Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(3): 113852, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38427558

ABSTRACT

The NLRP3 inflammasome is essential for caspase-1 activation and the release of interleukin (IL)-1ß, IL-18, and gasdermin-D in myeloid cells. However, research on species-specific NLRP3's physiological impact is limited. We engineer mice with the human NLRP3 gene, driven by either the human or mouse promoter, via syntenic replacement at the mouse Nlrp3 locus. Both promoters facilitate hNLRP3 expression in myeloid cells, but the mouse promoter responds more robustly to LPS. Investigating the disease impact of differential NLRP3 regulation, we introduce the D305N gain-of-function mutation into both humanized lines. Chronic inflammation is evident with both promoters; however, CNS outcomes vary significantly. Despite poor response to LPS, the human promoter results in D305N-associated aseptic meningitis, mirroring human pathology. The mouse promoter, although leading to increased CNS expression post-LPS, does not induce meningitis in D305N mutants. Therefore, human-like NLRP3 expression may be crucial for accurate modeling of its role in disease pathogenesis.


Subject(s)
Hereditary Autoinflammatory Diseases , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Animals , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Lipopolysaccharides/pharmacology , Inflammasomes/metabolism , Inflammation , Syndrome , Interleukin-1beta/metabolism , Caspase 1/metabolism
2.
J Pharmacol Exp Ther ; 388(3): 798-812, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38253384

ABSTRACT

The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is a central regulator of innate immunity, essential for processing and release of interleukin-1ß and pyroptotic cell death. As endogenous NLRP3 activating triggers are hallmarks of many human chronic inflammatory diseases, inhibition of NLRP3 has emerged as a therapeutic target. Here we identify NDT-19795 as a novel carboxylic acid-containing NLRP3 activation inhibitor in both human and mouse monocytes and macrophages. Remarkably, conversion of the carboxylate to an isopropyl-ester (NT-0796) greatly enhances NLRP3 inhibitory potency in human monocytes. This increase is attributed to the ester-containing pharmacophore being more cell-penetrant than the acid species and, once internalized, the ester being metabolized to NDT-19795 by carboxylesterase-1 (CES-1). Mouse macrophages do not express CES-1, and NT-0796 is ineffective in these cells. Mice also contain plasma esterase (Ces1c) activity which is absent in humans. To create a more human-like model, we generated a mouse line in which the genome was modified, removing Ces1c and replacing this segment of DNA with the human CES-1 gene driven by a mononuclear phagocyte-specific promoter. We show human CES-1 presence in monocytes/macrophages increases the ability of NT-0796 to inhibit NLRP3 activation both in vitro and in vivo. As NLRP3 is widely expressed by monocytes/macrophages, the co-existence of CES-1 in these same cells affords a unique opportunity to direct ester-containing NLRP3 inhibitors precisely to target cells of interest. Profiling NT-0796 in mice humanized with respect to CES-1 biology enables critical modeling of the pharmacokinetics and pharmacodynamics of this novel therapeutic candidate. SIGNIFICANCE STATEMENT: Inhibition of NLRP3 represents a desirable therapeutic strategy for the treatment of multiple human disorders. In this study pharmacological properties of a structurally-novel, ester-containing NLRP3 inhibitor NT-0796 are characterized. To study pharmacodynamics of NT-0796 in vivo, a mouse line was engineered possessing more human-like traits with respect to carboxylesterase biology. In the context of these hCES-1 mice, NT-0796 serves as a more effective inhibitor of NLRP3 activation than the corresponding acid, highlighting the full translational potential of the ester strategy.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Humans , Animals , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyrin Domain , Inflammasomes/metabolism , Caspase 1/metabolism , Esters , Carboxylic Ester Hydrolases/metabolism , Interleukin-1beta/metabolism
3.
Environ Health Perspect ; 131(12): 127021, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38150313

