. No drug-related toxicity was observed as assessed histologically in the spleen, lung, nasal cavity, and kidney for both single and repeated doses of 10 mg Ga /Kg of mice over 13 days. Our results strongly suggest that GaPP and Ga(NO3)3 in combination have excellent synergism and potential to be developed as a novel therapy for infections with S. aureus.


Gallium , Methicillin-Resistant Staphylococcus aureus , Animals , Mice , Protoporphyrins/pharmacology , Protoporphyrins/metabolism , Staphylococcus aureus , Tissue Distribution , Anti-Bacterial Agents/pharmacology , Gallium/pharmacology , Heme/metabolism , Iron/metabolism , Microbial Sensitivity Tests
3.
ACS Infect Dis ; 9(4): 716-738, 2023 04 14.
Article En | MEDLINE | ID: mdl-36995299

The treatment of infections is becoming more difficult due to emerging resistance of pathogens to existing drugs. As such, alternative druggable targets, particularly those that are essential for microbe viability and thus make it harder to develop resistance, are desperately needed. In turn, once identified, safe and effective agents that disrupt these targets must be developed. Microbial acquisition and use of iron is a promising novel target for antimicrobial drug development. In this Review we look at the various facets of iron metabolism critical to human infection with pathogenic microbes and the various ways in which it can be targeted, altered, disrupted, and taken advantage of to halt or eliminate microbial infections. Although a variety of agents will be touched upon, the primary focus will be on the potential use of one or more gallium complexes as a new class of antimicrobial agents. In vitro and in vivo data on the activity of gallium complexes against a variety of pathogens including ESKAPE pathogens, mycobacteria, emerging viruses, and fungi will be discussed in detail, as well as pharmacokinetics, novel formulations and delivery approaches, and early human clinical results.


Anti-Infective Agents , Gallium , Humans , Gallium/pharmacology , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Iron/metabolism , Drug Delivery Systems
4.
Microorganisms ; 12(1)2023 Dec 21.
Article En | MEDLINE | ID: mdl-38276186

In a recent effort to mitigate harm from human pathogens, many biosynthetic pathways have been extensively evaluated for their ability to inhibit pathogen growth and to determine drug targets. One of the important products/targets of such pathways is isopentenyl diphosphate. Isopentenyl diphosphate is the universal precursor of isoprenoids, which are essential for the normal functioning of microorganisms. In general, two biosynthetic pathways lead to the formation of isopentenyl diphosphate: (1) the mevalonate pathway in animals; and (2) the non-mevalonate or methylerythritol phosphate (MEP) in many bacteria, and some protozoa and plants. Because the MEP pathway is not found in mammalian cells, it is considered an attractive target for the development of antimicrobials against a variety of human pathogens, including Mycobacterium tuberculosis (M.tb). In the MEP pathway, 4-diphosphocytidyl-2-c-methyl-d-erythritol kinase (IspE) phosphorylates 4-diphosphocytidyl-2-C-methyl-D-erythritol (CDPME) to form 4-diphosphocytidyl-2-C-methyl-D-erythritol 2-phosphate (CDPME2P). A virtual high-throughput screening against 15 million compounds was carried out by docking IspE protein. We identified an active heterotricyclic compound which showed enzymatic activity; namely, IC50 of 6 µg/mL against M.tb IspE and a MIC of 12 µg/mL against M.tb (H37Rv). Hence, we designed and synthesized similar new heterotricyclic compounds and tested them against mycobacterium, observing a MIC of 5 µg/mL against M. avium. This study will provide the critical insight necessary for developing novel antimicrobials that target the MEP pathways in pathogens.

