Pub Date : 2025-02-14Epub Date: 2025-01-24DOI: 10.1021/acsinfecdis.4c00896
Paul J Hensbergen, Loes van Huijkelom, Jordy van Angeren, Arnoud H de Ru, Bart Claushuis, Peter A van Veelen, Wiep Klaas Smits, Jeroen Corver
Flagella are essential for motility and pathogenicity in many bacteria. The main component of the flagellar filament, flagellin (FliC), often undergoes post-translational modifications, with glycosylation being a common occurrence. In Pseudomonas aeruginosa PAO1, the b-type flagellin is O-glycosylated with a structure that includes a deoxyhexose, a phospho-group, and a previous unknown moiety. This structure resembles the well-characterized glycan (Type A) in Clostridioides difficile strain 630, which features an N-acetylglucosamine linked to an N-methylthreonine via a phosphodiester bond. This study aimed to characterize the b-type glycan structure in Pseudomonas aeruginosa PAO1 using a set of mass spectrometry experiments. For this purpose, we used wild-type P. aeruginosa PAO1 and several gene mutants from the b-type glycan biosynthetic cluster. Moreover, we compared the mass spectrometry characteristics of the b-type glycan with those of in vitro modified Type A-peptides from C. difficile strain 630Δerm. Our results demonstrate that the thus far unknown moiety of the b-type glycan in P. aeruginosa consists of an N,N-dimethylthreonine. These data allowed us to refine our model of the flagellin glycan biosynthetic pathway in both P. aeruginosa PAO1 and C. difficile strain 630.
{"title":"Elucidation of the Glycan Structure of the b-type Flagellin of <i>Pseudomonas aeruginosa</i> PAO1.","authors":"Paul J Hensbergen, Loes van Huijkelom, Jordy van Angeren, Arnoud H de Ru, Bart Claushuis, Peter A van Veelen, Wiep Klaas Smits, Jeroen Corver","doi":"10.1021/acsinfecdis.4c00896","DOIUrl":"10.1021/acsinfecdis.4c00896","url":null,"abstract":"<p><p>Flagella are essential for motility and pathogenicity in many bacteria. The main component of the flagellar filament, flagellin (FliC), often undergoes post-translational modifications, with glycosylation being a common occurrence. In <i>Pseudomonas aeruginosa</i> PAO1, the b-type flagellin is <i>O</i>-glycosylated with a structure that includes a deoxyhexose, a phospho-group, and a previous unknown moiety. This structure resembles the well-characterized glycan (Type A) in <i>Clostridioides difficile</i> strain 630, which features an <i>N</i>-acetylglucosamine linked to an <i>N</i>-methylthreonine via a phosphodiester bond. This study aimed to characterize the b-type glycan structure in <i>Pseudomonas aeruginosa</i> PAO1 using a set of mass spectrometry experiments. For this purpose, we used wild-type <i>P. aeruginosa</i> PAO1 and several gene mutants from the b-type glycan biosynthetic cluster. Moreover, we compared the mass spectrometry characteristics of the b-type glycan with those of <i>in vitro</i> modified Type A-peptides from <i>C. difficile</i> strain 630Δ<i>erm</i>. Our results demonstrate that the thus far unknown moiety of the b-type glycan in <i>P. aeruginosa</i> consists of an <i>N,N</i>-dimethylthreonine. These data allowed us to refine our model of the flagellin glycan biosynthetic pathway in both <i>P. aeruginosa</i> PAO1 and <i>C. difficile</i> strain 630.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":" ","pages":"518-528"},"PeriodicalIF":4.0,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11833859/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031496","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-14Epub Date: 2025-01-29DOI: 10.1021/acsinfecdis.4c00636
Xingyou Wang, Masha M Rosenberg, Youngchang Kim, Natalia Maltseva, Gregory D Cuny, Andrzej Joachimiak, Petr Kuzmič, Lizbeth Hedstrom
Inosine 5'-monophosphate dehydrogenase (IMPDH) is a promising antibiotic target. This enzyme catalyzes the NAD-dependent oxidation of inosine 5'-monophosphate (IMP) to xanthosine 5'-monophosphate (XMP), which is the rate-limiting step in guanine nucleotide biosynthesis. Bacterial IMPDH-specific inhibitors have been developed that bind to the NAD+ site. These inhibitors display varied affinities to different bacterial IMPDHs that are not easily rationalized by X-ray crystal structures of enzyme-inhibitor complexes. Inspection of X-ray crystal structures of 25 enzyme-inhibitor complexes, including 10 newly described, suggested that a mobile active site flap may be a structural determinant of inhibitor potency. Saturation transfer difference NMR experiments also suggested that the flap may contact the inhibitors to varying extents in different IMPDHs. Flap residue Leu413 contacted some inhibitors but was not structured in the crystal structures of other inhibitor complexes. The substitution of Leu413 with Phe or Ala in Bacillus anthracis IMPDH had inhibitor-selective effects, suggesting residue 413 could be a structural determinant of affinity. Curiously, the Ala substitution increased the potency of most inhibitors, even those that contacted Leu413 in the crystal structures. Presteady-state and steady-state kinetics experiments showed that the Leu413Ala substitution had comparable effects on inhibitor binding to the noncovalent E·IMP complex and the covalent intermediate E-XMP*, suggesting that the flap had similar interactions in both complexes. These results demonstrate that contacts do not necessarily indicate favorable interactions, and poorly structured mobile regions should not be discounted when assessing binding determinants.