ABSTRACT

BACKGROUND: Chronic exposure to inorganic arsenic (iAs) has been associated with type 2 diabetes (T2D). However, potential sex divergence and the underlying mechanisms remain understudied. iAs is not metabolized uniformly across species, which is a limitation of typical exposure studies in rodent models. The development of a new "humanized" mouse model overcomes this limitation. In this study, we leveraged this model to study sex differences in the context of iAs exposure. OBJECTIVES: The aim of this study was to determine if males and females exhibit different liver and adipose molecular profiles and metabolic phenotypes in the context of iAs exposure. METHODS: Our study was performed on wild-type (WT) 129S6/SvEvTac and humanized arsenic +3 methyl transferase (human AS3MT) 129S6/SvEvTac mice treated with 400 ppb of iAs via drinking water ad libitum. After 1 month, mice were sacrificed and the liver and gonadal adipose depots were harvested for iAs quantification and sequencing-based microRNA and gene expression analysis. Serum blood was collected for fasting blood glucose, fasting plasma insulin, and homeostatic model assessment for insulin resistance (HOMA-IR). RESULTS: We detected sex divergence in liver and adipose markers of diabetes (e.g., miR-34a, insulin signaling pathways, fasting blood glucose, fasting plasma insulin, and HOMA-IR) only in humanized (not WT) mice. In humanized female mice, numerous genes that promote insulin sensitivity and glucose tolerance in both the liver and adipose are elevated compared to humanized male mice. We also identified Klf11 as a putative master regulator of the sex divergence in gene expression in humanized mice. DISCUSSION: Our study underscored the importance of future studies leveraging the humanized mouse model to study iAs-associated metabolic disease. The findings suggested that humanized males are at increased risk for metabolic dysfunction relative to humanized females in the context of iAs exposure. Future investigations should focus on the detailed mechanisms that underlie the sex divergence. https://doi.org/10.1289/EHP12785.


Subject(s)
Arsenic , Arsenicals , Diabetes Mellitus, Type 2 , Insulin Resistance , Female , Male , Mice , Humans , Animals , Arsenic/analysis , Blood Glucose/analysis , Diabetes Mellitus, Type 2/chemically induced , Insulin , Obesity , Methyltransferases/genetics
4.
Sci Rep ; 13(1): 3660, 2023 03 04.
Article in English | MEDLINE | ID: mdl-36871058

ABSTRACT

Although mice are widely used to study adverse effects of inorganic arsenic (iAs), higher rates of iAs methylation in mice than in humans may limit their utility as a model organism. A recently created 129S6 mouse strain in which the Borcs7/As3mt locus replaces the human BORCS7/AS3MT locus exhibits a human-like pattern of iAs metabolism. Here, we evaluate dosage dependency of iAs metabolism in humanized (Hs) mice. We determined tissue and urinary concentrations and proportions of iAs, methylarsenic (MAs), and dimethylarsenic (DMAs) in male and female Hs and wild-type (WT) mice that received 25- or 400-ppb iAs in drinking water. At both exposure levels, Hs mice excrete less total arsenic (tAs) in urine and retain more tAs in tissues than WT mice. Tissue tAs levels are higher in Hs females than in Hs males, particularly after exposure to 400-ppb iAs. Tissue and urinary fractions of tAs present as iAs and MAs are significantly greater in Hs mice than in WT mice. Notably, tissue tAs dosimetry in Hs mice resembles human tissue dosimetry predicted by a physiologically based pharmacokinetic model. These data provide additional support for use of Hs mice in laboratory studies examining effects of iAs exposure in target tissues or cells.


Subject(s)
Arsenic , Arsenicals , Arsenites , Drinking Water , Humans , Female , Male , Animals , Mice , Methyltransferases
5.
PLoS Pathog ; 19(2): e1011168, 2023 02.
Article in English | MEDLINE | ID: mdl-36812267

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2), part of the renin-angiotensin system (RAS), serves as an entry point for SARS-CoV-2, leading to viral proliferation in permissive cell types. Using mouse lines in which the Ace2 locus has been humanized by syntenic replacement, we show that regulation of basal and interferon induced ACE2 expression, relative expression levels of different ACE2 transcripts, and sexual dimorphism in ACE2 expression are unique to each species, differ between tissues, and are determined by both intragenic and upstream promoter elements. Our results indicate that the higher levels of expression of ACE2 observed in the lungs of mice relative to humans may reflect the fact that the mouse promoter drives expression of ACE2 in populous airway club cells while the human promoter drives expression in alveolar type 2 (AT2) cells. In contrast to transgenic mice in which human ACE2 is expressed in ciliated cells under the control of the human FOXJ1 promoter, mice expressing ACE2 in club cells under the control of the endogenous Ace2 promoter show a robust immune response after infection with SARS-CoV-2, leading to rapid clearance of the virus. This supports a model in which differential expression of ACE2 determines which cell types in the lung are infected, and this in turn modulates the host response and outcome of COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Receptors, Virus , Animals , Humans , Mice , Angiotensin-Converting Enzyme 2/genetics , COVID-19/genetics , Mice, Transgenic , Receptors, Virus/genetics , SARS-CoV-2 , Viral Tropism
6.
J Allergy Clin Immunol ; 151(4): 966-975, 2023 04.
Article in English | MEDLINE | ID: mdl-36592703