5.
ACS Infect Dis ; 8(10): 2096-2105, 2022 10 14.
Article En | MEDLINE | ID: mdl-36049087

Pseudomonas aeruginosa is a highly antibiotic-resistant opportunistic pathogenic bacteria that is responsible for thousands of deaths each year. Infections with P. aeruginosa disproportionately impact individuals with compromised immune systems as well as cystic fibrosis patients, where P. aeruginosa lung infection is a leading cause of morbidity and mortality. In previous work, we showed that a combination of gallium (Ga) nitrate and Ga protoporphyrin worked well in several bacterial infection models but its mechanism of action (MOA) is unknown. In the current work, we have investigated the MOA of Ga combination therapy in P. aeruginosa and its analysis in the in vivo model. In P. aeruginosa treated with Ga combination therapy, we saw a decrease in catalase and superoxide dismutase (SOD) activity, key antioxidant enzymes, which could correlate with a higher potential for oxidative stress. Consistent with this hypothesis, we found that, following combination therapy, P. aeruginosa demonstrated higher levels of reactive oxygen species, as measured using the redox-sensitive fluorescent probe, H2DCFDA. We also saw that the Ga combination therapy killed phagocytosed bacteria inside macrophages in vitro. The therapy with low dose was able to fully prevent mortality in a murine model of P. aeruginosa lung infection and also significantly reduced lung damage. These results support our previous data that Ga combination therapy acts synergistically to kill P. aeruginosa, and we now show that this may occur through increasing the organism's susceptibility to oxidative stress. Ga combination therapy also showed itself to be effective at treating infection in a murine pulmonary-infection model.


Gallium , Pseudomonas aeruginosa , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antioxidants/pharmacology , Bacteria , Catalase/pharmacology , Fluorescent Dyes , Gallium/pharmacology , Humans , Mice , Nitrates/pharmacology , Protoporphyrins/pharmacology , Reactive Oxygen Species , Superoxide Dismutase
6.
ACS Chem Neurosci ; 13(8): 1165-1177, 2022 04 20.
Article En | MEDLINE | ID: mdl-35385645

Cannabidiol is a nonpsychoactive phytocannabinoid produced by the Cannabis sativa plant and possesses a wide range of pharmacological activities, including anti-inflammatory, antioxidant, and neuroprotective activities. Cannabidiol functions in a neuroprotective manner, in part through the activation of cellular antioxidant pathways. The glyoxalase pathway detoxifies methylglyoxal, a highly reactive metabolic byproduct that can accumulate in the brain, and contributes to the severity of neurodegenerative diseases, including Alzheimer's disease. While cannabidiol's antioxidant properties have been investigated, it is currently unknown how it may modulate the glyoxalase pathway. In this research paper, we examine the effects of Cannabidiol on cerebellar neurons and in several Caenorhabditis elegans strains. We determined that a limited amount of Cannabidiol can prevent methylglyoxal-mediated cellular damage through enhancement of the neural glyoxalase pathway and extend the lifespan and survival of C. elegans, including a transgenic C. elegans strain modeling Alzheimer's disease.


Alzheimer Disease , Caenorhabditis elegans Proteins , Cannabidiol , Lactoylglutathione Lyase , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Antioxidants/pharmacology , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/pharmacology , Cannabidiol/pharmacology , Lactoylglutathione Lyase/metabolism , Longevity , Pyruvaldehyde/metabolism
7.
Bioorg Med Chem Lett ; 62: 128645, 2022 04 15.
Article En | MEDLINE | ID: mdl-35219822

Bacterial heme uptake pathways offer a novel target for antimicrobial drug discovery. Recently, gallium (Ga) porphyrin complexes were found to be effective against mycobacterial heme uptake pathways. The goal of the current study is to build on this foundation and develop a new Ga(III) porphyrin and its nanoparticles, formulated by a single emulsion-evaporation technique to inhibit the growth of Mycobacterium avium complex (MAC) with enhanced properties. Gallium 5,10,15,20-tetrakis(4-methoxyphenyl)porphyrin chloride (GaMeOTP) was synthesized from 5,10,15,20-tetrakis(4-methoxyphenyl)porphyrin and GaCl3. GaMeOTP showed enhanced antimicrobial activity against MAC104 and some clinical M. avium isolates. The synthesized Ga(III) porphyrin antimicrobial activity resulted in the overproduction of reactive oxygen species. Our study also demonstrated that F127 nanoparticles encapsulating GaMeOTP exhibited a smaller size than GaTP nanoparticles and a better duration of activity in MAC-infected macrophages compared to the free GaMeOTP. The nanoparticles were trafficked to endosomal compartments within MAC-infected macrophages, likely contributing to the antimicrobial activity of the drug.