{"title":"Role of the Mobile Active Site Flap in IMP Dehydrogenase Inhibitor Binding.","authors":"Xingyou Wang, Masha M Rosenberg, Youngchang Kim, Natalia Maltseva, Gregory D Cuny, Andrzej Joachimiak, Petr Kuzmič, Lizbeth Hedstrom","doi":"10.1021/acsinfecdis.4c00636","DOIUrl":"10.1021/acsinfecdis.4c00636","url":null,"abstract":"<p><p>Inosine 5'-monophosphate dehydrogenase (IMPDH) is a promising antibiotic target. This enzyme catalyzes the NAD-dependent oxidation of inosine 5'-monophosphate (IMP) to xanthosine 5'-monophosphate (XMP), which is the rate-limiting step in guanine nucleotide biosynthesis. Bacterial IMPDH-specific inhibitors have been developed that bind to the NAD<sup>+</sup> site. These inhibitors display varied affinities to different bacterial IMPDHs that are not easily rationalized by X-ray crystal structures of enzyme-inhibitor complexes. Inspection of X-ray crystal structures of 25 enzyme-inhibitor complexes, including 10 newly described, suggested that a mobile active site flap may be a structural determinant of inhibitor potency. Saturation transfer difference NMR experiments also suggested that the flap may contact the inhibitors to varying extents in different IMPDHs. Flap residue Leu413 contacted some inhibitors but was not structured in the crystal structures of other inhibitor complexes. The substitution of Leu413 with Phe or Ala in <i>Bacillus anthracis</i> IMPDH had inhibitor-selective effects, suggesting residue 413 could be a structural determinant of affinity. Curiously, the Ala substitution increased the potency of most inhibitors, even those that contacted Leu413 in the crystal structures. Presteady-state and steady-state kinetics experiments showed that the Leu413Ala substitution had comparable effects on inhibitor binding to the noncovalent E·IMP complex and the covalent intermediate E-XMP*, suggesting that the flap had similar interactions in both complexes. These results demonstrate that contacts do not necessarily indicate favorable interactions, and poorly structured mobile regions should not be discounted when assessing binding determinants.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":" ","pages":"442-452"},"PeriodicalIF":4.0,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11841048/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062130","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-14Epub Date: 2025-01-13DOI: 10.1021/acsinfecdis.4c00849
Amit Sharma, Sonali J Jain, Prabhat Nath Jha, Santosh Rudrawar, Sandip B Bharate, Hemant R Jadhav
Antimicrobial drug resistance is a significant global health challenge, causing hundreds of thousands of deaths annually and severely impacting healthcare systems worldwide. Several reported antimicrobial compounds have a guanidine motif, as the positive charge on guanidine promotes cell lysis. Therefore, pyrrole- and indole-based allylidene hydrazine carboximidamide derivatives with guanidine motifs are proposed as antimicrobial agents that mimic cationic antimicrobial peptides (CAMPs). A total of 72 derivatives having pyrrol-2-yl-phenyl allylidene hydrazine carboximidamide and indol-3-yl-phenyl allylidene hydrazine carboximidamide scaffolds were assessed for their inhibitory potential against a panel of Gram-positive and Gram-negative bacteria. Analogs 1j, 1k, 1s, 2j, 2q, 4a, 4c, 4h, 5b, 6a, and 6d exhibited potent broad-spectrum antimicrobial activity better than the standard antibiotics. Also, these compounds showed no cytotoxicity up to 3-fold of the minimum inhibitory concentration, and structure-activity relationship was established. Further, the most active compound, 6a, showed a strong biofilm disruption, acted on the bacterial membrane, and lysed it. The further development of these compounds as novel antimicrobial agents is warranted.