ABSTRACT

BACKGROUND: Type 2 endotype asthma is driven by IL-4 and IL-13 signaling via IL-4Ra, which is highly expressed on airway epithelium, airway smooth muscle, and immunocytes in the respiratory mucosa, suggesting potential advantages of an inhalable antagonist. Lipocalin 1 (Lcn1), a 16 kDa protein abundant in human periciliary fluid, has a robust drug-like structure well suited to protein engineering, but it has never been used to make an inhaled Anticalin protein therapeutic. OBJECTIVES: We sought to reengineer Lcn1 into an inhalable IL-4Ra antagonist and assess its pharmacodynamic/kinetic profile. METHODS: Lcn1 was systematically modified by directed protein mutagenesis yielding a high-affinity, slowly dissociating, long-acting full antagonist of IL-4Ra designated PRS-060 with properties analogous to dupilumab, competitively antagonizing IL-4Ra-dependent cell proliferation, mucus induction, and eotaxin expression in vitro. Because PRS-060 displayed exquisite specificity for human IL-4Ra, with no cross-reactivity to rodents or higher primates, we created a new triple-humanized mouse model substituting human IL-4Ra, IL-4, and IL-13 at their correct syntenic murine loci to model clinical dosing. RESULTS: Inhaled PRS-060 strongly suppressed acute allergic inflammation indexes in triple-humanized mice with a duration of action longer than its bulk clearance, suggesting that it may act locally in the lung. CONCLUSION: Lcn1 can be reengineered into the Anticalin antagonist PRS-060 (elarekibep), exemplifying a new class of inhaled topical, long-acting therapeutic drugs with the potential to treat type 2 endotype asthma.


Subject(s)
Asthma , Interleukin-13 , Animals , Humans , Mice , Asthma/drug therapy , Disease Models, Animal , Interleukin-4/genetics , Lung , Proteins , Nebulizers and Vaporizers , Receptors, Interleukin-4/immunology
7.
J Am Heart Assoc ; 11(19): e026581, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36172956

ABSTRACT

Background A beneficial role for prostanoids in hypertension is suggested by clinical studies showing nonsteroidal anti-inflammatory drugs, which block the production of all prostanoids, cause sodium retention and exacerbate hypertension. Among prostanoids, prostaglandin E2 and its E-prostanoid receptor 4 receptor (EP4R) have been implicated in blood pressure control. Our previous study found that conditional deletion of EP4R from all tissues in adult mice exacerbates angiotensin II-dependent hypertension, suggesting a powerful effect of EP4R to resist blood pressure elevation. We also found that elimination of EP4R from vascular smooth muscle cells did not affect the severity of hypertension, suggesting nonvascular targets of prostaglandin E mediate this antihypertensive effect. Methods and Results Here we generated mice with cell-specific deletion of EP4R from macrophage-specific EP4 receptor knockouts or kidney epithelial cells (KEKO) to assess the contributions of EP4R in these cells to hypertension pathogenesis. Macrophage-specific EP4 receptor knockouts showed similar blood pressure responses to alterations in dietary sodium or chronic angiotensin II infusion as Controls. By contrast, angiotensin II-dependent hypertension was significantly augmented in KEKOs (mean arterial pressure: 146±3 mm Hg) compared with Controls (137±4 mm Hg; P=0.02), which was accompanied by impaired natriuresis in KEKOs. Because EP4R expression in the kidney is enriched in the collecting duct, we compared responses to amiloride in angiotensin II-infused KEKOs and Controls. Blockade of the epithelial sodium channel with amiloride caused exaggerated natriuresis in KEKOs compared with Controls (0.21±0.01 versus 0.15±0.02 mmol/24 hour per 20 g; P=0.015). Conclusions Our data suggest EP4R in kidney epithelia attenuates hypertension. This antihypertension effect of EP4R may be mediated by reducing the activity of the epithelial sodium channel, thereby promoting natriuresis.