Anti-Infective Agents , Gallium , Nanoparticles , Porphyrins , Anti-Bacterial Agents/pharmacology , Gallium/pharmacology , Heme , Mycobacterium avium Complex , Porphyrins/pharmacology
9.
ACS Infect Dis ; 7(8): 2299-2309, 2021 08 13.
Article En | MEDLINE | ID: mdl-34314150

The emergence of drug-resistant pathogens causes the greatest challenge for drug development research. Recently, gallium(III)-based compounds have received great attention as novel antimicrobial agents against drug-resistant pathogens. Here, we synthesized a new ß-cyclodextrin Ga nanoparticle (CDGaTP) using Ga tetraphenylporphyrin (GaTP, a hemin analogue) and ß-cyclodextrin. The newly synthesized nanoparticle was nontoxic and efficient at a single dose, showing sustained drug release for 15 days in vitro. CDGaTP's activity with transferrin or lactoferrin was tested, and synergism in activity was observed against nontuberculosis mycobacteria (NTM), Mycobacterium avium (M. avium), and Mycobacteroides abscessus. Human serum albumin (HSA) decreased the efficacy of both GaTP and CDGaTP in a concentration-dependent manner. The NTMs incubated with GaTP or CDGaTP significantly produced reactive oxygen species (ROS), indicating potential inhibition of antioxidant enzymes, such as catalase. The single-dose CDGaTP displayed a prolonged intracellular inhibitory activity in an in vitro macrophage infection model against both NTMs. In addition, CDGaTP, not GaTP, was effective in a murine lung M. avium infection model when delivered via intranasal administration. These results suggest that CDGaTP provides new opportunities for the development of gallium-porphyrin based antibiotics.


Gallium , Mycobacterium abscessus , Porphyrins , beta-Cyclodextrins , Animals , Anti-Bacterial Agents/pharmacology , Gallium/pharmacology , Humans , Mice , Microbial Sensitivity Tests , Mycobacterium avium , Porphyrins/pharmacology
10.
Bioorg Med Chem Lett ; 47: 128203, 2021 09 01.
Article En | MEDLINE | ID: mdl-34139327

Menaquinone (MK) plays essential role in the electron transport chain (ETC), suggesting MK biosynthesis enzymes as potential targets for drug development. Previously, we demonstrated that Methicillin-resistant Staphylococcus aureus (MRSA) is susceptible to naphthol-based compounds which were developed by mimicking demethylmenaquinone, a product of MenA enzymatic reaction. Here, a series of new MenA inhibitors (4-19) were synthesized and evaluated as MenA inhibitors in this study. The inhibitors were designed to improve growth inhibitory activity against MRSA. Among the MenA inhibitors, bicyclic substituted amine 3 showed MIC of 3 µg/mL, and alkenyl substituted amine 11 showed MIC of 8 µg/mL against USA300. Regrowth of MRSA was observed on addition of MK when exposed to 8 µg/mL of inhibitor 11, supporting inhibition of MK biosynthesis. However, inhibitor 11 did not show efficacy in treating USA300 infected C. elegans up to 25 µg/mL concentration. However, all infected C. elegans survived when exposed to a bicyclic substituted amine 3. Hence, a bicyclic substituted amine was tested in mice for tolerability and biodistribution and observed 100% tolerable and high level of compound accumulation in lungs.