{"title":"Unfolding the Potential of Pyrrole- and Indole-Based Allylidene Hydrazine Carboximidamides as Antimicrobial Agents.","authors":"Amit Sharma, Sonali J Jain, Prabhat Nath Jha, Santosh Rudrawar, Sandip B Bharate, Hemant R Jadhav","doi":"10.1021/acsinfecdis.4c00849","DOIUrl":"10.1021/acsinfecdis.4c00849","url":null,"abstract":"<p><p>Antimicrobial drug resistance is a significant global health challenge, causing hundreds of thousands of deaths annually and severely impacting healthcare systems worldwide. Several reported antimicrobial compounds have a guanidine motif, as the positive charge on guanidine promotes cell lysis. Therefore, pyrrole- and indole-based allylidene hydrazine carboximidamide derivatives with guanidine motifs are proposed as antimicrobial agents that mimic cationic antimicrobial peptides (CAMPs). A total of 72 derivatives having pyrrol-2-yl-phenyl allylidene hydrazine carboximidamide and indol-3-yl-phenyl allylidene hydrazine carboximidamide scaffolds were assessed for their inhibitory potential against a panel of Gram-positive and Gram-negative bacteria. Analogs <b>1j</b>, <b>1k</b>, <b>1s</b>, <b>2j</b>, <b>2q</b>, <b>4a</b>, <b>4c</b>, <b>4h</b>, <b>5b</b>, <b>6a</b>, and <b>6d</b> exhibited potent broad-spectrum antimicrobial activity better than the standard antibiotics. Also, these compounds showed no cytotoxicity up to 3-fold of the minimum inhibitory concentration, and structure-activity relationship was established. Further, the most active compound, <b>6a</b>, showed a strong biofilm disruption, acted on the bacterial membrane, and lysed it. The further development of these compounds as novel antimicrobial agents is warranted.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":" ","pages":"493-505"},"PeriodicalIF":4.0,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142968651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-14Epub Date: 2025-01-28DOI: 10.1021/acsinfecdis.4c00806
Sapna Saini, Sunny Pal, Rashmi Sharma
Tuberculosis (TB), a leading infectious disease caused by the pathogen Mycobacterium tuberculosis, poses a significant treatment challenge due to its unique characteristics and resistance to existing drugs. The conventional treatment regimens, which are lengthy and involve multiple drugs, often result in poor patient adherence and subsequent drug resistance, particularly with multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains. This highlights the urgent need for novel anti-TB therapies and new drug targets. Antimicrobial peptides (AMPs), which are natural host defense molecules present in all living organisms, offer a promising alternative to traditional small-molecule drugs. AMPs have several advantages, including their broad-spectrum activity and the potential to circumvent existing resistance mechanisms. However, their clinical application faces challenges such as stability, delivery, and potential toxicity. This review aims to provide essential information on AMPs, including their sources, classification, mode of action, induction within the host under stress, efficacy against M. tuberculosis, clinical status and hurdles to their use. It also highlights future research directions to address these challenges and advance the development of AMP-based therapies for TB.