Subject(s)
Hypertension , Receptors, Prostaglandin E, EP4 Subtype , Amiloride/therapeutic use , Angiotensin II/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Antihypertensive Agents/therapeutic use , Dinoprostone/metabolism , Epithelial Cells , Epithelial Sodium Channels/genetics , Hypertension/drug therapy , Kidney , Macrophages/metabolism , Mice , Prostaglandins , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sodium/metabolism , Sodium Chloride, Dietary/metabolism
8.
Am J Respir Cell Mol Biol ; 65(5): 500-512, 2021 11.
Article in English | MEDLINE | ID: mdl-34126877

ABSTRACT

Ozone (O3) is a prevalent air pollutant causing lung inflammation. Previous studies demonstrate that O3 oxidizes lipids, such as cholesterol, in the airway to produce oxysterols, such as secosterol A (SecoA), which are electrophiles that are capable of forming covalent linkages preferentially with lysine residues and that consequently modify protein function. The breadth of proteins modified by this oxysterol as well as the biological consequences in the lung are unknown. By using an alkynyl-tagged form of SecoA and shotgun proteomics, we identified 135 proteins as being modified in bronchial epithelial cells. Among them was NLRP2 (NLR family pyrin domain-containing protein 2), which forms an alkynyl-tagged SecoA-protein adduct at lysine residue 1019 (K1019) in the terminal leucine-rich repeat region, a known regulatory region for NLR proteins. NLRP2 expression in airway epithelial cells was characterized, and CRISPR-Cas9 knockout (KO) and shRNA knockdown of NLRP2 were used to determine its function in O3-induced inflammation. No evidence for NLPR2 inflammasome formation or an NLRP2-dependent increase in caspase-1 activity in response to O3 was observed. O3-induced proinflammatory gene expression for CXCL2 and CXCL8/IL8 was further enhanced in NLRP2-KO cells, suggesting a negative regulatory role. Reconstitution of NLRP2-KO cells with the NLRP2 K1019 mutated to arginine partially blocked SecoA adduction and enhanced O3-induced IL-8 release as compared with wild-type NLRP2. Together, our findings uncover NLRP2 as a highly abundant, key component of proinflammatory signaling pathways in airway epithelial cells and as a novel mediator of O3-induced inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Inflammation/metabolism , Oxysterols/metabolism , Ozone/adverse effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Substitution , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Bronchi/cytology , Epithelial Cells , Gene Expression Regulation/drug effects , Humans , Immunoblotting , Inflammasomes/metabolism , Inflammation/chemically induced , Inflammation/pathology , Interleukin-8/metabolism , Oxysterols/chemistry
9.
Nat Commun ; 12(1): 2574, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976140

ABSTRACT

Allergic asthma is characterized by elevated levels of IgE antibodies, type 2 cytokines such as interleukin-4 (IL-4) and IL-13, airway hyperresponsiveness (AHR), mucus hypersecretion and eosinophilia. Approved therapeutic monoclonal antibodies targeting IgE or IL-4/IL-13 reduce asthma symptoms but require costly lifelong administrations. Here, we develop conjugate vaccines against mouse IL-4 and IL-13, and demonstrate their prophylactic and therapeutic efficacy in reducing IgE levels, AHR, eosinophilia and mucus production in mouse models of asthma analyzed up to 15 weeks after initial vaccination. More importantly, we also test similar vaccines specific for human IL-4/IL-13 in mice expressing human IL-4/IL-13 and the related receptor, IL-4Rα, to find efficient neutralization of both cytokines and reduced IgE levels for at least 11 weeks post-vaccination. Our results imply that dual IL-4/IL-13 vaccination may represent a cost-effective, long-term therapeutic strategy for the treatment of allergic asthma as demonstrated in mouse models, although additional studies are warranted to assess its safety and feasibility.