Amines/pharmacology , Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Microfilament Proteins/antagonists & inhibitors , Amines/chemical synthesis , Amines/chemistry , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Female , Methicillin-Resistant Staphylococcus aureus/metabolism , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microfilament Proteins/metabolism , Molecular Structure , Structure-Activity Relationship
11.
ACS Infect Dis ; 6(10): 2582-2591, 2020 10 09.
Article En | MEDLINE | ID: mdl-32845117

There is an urgent need for new effective and safe antibiotics active against pathogenic mycobacterial species. Gallium (Ga) nitrate (Ga(NO3)3) and Ga porphyrin (GaPP) have each been shown to inhibit the growth of a variety of mycobacterial species. The Ga(III) ion derived from Ga(NO3)3 has the potential to disrupt the mycobacterial Fe(III) uptake mechanisms and utilization, including replacing iron (Fe) in the active site of enzymes, resulting in the disruption of function. Similarly, noniron metalloporphyrins such as heme mimetics, which can be transported across the bacterial membrane via heme-uptake pathways, would potentially block the acquisition of iron-containing heme and bind to heme-utilizing proteins, making them nonfunctional. Given that they likely act on different aspects of mycobacterial Fe metabolism, the efficacy of combining Ga(NO3)3 and GaPP was studied in vitro against Mycobacterium avium, Mycobacterium abscessus, and Mycobacterium tuberculosis (M. tb). The combination was then assessed in vivo in a murine pulmonary infection model of M. abscessus. We observed that Ga(NO3)3 in combination with GaPP exhibited synergistic inhibitory activity against the growth of M. avium, M. tb, and M. abscessus, being most active against M. abscessus. Activity assays indicated that Ga(NO3)3 and GaPP inhibited both catalase and aconitase at high concentrations. However, the combination showed a synergistic effect on the aconitase activity of M. abscessus. The Ga(NO3)3/GaPP combination via intranasal administration showed significant antimicrobial activity in mice infected with M. abscessus. M. abscessus CFU from the lungs of the Ga(NO3)3/GaPP-treated mice was significantly less compared to that of nontreated or single Ga(III)-treated mice. These findings suggest that combinations of different Ga(III) compounds can synergistically target multiple iron/heme-utilizing mycobacterial enzymes. The results support the potential of combination Ga therapy for development against mycobacterial pathogens.


Gallium , Porphyrins , Animals , Gallium/pharmacology , Heme , Iron , Mice , Microbial Sensitivity Tests , Porphyrins/pharmacology
12.
ACS Chem Neurosci ; 11(3): 356-366, 2020 02 05.
Article En | MEDLINE | ID: mdl-31909963

The glyoxalase pathway (GP) is an antioxidant defense system that detoxifies metabolic byproduct methylglyoxal (MG). Through sequential reactions, reduced glutathione (GSH), glyoxalase I (glo-1), and glyoxalase II (glo-2) convert MG into d-lactate. Spontaneous reactions involving MG alter the structure and function of cellular macromolecules through the formation of inflammatory advanced glycation endproducts (AGEs). Accumulation of MG and AGEs in neural cells contributes to oxidative stress (OS), a state of elevated inflammation commonly found in neurodegenerative diseases including Alzheimer's disease (AD). Morin is a common plant-produced flavonoid polyphenol that exhibits the ability to enhance the GP-mediated detoxification of MG. We hypothesize that structural modifications to morin will improve its inherent GP enhancing ability. Here we synthesized a morin derivative, dibromo-morin (DBM), formulated a morin encapsulated nanoparticle (MNP), and examined their efficacy in enhancing neural GP activity. Cultured mouse primary cerebellar neurons and Caenorhabditis elegans were induced to a state of OS with MG and treated with morin, DBM, and MNP. Results indicated the morin derivatives were more effective compared to the parent compound in neural GP enhancement and preventing MG-mediated OS in an AD model.


Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Flavonoids/pharmacology , Lactoylglutathione Lyase/pharmacology , Animals , Antioxidants/pharmacology , Flavonoids/chemistry , Inflammation/metabolism , Lactoylglutathione Lyase/metabolism , Mice, Inbred C57BL , Neural Pathways/metabolism , Neurons/drug effects , Oxidative Stress/drug effects , Pyruvaldehyde/metabolism , Pyruvaldehyde/pharmacology
13.
mSphere ; 4(4)2019 07 24.
Article En | MEDLINE | ID: mdl-31341073