{"title":"Decoding the Role of Antimicrobial Peptides in the Fight against <i>Mycobacterium tuberculosis</i>.","authors":"Sapna Saini, Sunny Pal, Rashmi Sharma","doi":"10.1021/acsinfecdis.4c00806","DOIUrl":"10.1021/acsinfecdis.4c00806","url":null,"abstract":"<p><p>Tuberculosis (TB), a leading infectious disease caused by the pathogen <i>Mycobacterium tuberculosis</i>, poses a significant treatment challenge due to its unique characteristics and resistance to existing drugs. The conventional treatment regimens, which are lengthy and involve multiple drugs, often result in poor patient adherence and subsequent drug resistance, particularly with multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains. This highlights the urgent need for novel anti-TB therapies and new drug targets. Antimicrobial peptides (AMPs), which are natural host defense molecules present in all living organisms, offer a promising alternative to traditional small-molecule drugs. AMPs have several advantages, including their broad-spectrum activity and the potential to circumvent existing resistance mechanisms. However, their clinical application faces challenges such as stability, delivery, and potential toxicity. This review aims to provide essential information on AMPs, including their sources, classification, mode of action, induction within the host under stress, efficacy against <i>M. tuberculosis</i>, clinical status and hurdles to their use. It also highlights future research directions to address these challenges and advance the development of AMP-based therapies for TB.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":" ","pages":"350-365"},"PeriodicalIF":4.0,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143051040","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-14Epub Date: 2025-01-18DOI: 10.1021/acsinfecdis.4c00880
Iqball Faheem, Valakunja Nagaraja
Tuberculosis (TB) continues to be a major cause of death worldwide despite having an effective combinatorial therapeutic regimen and vaccine. Being one of the most successful human pathogens, Mycobacterium tuberculosis retains the ability to adapt to diverse intracellular and extracellular environments encountered by it during infection, persistence, and transmission. Designing and developing new therapeutic strategies to counter the emergence of multidrug-resistant and extensively drug-resistant TB remains a major task. DNA topoisomerases make up a unique class of ubiquitous enzymes that ensure steady-state level supercoiling and solve topological problems occurring during DNA transactions in cells. They continue to be attractive targets for the discovery of novel classes of antibacterials and to develop better molecules from existing drugs by virtue of their reaction mechanism. The limited repertoire of topoisomerases in M. tuberculosis, key differences in their properties compared to topoisomerases from other bacteria, their essentiality for the pathogen's survival, and validation as candidates for drug discovery provide an opportunity to exploit them in drug discovery efforts. The present review provides insights into their organization, structure, function, and regulation to further efforts in targeting them for new inhibitor discovery. First, the structure and biochemical properties of DNA gyrase and Topoisomerase I (TopoI) of mycobacteria are described compared to the well-studied counterparts from other bacteria. Next, we provide an overview of known inhibitors of DNA gyrase and emerging novel bacterial topoisomerase inhibitors (NBTIs). We also provide an update on TopoI-specific compounds, highlighting mycobacteria-specific inhibitors.
{"title":"Multifunctional Mycobacterial Topoisomerases with Distinctive Features.","authors":"Iqball Faheem, Valakunja Nagaraja","doi":"10.1021/acsinfecdis.4c00880","DOIUrl":"10.1021/acsinfecdis.4c00880","url":null,"abstract":"<p><p>Tuberculosis (TB) continues to be a major cause of death worldwide despite having an effective combinatorial therapeutic regimen and vaccine. Being one of the most successful human pathogens, <i>Mycobacterium tuberculosis</i> retains the ability to adapt to diverse intracellular and extracellular environments encountered by it during infection, persistence, and transmission. Designing and developing new therapeutic strategies to counter the emergence of multidrug-resistant and extensively drug-resistant TB remains a major task. DNA topoisomerases make up a unique class of ubiquitous enzymes that ensure steady-state level supercoiling and solve topological problems occurring during DNA transactions in cells. They continue to be attractive targets for the discovery of novel classes of antibacterials and to develop better molecules from existing drugs by virtue of their reaction mechanism. The limited repertoire of topoisomerases in <i>M. tuberculosis</i>, key differences in their properties compared to topoisomerases from other bacteria, their essentiality for the pathogen's survival, and validation as candidates for drug discovery provide an opportunity to exploit them in drug discovery efforts. The present review provides insights into their organization, structure, function, and regulation to further efforts in targeting them for new inhibitor discovery. First, the structure and biochemical properties of DNA gyrase and Topoisomerase I (TopoI) of mycobacteria are described compared to the well-studied counterparts from other bacteria. Next, we provide an overview of known inhibitors of DNA gyrase and emerging novel bacterial topoisomerase inhibitors (NBTIs). We also provide an update on TopoI-specific compounds, highlighting mycobacteria-specific inhibitors.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":" ","pages":"366-385"},"PeriodicalIF":4.0,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142995994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-14Epub Date: 2025-01-19DOI: 10.1021/acsinfecdis.4c00864
Victoria Banas, Mostafa A Elfawal, Bruce A Rosa, Matthew Mahoney, Jacquelyn Kauffman, Emily Goetz, Paulina Chen, Raffi V Aroian, Makedonka Mitreva, James W Janetka
Soil-transmitted helminth (STH) infections affect one-fourth of the global population and pose a significant threat to human and animal health, with limited treatment options and emerging drug resistance. Trichuris trichiura (whipworm) stands out as a neglected disease, necessitating new drugs to address this unmet medical need. We discovered that several different chemical series of related human Provirus Integration sites for Moloney murine leukemia virus (PIM) family kinase inhibitors possess potent anthelmintic activity by using whole-worm motility assays. Systematic structure-activity relationship (SAR) studies based on the pan-PIM kinase inhibitor CX-6258 were conducted to identify compounds displaying improved in vitro motility inhibition of both adult hookworm (Ancylostoma ceylanicum) and adult whipworm (Trichuris muris) nematodes. A broad kinase selectivity screen of >450 human kinases confirms PIM1 kinase and others as potential targets for CX-6258 and analogues thereof. In addition, we demonstrated that CX-6258 significantly reduced worm burden and egg counts in the T. muris infection model of mice, establishing it as a new oral small molecule anthelmintic therapeutic.