Tuberculosis (TB), caused by Mycobacterium tuberculosis, remains a global threat. The course of TB is negatively impacted by coexistent infection with human immunodeficiency virus type 1 (HIV). Macrophage infection with these pathogens modulates their production of pro- and anti-inflammatory cytokines, which could play a crucial role in pathogenesis. Despite the important role of macrophages in containing infection by a variety of microbes, both HIV and M. tuberculosis infect and replicate within these cells during the course of HIV-M. tuberculosis coinfection. Both M. tuberculosis and HIV require iron for growth and replication. We have previously shown that gallium encapsulated in nanoparticles, which interferes with cellular iron acquisition and utilization, inhibited the growth of HIV and an attenuated strain of M. tuberculosis within human monocyte-derived macrophages (MDMs) in vitro Whether this was true for a fully virulent strain of M. tuberculosis and whether gallium treatment modulates cytokine production by HIV- and/or M. tuberculosis-infected macrophages have not been previously addressed. Therefore, coinfection of MDMs with HIV and a virulent M. tuberculosis strain (H37Rv) was studied in the presence of different gallium nanoparticles (GaNP). All GaNP were readily internalized by the MDMs, which provided sustained drug (gallium) release for 15 days. This led to significant growth inhibition of both HIV and M. tuberculosis within MDMs for up to 15 days after loading of the cells with all GaNP tested in our study. Cytokine analysis showed that HIV-M. tuberculosis coinfected macrophages secreted large amounts of interleukin 6 (IL-6) and IL-8 and smaller amounts of IL-1ß, IL-4, and tumor necrosis factor alpha (TNF-α) cytokines. However, all GaNP were able to regulate the release of cytokines significantly. GaNP interrupts iron-mediated enzymatic reactions, leading to growth inhibition of HIV-M. tuberculosis coinfection in macrophages, and also modulates release of cytokines that may contribute to HIV-TB pathogenesis.IMPORTANCE GaNP interrupts iron-mediated enzymatic reactions, leading to growth inhibition of virulent HIV-M. tuberculosis coinfection in macrophages, and also modulates release of cytokines that may contribute to HIV-TB pathogenesis. Macrophage-targeting GaNP are a promising therapeutic approach to provide sustained antimicrobial activity against HIV-M. tuberculosis coinfection.


Cytokines/immunology , Gallium/pharmacology , HIV-1/drug effects , Metal Nanoparticles/chemistry , Mycobacterium tuberculosis/drug effects , Cells, Cultured , Humans , Iron/metabolism , Macrophages/microbiology , Macrophages/virology , Mycobacterium tuberculosis/pathogenicity
14.
ACS Infect Dis ; 5(9): 1559-1569, 2019 09 13.
Article En | MEDLINE | ID: mdl-31264851

Iron- and heme-uptake pathways and metabolism are promising targets for the development of new antimicrobial agents, as their disruption would lead to nutritional iron starvation and inhibition of bacterial growth. Salts of gallium(III) (Ga), an iron mimetic metal, disrupt iron-dependent biological processes by binding iron-utilizing proteins and competing with iron for uptake by bacterial siderophore-mediated iron uptake systems. Ga porphyrins, heme mimetic complexes, disrupt heme-utilizing hemoproteins. Because Ga(NO3)3 and Ga porphyrin disrupt different pathways of bacterial ion acquisition and utilization, we hypothesized that if used in combination, they would result in enhanced antimicrobial activity. Antimicrobial activity of Ga porphyrins (Ga protoporphyrin, GaPP, or Ga mesoporphyrin, GaMP) alone and in combination with Ga(NO3)3 were evaluated against Pseudomonas aeruginosa, Klebsiella pneumoniae, Acinetobacter baumannii, and methicillin-resistant Staphylococcus aureus (MRSA) under iron-limited conditions. The Ga porphyrin/Ga(NO3)3 combination demonstrated substantial synergism against K. pneumoniae, P. aeruginosa, and MRSA. Time-kill assays revealed that the synergistic combination of GaPP/Ga(NO3)3 was bacteriostatic against K. pneumoniae and MRSA and bactericidal against P. aeruginosa. The GaPP/Ga(NO3)3 combination significantly disrupted K. pneumoniae and P. aeruginosa biofilms on plasma-coated surfaces and increased the survival of Caenorhabditis elegans infected with K. pneumoniae or P. aeruginosa. When assessing the antibacterial activity of the Ga(III)/antibiotic combinations, GaPP/colistin and Ga(NO3)3/colistin combinations also showed synergistic activity against K. pneumoniae and P. aeruginosa. Our results demonstrate that GaPP and Ga(NO3)3 have significant synergistic effects against several important human bacterial pathogens through dual inhibition of iron and heme metabolism.


Bacteria/metabolism , Gallium/chemistry , Iron/metabolism , Porphyrins/pharmacology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/metabolism , Bacteria/drug effects , Biofilms/drug effects , Drug Synergism , Gallium/pharmacology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/metabolism , Metabolic Networks and Pathways/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Porphyrins/chemistry , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism
15.
Article En | MEDLINE | ID: mdl-30962346

Mycobacterium tuberculosis is the leading cause of morbidity and death resulting from infectious disease worldwide. The incredible disease burden, combined with the long course of drug treatment and an increasing incidence of antimicrobial resistance among M. tuberculosis isolates, necessitates novel drugs and drug targets for treatment of this deadly pathogen. Recent work has produced several promising clinical candidates targeting components of the electron transport chain (ETC) of M. tuberculosis, highlighting this pathway's potential as a drug target. Menaquinone is an essential component of the M. tuberculosis ETC, as it functions to shuttle electrons through the ETC to produce the electrochemical gradient required for ATP production for the cell. We show that inhibitors of MenA, a component of the menaquinone biosynthetic pathway, are highly active against M. tuberculosis MenA inhibitors are bactericidal against M. tuberculosis under both replicating and nonreplicating conditions, with 10-fold higher bactericidal activity against nutrient-starved bacteria than against replicating cultures. MenA inhibitors have enhanced activity in combination with bedaquiline, clofazimine, and inhibitors of QcrB, a component of the cytochrome bc1 oxidase. Together, these data support MenA as a viable target for drug treatment against M. tuberculosis MenA inhibitors not only kill M. tuberculosis in a variety of physiological states but also show enhanced activity in combination with ETC inhibitors in various stages of clinical trial testing.


Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Bacterial Proteins/metabolism , Clofazimine/pharmacology , Diarylquinolines/pharmacology , Electron Transport/drug effects , Microbial Sensitivity Tests , Mycobacterium tuberculosis/metabolism , Oxidation-Reduction/drug effects
16.
Article En | MEDLINE | ID: mdl-30782994

Iron/heme acquisition systems are critical for microorganisms to acquire iron from the human host, where iron sources are limited due to the nutritional immune system and insolubility of the ferric form of iron. Prior work has shown that a variety of gallium compounds can interfere with bacterial iron acquisition. This study explored the intra- and extracellular antimicrobial activities of gallium protoporphyrin (GaPP), gallium mesoporphyrin (GaMP), and nanoparticles encapsulating GaPP or GaMP against the Gram-negative pathogens Pseudomonas aeruginosa and Acinetobacter baumannii, including clinical isolates. All P. aeruginosa and A. baumannii isolates were susceptible to GaPP and GaMP, with MICs ranging from 0.5 to ∼32 µg/ml in iron-depleted medium. Significant intra- and extracellular growth inhibition was observed against P. aeruginosa cultured in macrophages at a gallium concentration of 3.3 µg/ml (5 µM) of all Ga(III) compounds, including nanoparticles. Nanoparticle formulations showed prolonged activity against both P. aeruginosa and A. baumannii in previously infected macrophages. When the macrophages were loaded with the nanoparticles 3 days prior to infection, there was a 5-fold decrease in growth of P. aeruginosa in the presence of single emulsion F127 copolymer nanoparticles encapsulating GaMP (eFGaMP). In addition, all Ga(III) porphyrins and nanoparticles showed significant intracellular and antibiofilm activity against both pathogens, with the nanoparticles exhibiting intracellular activity for 3 days. Ga nanoparticles also increased the survival rate of Caenorhabditis elegans nematodes infected by P. aeruginosa and A. baumannii Our results demonstrate that Ga nanoparticles have prolonged in vitro and in vivo activities against both P. aeruginosa and A. baumannii, including disruption of their biofilms.