{"title":"Discovery of Human PIM Kinase Inhibitors as a Class of Anthelmintic Drugs to Treat Intestinal Nematode Infections.","authors":"Victoria Banas, Mostafa A Elfawal, Bruce A Rosa, Matthew Mahoney, Jacquelyn Kauffman, Emily Goetz, Paulina Chen, Raffi V Aroian, Makedonka Mitreva, James W Janetka","doi":"10.1021/acsinfecdis.4c00864","DOIUrl":"10.1021/acsinfecdis.4c00864","url":null,"abstract":"<p><p>Soil-transmitted helminth (STH) infections affect one-fourth of the global population and pose a significant threat to human and animal health, with limited treatment options and emerging drug resistance. <i>Trichuris trichiura</i> (whipworm) stands out as a neglected disease, necessitating new drugs to address this unmet medical need. We discovered that several different chemical series of related human Provirus Integration sites for Moloney murine leukemia virus (PIM) family kinase inhibitors possess potent anthelmintic activity by using whole-worm motility assays. Systematic structure-activity relationship (SAR) studies based on the <i>pan</i>-PIM kinase inhibitor CX-6258 were conducted to identify compounds displaying improved <i>in vitro</i> motility inhibition of both adult hookworm (<i>Ancylostoma ceylanicum</i>) and adult whipworm (<i>Trichuris muris</i>) nematodes. A broad kinase selectivity screen of >450 human kinases confirms PIM1 kinase and others as potential targets for CX-6258 and analogues thereof. In addition, we demonstrated that CX-6258 significantly reduced worm burden and egg counts in the <i>T. muris</i> infection model of mice, establishing it as a new oral small molecule anthelmintic therapeutic.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":" ","pages":"506-517"},"PeriodicalIF":4.0,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142995991","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1021/acsinfecdis.4c0101610.1021/acsinfecdis.4c01016
Wenrui Li, Chuan Hao Tan, Jong-Suep Baek, Lai Jiang, Noele Kai Jing Ng, Kelvin Kian Long Chong, Jun Jie Wong, Liheng Gao, Kimberly A. Kline and Say Chye Joachim Loo*,
Methicillin-resistant Staphylococcus aureus (MRSA) is a significant concern for skin and soft tissue infections. Apart from biofilm formation, these bacteria can reside intracellularly in phagocytic and nonphagocytic mammalian cells, complicating treatment with conventional antibiotics. Lipid-polymer hybrid nanoparticle (LPN) systems, combining the advantages of polymeric nanoparticles and liposomes, represent a new generation of nanocarriers with the potential to address these therapeutic challenges. In this study, gentamicin (Gen) and vancomycin (Van) were encapsulated in LPNs and evaluated for their ability to eliminate intracellular MRSA in phagocytic macrophage RAW-Blue cells and nonphagocytic epithelial HaCaT cells. Compared to free antibiotics at 100 μg/mL, LPN formulations significantly reduced intracellular bacterial loads in both cell lines. Specifically, LPN-Van resulted in approximately 0.7 Log CFU/well reduction in RAW-Blue cells and 0.3 Log CFU/well reduction in HaCaT cells. LPN-Gen showed a more pronounced reduction, with approximately 1.26 Log CFU/well reduction in RAW-Blue cells and 0.45 Log CFU/well reduction in HaCaT cells. In vivo, LPN-Van at 500 μg/mL significantly reduced MRSA biofilm viability compared to untreated controls (p < 0.001), achieving 98% eradication based on median values. In comparison, free vancomycin achieved a nonstatistically significant 79.2% reduction in biofilm viability compared to control. Prophylactically, LPN-Van at 500 μg/mL decreased MRSA levels to the limit of detection, resulting in a ∼3.5 Log reduction in the median CFU/wound compared to free vancomycin. No acute dermal toxicity was observed for LPN-Van based on histological analysis. These data indicate that LPNs show promise as a drug delivery platform technology to address intracellular infections.