Acinetobacter Infections/drug therapy , Acinetobacter baumannii/drug effects , Gallium/pharmacology , Heme/metabolism , Iron/metabolism , Nanoparticles/administration & dosage , Pseudomonas aeruginosa/drug effects , Acinetobacter Infections/metabolism , Acinetobacter Infections/microbiology , Acinetobacter baumannii/metabolism , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Cells, Cultured , Humans , Microbial Sensitivity Tests/methods , Protoporphyrins/metabolism , Pseudomonas aeruginosa/metabolism , THP-1 Cells
17.
Mol Pharm ; 15(3): 1215-1225, 2018 03 05.
Article En | MEDLINE | ID: mdl-29421865

The nontuberculous mycobacterial (NTM) pathogens, M. avium complex (MAC) and M. abscessus, can result in severe pulmonary infections. Current antibiotics confront significant challenges for treatment of these NTM infections due to emerging multidrug-resistance. Thus, development of new antibiotics targeted against these agents is needed. We examined the inhibitory activities of Ga(NO3)3, GaCl3, gallium meso-tetraphenylporphyrine (GaTP), and gallium nanoparticles (GaNP) against intra- and extracellular M. avium and M. abscessus. GaTP, an analogue of natural heme, inhibited growth of both M. avium and M. abscessus with MICs in Fe-free 7H9 media of 0.5 and 2 µg/mL, respectively. GaTP was more active than Ga(NO3)3 and GaCl3. Ga(NO3)3 and GaCl3 were not as active in Fe-rich media compared to Fe-free media. However, GaTP was much less impacted by exogenous Fe, with MICs against M. avium and M. abscessus of 2 and 4 µg/mL, respectively, in 7H9 OADC media (Fe rich). Confocal microscopy showed that GaNP penetrates the M. avium cell wall. As assessed by determining colony forming units, GaNP inhibited the growth of NTM growing in THP-1 macrophages up to 15 days after drug-loading of the cells, confirming a prolonged growth inhibitory activity of the GaNP. Biodistribution studies of GaNP conducted in mice showed that intraperitoneal injection is more effective than intramuscular injection in delivering Ga(III) into lung tissue. GaTP exhibits potential as a lead compound for development of anti-NTM agents that target heme-bound iron uptake mechanisms by mycobacteria and inhibit growth by disrupting mycobacterial iron acquisition/utilization.


Anti-Bacterial Agents/pharmacology , Gallium/pharmacology , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium abscessus/drug effects , Mycobacterium avium/drug effects , Respiratory Tract Infections/drug therapy , Animals , Anti-Bacterial Agents/therapeutic use , Cell Line, Tumor , Female , Gallium/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Models, Animal , Mycobacterium Infections, Nontuberculous/microbiology , Nanoparticles/chemistry , Porphyrins/chemistry , Respiratory Tract Infections/microbiology , Tissue Distribution
18.
Redox Biol ; 14: 465-473, 2018 04.
Article En | MEDLINE | ID: mdl-29080525

The glyoxalase pathway functions to detoxify reactive dicarbonyl compounds, most importantly methylglyoxal. The glyoxalase pathway is an antioxidant defense mechanism that is essential for neuroprotection. Excessive concentrations of methylglyoxal have deleterious effects on cells, leading to increased levels of inflammation and oxidative stress. Neurodegenerative diseases - including Alzheimer's, Parkinson's, Aging and Autism Spectrum Disorder - are often induced or exacerbated by accumulation of methylglyoxal. Antioxidant compounds possess several distinct mechanisms that enhance the glyoxalase pathway and function as neuroprotectants. Flavonoids are well-researched secondary plant metabolites that appear to be effective in reducing levels of oxidative stress and inflammation in neural cells. Novel flavonoids could be designed, synthesized and tested to protect against neurodegenerative diseases through regulating the glyoxalase pathway.