{"title":"Anti-Intracellular MRSA Activity of Antibiotic-Loaded Lipid-Polymer Hybrid Nanoparticles and Their Effectiveness in Murine Skin Wound Infection Models","authors":"Wenrui Li, Chuan Hao Tan, Jong-Suep Baek, Lai Jiang, Noele Kai Jing Ng, Kelvin Kian Long Chong, Jun Jie Wong, Liheng Gao, Kimberly A. Kline and Say Chye Joachim Loo*, ","doi":"10.1021/acsinfecdis.4c0101610.1021/acsinfecdis.4c01016","DOIUrl":"https://doi.org/10.1021/acsinfecdis.4c01016https://doi.org/10.1021/acsinfecdis.4c01016","url":null,"abstract":"<p >Methicillin-resistant <i>Staphylococcus aureus</i> (MRSA) is a significant concern for skin and soft tissue infections. Apart from biofilm formation, these bacteria can reside intracellularly in phagocytic and nonphagocytic mammalian cells, complicating treatment with conventional antibiotics. Lipid-polymer hybrid nanoparticle (LPN) systems, combining the advantages of polymeric nanoparticles and liposomes, represent a new generation of nanocarriers with the potential to address these therapeutic challenges. In this study, gentamicin (Gen) and vancomycin (Van) were encapsulated in LPNs and evaluated for their ability to eliminate intracellular MRSA in phagocytic macrophage RAW-Blue cells and nonphagocytic epithelial HaCaT cells. Compared to free antibiotics at 100 μg/mL, LPN formulations significantly reduced intracellular bacterial loads in both cell lines. Specifically, LPN-Van resulted in approximately 0.7 Log CFU/well reduction in RAW-Blue cells and 0.3 Log CFU/well reduction in HaCaT cells. LPN-Gen showed a more pronounced reduction, with approximately 1.26 Log CFU/well reduction in RAW-Blue cells and 0.45 Log CFU/well reduction in HaCaT cells. In vivo, LPN-Van at 500 μg/mL significantly reduced MRSA biofilm viability compared to untreated controls (<i>p</i> < 0.001), achieving 98% eradication based on median values. In comparison, free vancomycin achieved a nonstatistically significant 79.2% reduction in biofilm viability compared to control. Prophylactically, LPN-Van at 500 μg/mL decreased MRSA levels to the limit of detection, resulting in a ∼3.5 Log reduction in the median CFU/wound compared to free vancomycin. No acute dermal toxicity was observed for LPN-Van based on histological analysis. These data indicate that LPNs show promise as a drug delivery platform technology to address intracellular infections.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 3","pages":"750–761 750–761"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143608877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13DOI: 10.1021/acsinfecdis.4c0076710.1021/acsinfecdis.4c00767
Krishna Jadhav, Agrim Jhilta, Raghuraj Singh, Swarnima Negi, Shweta Sharma, Rahul Shukla, Amit Kumar Singh* and Rahul Kumar Verma*,
Mycobacterium tuberculosis (Mtb) demonstrates a proclivity for infecting extrapulmonary sites, notably the brain. Treating these extrapulmonary tuberculosis (TB) manifestations is challenging due to the difficulty of drug delivery across the blood-brain barrier. Clofazimine (CLF) has exhibited promising activity against Mtb, including multidrug-resistant variants, in vitro and in preclinical animal models. However, its clinical implication is restricted owing to poor physicochemical and pharmacokinetic properties. This study aims to develop CLF nano-in-microparticles (CLF-NIMs) for brain drug delivery for central nervous system TB (CNS-TB) treatment via the intranasal route. Simultaneously, the potential dissemination of TB bacilli to the brain was investigated. Following treatment, colony-forming unit (CFU) enumeration was conducted in both the brain and lung tissues to assess mycobacterial burden. Concurrently, drug concentrations were quantified in serum, brain, and lung tissue, enabling a comprehensive evaluation of pharmacokinetics and tissue-specific drug distribution. In pharmacokinetic investigations of CLF-NIMs, significant accumulation of CLF was observed in brain tissue compared to orally administered CLF, surpassing the minimum inhibitory concentration of CLF. In a murine CNS-TB model, intranasal insufflation of CLF-NIMs for 4 weeks led to a substantial reduction (∼0.99 ± 0.57 Log10CFU/gram) in CFU count in the brain compared to oral administration of CLF (2.45 ± 0.47 Log10CFU/gram). These promising preclinical results indicate that CLF-NIMs are well-tolerated and exhibit significant anti-TB activity in a murine CNS-TB model.
{"title":"Pharmacokinetic and Pharmacodynamics of Clofazimine Nano-in-Microparticles: Enhanced Brain Delivery and CNS Tuberculosis Amelioration via Intranasal Administration","authors":"Krishna Jadhav, Agrim Jhilta, Raghuraj Singh, Swarnima Negi, Shweta Sharma, Rahul Shukla, Amit Kumar Singh* and Rahul Kumar Verma*, ","doi":"10.1021/acsinfecdis.4c0076710.1021/acsinfecdis.4c00767","DOIUrl":"https://doi.org/10.1021/acsinfecdis.4c00767https://doi.org/10.1021/acsinfecdis.4c00767","url":null,"abstract":"<p ><i>Mycobacterium tuberculosis</i> (Mtb) demonstrates a proclivity for infecting extrapulmonary sites, notably the brain. Treating these extrapulmonary tuberculosis (TB) manifestations is challenging due to the difficulty of drug delivery across the blood-brain barrier. Clofazimine (CLF) has exhibited promising activity against Mtb, including multidrug-resistant variants, in vitro and in preclinical animal models. However, its clinical implication is restricted owing to poor physicochemical and pharmacokinetic properties. This study aims to develop CLF nano-in-microparticles (CLF-NIMs) for brain drug delivery for central nervous system TB (CNS-TB) treatment via the intranasal route. Simultaneously, the potential dissemination of TB bacilli to the brain was investigated. Following treatment, colony-forming unit (CFU) enumeration was conducted in both the brain and lung tissues to assess mycobacterial burden. Concurrently, drug concentrations were quantified in serum, brain, and lung tissue, enabling a comprehensive evaluation of pharmacokinetics and tissue-specific drug distribution. In pharmacokinetic investigations of CLF-NIMs, significant accumulation of CLF was observed in brain tissue compared to orally administered CLF, surpassing the minimum inhibitory concentration of CLF. In a murine CNS-TB model, intranasal insufflation of CLF-NIMs for 4 weeks led to a substantial reduction (∼0.99 ± 0.57 Log10CFU/gram) in CFU count in the brain compared to oral administration of CLF (2.45 ± 0.47 Log10CFU/gram). These promising preclinical results indicate that CLF-NIMs are well-tolerated and exhibit significant anti-TB activity in a murine CNS-TB model.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 3","pages":"665–675 665–675"},"PeriodicalIF":4.0,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143608890","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-12DOI: 10.1021/acsinfecdis.4c0066610.1021/acsinfecdis.4c00666
Andrew Spaulding, Amrita Sharma, Miriam A. Giardini, Benjamin Hoffman, Jean A. Bernatchez, Laura-Isobel McCall, Claudia M. Calvet, Jasmin Ackermann, Julia M. Souza, Diane Thomas, Caroline C. Millard, William G. Devine, Baljinder Singh, Everton M. Silva, Susan E. Leed, Norma E. Roncal, Erica C. Penn, Jessey Erath, Gaurav Kumar, Yadira Sepulveda, Arnold Garcia, Ana Rodriguez, Nelly El-Sakkary, Richard J. Sciotti, Robert F. Campbell, Jeremiah D. Momper, James H. McKerrow, Conor R. Caffrey, Jair L. Siqueira-Neto, Michael P. Pollastri, Kojo Mensa-Wilmot and Lori Ferrins*,
Neglected tropical diseases such as Chagas disease, human African trypanosomiasis, leishmaniasis, and schistosomiasis have a significant global health impact in predominantly developing countries, although these diseases are spreading due to increased international travel and population migration. Drug repurposing with a focus on increasing antiparasitic potency and drug-like properties is a cost-effective and efficient route to the development of new therapies. Here we identify compounds that have potent activity against Trypanosoma cruzi and Leishmania donovani, and the latter were progressed into the murine model of infection. Despite the potent in vitro activity, there was no effect on parasitemia, necessitating further work to improve the pharmacokinetic properties of this series. Nonetheless, valuable insights have been obtained into the structure–activity and structure–property relationships of this compound series.