Antioxidants/therapeutic use , Flavonoids/therapeutic use , Lactoylglutathione Lyase/metabolism , Neurodegenerative Diseases/drug therapy , Neuroprotection/drug effects , Oxidative Stress/drug effects , Aging/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Antioxidants/pharmacology , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Flavonoids/pharmacology , Humans , Neurodegenerative Diseases/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Pyruvaldehyde/metabolism , Signal Transduction/drug effects
19.
Sci Rep ; 7(1): 5126, 2017 07 11.
Article En | MEDLINE | ID: mdl-28698611

Oxidative stress is damaging to cells and contributes to aging and neurodegenerative disease. This state is mediated by production of imbalanced molecules, and reactive dicarbonyl compounds - mainly methylglyoxal. The glyoxalase pathway is an antioxidant defense system utilized to detoxify methylglyoxal and neutralize free radicals. Pathway dysfunction leads to overproduction and accumulation of toxic, prooxidant compounds. We hypothesize flavonoid treatment as a means to enhance the glyoxalase pathway's ability to detoxify in neurons. This study found that flavonoid treatment in methylglyoxal treated cerebellar neurons increased the functioning of glyoxalase pathway by enhancing expression of glyoxalase-1 and glyoxalase-2 proteins, decreased cell death and increased cellular viability. Flavonoids also significantly contributed in the retention of synaptic functions (VGLUT1 and GAD65) in cerebellar neurons. In addition, flavonoids were found to be involved in pAkt - NF-κB signaling pathway through a reduction in phosphorylation of Akt. The data here show flavonoid compounds have the potential to protect the brain from aging and neurodegenerative disease.


Cerebellum/cytology , Flavonoids/pharmacology , Lactoylglutathione Lyase/metabolism , Thiolester Hydrolases/metabolism , Up-Regulation , Animals , Cell Death/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/metabolism , Mice , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress , Signal Transduction/drug effects , Vesicular Glutamate Transport Protein 1/metabolism
20.
PLoS One ; 12(5): e0177987, 2017.
Article En | MEDLINE | ID: mdl-28542623

New treatments and novel drugs are required to counter the growing problem of drug-resistant strains of Mycobacterium tuberculosis (M.tb). Our approach against drug resistant M.tb, as well as other intracellular pathogens, is by targeted drug delivery using nanoformulations of drugs already in use, as well as drugs in development. Among the latter are gallium (III) (Ga)-based compounds. In the current work, six different types of Ga and rifampin nanoparticles were prepared in such a way as to enhance targeting of M.tb infected-macrophages. They were then tested for their ability to inhibit growth of a fully pathogenic strain (H37Rv) or a non-pathogenic strain (H37Ra) of M.tb. Encapsulating Ga in folate- or mannose-conjugated block copolymers provided sustained Ga release for 15 days and significantly inhibited M.tb growth in human monocyte-derived macrophages. Nanoformulations with dendrimers encapsulating Ga or rifampin also showed promising anti-tuberculous activity. The nanoparticles co-localized with M.tb containing phagosomes, as measured by detection of mature cathepsin D (34 kDa, lysosomal hydrogenase). They also promoted maturation of the phagosome, which would be expected to increase macrophage-mediated killing of the organism. Delivery of Ga or rifampin in the form of nanoparticles to macrophages offers a promising approach for the development of new therapeutic anti-tuberculous drugs.


Gallium/pharmacology , Macrophages/drug effects , Macrophages/microbiology , Metal Nanoparticles/chemistry , Mycobacterium tuberculosis/drug effects , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Survival/drug effects , Cells, Cultured , Dendrimers/chemistry , Folic Acid/chemistry , Galectin 3/genetics , Galectin 3/metabolism , Gallium/analysis , Gallium/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Mannose/chemistry , Mycobacterium tuberculosis/physiology , Particle Size , Phagosomes/metabolism , Phagosomes/microbiology , Polymers/chemistry , Rifampin/chemistry , Rifampin/pharmacology , Virulence/drug effects
...