{"title":"Identification of Substituted 4-Aminocinnolines as Broad-Spectrum Antiparasitic Agents","authors":"Andrew Spaulding, Amrita Sharma, Miriam A. Giardini, Benjamin Hoffman, Jean A. Bernatchez, Laura-Isobel McCall, Claudia M. Calvet, Jasmin Ackermann, Julia M. Souza, Diane Thomas, Caroline C. Millard, William G. Devine, Baljinder Singh, Everton M. Silva, Susan E. Leed, Norma E. Roncal, Erica C. Penn, Jessey Erath, Gaurav Kumar, Yadira Sepulveda, Arnold Garcia, Ana Rodriguez, Nelly El-Sakkary, Richard J. Sciotti, Robert F. Campbell, Jeremiah D. Momper, James H. McKerrow, Conor R. Caffrey, Jair L. Siqueira-Neto, Michael P. Pollastri, Kojo Mensa-Wilmot and Lori Ferrins*, ","doi":"10.1021/acsinfecdis.4c0066610.1021/acsinfecdis.4c00666","DOIUrl":"https://doi.org/10.1021/acsinfecdis.4c00666https://doi.org/10.1021/acsinfecdis.4c00666","url":null,"abstract":"<p >Neglected tropical diseases such as Chagas disease, human African trypanosomiasis, leishmaniasis, and schistosomiasis have a significant global health impact in predominantly developing countries, although these diseases are spreading due to increased international travel and population migration. Drug repurposing with a focus on increasing antiparasitic potency and drug-like properties is a cost-effective and efficient route to the development of new therapies. Here we identify compounds that have potent activity against <i>Trypanosoma cruzi</i> and <i>Leishmania donovani</i>, and the latter were progressed into the murine model of infection. Despite the potent <i>in vitro</i> activity, there was no effect on parasitemia, necessitating further work to improve the pharmacokinetic properties of this series. Nonetheless, valuable insights have been obtained into the structure–activity and structure–property relationships of this compound series.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 3","pages":"584–599 584–599"},"PeriodicalIF":4.0,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acsinfecdis.4c00666","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143608928","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-11DOI: 10.1021/acsinfecdis.4c0068910.1021/acsinfecdis.4c00689
Taher Uddin, Jing Xia, Yong Fu, Case W. McNamara, Arnab K. Chatterjee and L. David Sibley*,
Toxoplasma gondii causes widespread chronic infections that are not cured by current treatments due to the inability to affect semidormant bradyzoite stages within tissue cysts. To identify compounds to eliminate chronic infection, we developed an HTS using a recently characterized strain of T. gondii that undergoes efficient conversion to bradyzoites in vitro. Stage-specific expression of luciferase was used to selectively monitor the growth inhibition of bradyzoites by the Library of Pharmacological Active Compounds, consisting of 1280 drug-like compounds. We identified 44 compounds with >50% inhibitory effects against bradyzoites, including new highly potent compounds, several of which have precedent for antimicrobial activity. Subsequent characterization of the compound sanguinarine sulfate revealed potent and rapid killing against in vitro-produced bradyzoites and bradyzoites harvested from chronically infected mice, including potent activity against intact cysts. These findings provide a platform for expanded screening and identify promising compounds for further preclinical development against T. gondii bradyzoites that are responsible for chronic infection.
{"title":"High-Throughput Repurposing Screen Reveals Compounds with Activity against Toxoplasma gondii Bradyzoites","authors":"Taher Uddin, Jing Xia, Yong Fu, Case W. McNamara, Arnab K. Chatterjee and L. David Sibley*, ","doi":"10.1021/acsinfecdis.4c0068910.1021/acsinfecdis.4c00689","DOIUrl":"https://doi.org/10.1021/acsinfecdis.4c00689https://doi.org/10.1021/acsinfecdis.4c00689","url":null,"abstract":"<p ><i>Toxoplasma gondii</i> causes widespread chronic infections that are not cured by current treatments due to the inability to affect semidormant bradyzoite stages within tissue cysts. To identify compounds to eliminate chronic infection, we developed an HTS using a recently characterized strain of <i>T. gondii</i> that undergoes efficient conversion to bradyzoites in vitro. Stage-specific expression of luciferase was used to selectively monitor the growth inhibition of bradyzoites by the Library of Pharmacological Active Compounds, consisting of 1280 drug-like compounds. We identified 44 compounds with >50% inhibitory effects against bradyzoites, including new highly potent compounds, several of which have precedent for antimicrobial activity. Subsequent characterization of the compound sanguinarine sulfate revealed potent and rapid killing against in vitro-produced bradyzoites and bradyzoites harvested from chronically infected mice, including potent activity against intact cysts. These findings provide a platform for expanded screening and identify promising compounds for further preclinical development against <i>T. gondii</i> bradyzoites that are responsible for chronic infection.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 3","pages":"600–609 600–609"},"PeriodicalIF":4.0,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acsinfecdis.4c00689","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143608882","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}