Subject(s)
Asthma/therapy , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Vaccination/methods , Animals , Asthma/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Chronic Disease/therapy , Disease Models, Animal , Female , Humans , Injections, Intramuscular , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Transgenic , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
10.
Epigenetics ; 16(3): 289-299, 2021 03.
Article in English | MEDLINE | ID: mdl-32660355

ABSTRACT

Studies have suggested that abrogated expression of detoxification enzymes, UGT2B15 and UGT2B17, are associated with prostate tumour risk and progression. We investigated the role of EGF on the expression of these enzymes since it interacts with signalling pathways to also affect prostate tumour progression and is additionally associated with decreased DNA methylation. The expression of UGT2B15, UGT2B17, de novo methyltransferases, DNMT3A and DNMT3B was assessed in prostate cancer cells (LNCaP) treated with EGF, an EGFR inhibitor PD16893, and the methyltransferase inhibitor, 5-azacytidine, respectively. The results showed that EGF treatment decreased levels of expression of all four genes and that their expression was reversed by PD16893. Treatment with 5-azacytidine, markedly decreased expression of UGT2B15 and UGT2B17 over 85% as well as significantly decreased expression of DNMT3B, but not the expression of DNMT3A. DNMT3B siRNA treated LNCaP cells had decreased expression of UGT2B15 and UGT2B17, while DNMT3A siRNA treated cells had only moderately decreased UGT2B15 expression. Treatment with DNMT methyltransferase inhibitor, RG108, significantly decreased UGT2B17 expression. Additionally, methylation differences between prostate cancer samples and benign prostate samples from an Illumina 450K Methylation Array study were assessed. The results taken together suggest that hypomethylation of the UGT2B15 and UGT2B17 genes contributes to increased risk of prostate cancer and may provide a putative biomarker or epigenetic target for chemotherapeutics. Mechanistic studies are warranted to determine the role of the methylation marks in prostate cancer.


Subject(s)
DNA Methylation , Glucuronosyltransferase , Prostatic Neoplasms , Gene Expression Regulation, Neoplastic , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Male , Minor Histocompatibility Antigens/genetics , Prostatic Neoplasms/genetics
11.
Environ Health Perspect ; 128(8): 87003, 2020 08.
Article in English | MEDLINE | ID: mdl-32779937

ABSTRACT

BACKGROUND: Chronic exposure to inorganic arsenic (iAs) is a significant public health problem. Methylation of iAs by arsenic methyltransferase (AS3MT) controls iAs detoxification and modifies risks of iAs-induced diseases. Mechanisms underlying these diseases have been extensively studied using animal models. However, substantive differences between humans and laboratory animals in efficiency of iAs methylation have hindered the translational potential of the laboratory studies. OBJECTIVES: The goal of this study was to determine whether humanization of the As3mt gene confers a human-like pattern of iAs metabolism in mice. METHODS: We generated a mouse strain in which the As3mt gene along with the adjacent Borcs7 gene was humanized by syntenic replacement. We compared expression of the mouse As3mt and the human AS3MT and the rate and pattern of iAs metabolism in the wild-type and humanized mice. RESULTS: AS3MT expression in mouse tissues closely modeled that of human and differed substantially from expression of As3mt. Detoxification of iAs was much less efficient in the humanized mice than in wild-type mice. Profiles for iAs and its methylated metabolites in tissues and excreta of the humanized mice were consistent with those reported in humans. Notably, the humanized mice expressed both the full-length AS3MT that catalyzes iAs methylation and the human-specific AS3MTd2d3 splicing variant that has been linked to schizophrenia. CONCLUSIONS: These results suggest that AS3MT is the primary genetic locus responsible for the unique pattern of iAs metabolism in humans. Thus, the humanized mouse strain can be used to study the role of iAs methylation in the pathogenesis of iAs-induced diseases, as well as to evaluate the role of AS3MTd2d3 in schizophrenia. https://doi.org/10.1289/EHP6943.


Subject(s)
Arsenic/metabolism , Methyltransferases/metabolism , Animals , Arsenicals , Humans , Methyltransferases/genetics , Mice
12.
Chem Res Toxicol ; 33(8): 2043-2046, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32700902

ABSTRACT

To investigate the role of glutathione transferases (GSTs) in the metabolism of inorganic arsenic (iAs), we compared the disposition of iAs and its metabolites in wild-type mice and mice lacking genes encoding GST-P, -M and -T after exposure to 100 ppb iAs in drinking water. We found no differences between the two genotypes in the concentrations of total arsenic or arsenic species in urine, liver, and kidneys. No genotype-dependent differences were found in proportions of arsenicals in the tissues, and only small differences were observed in the urine. Thus, under these conditions, GST-P, -M and -T did not play a significant role in iAs metabolism in mice.


Subject(s)
Arsenic/metabolism , Animals , Arsenic/administration & dosage , Arsenic/analysis , Drinking Water/administration & dosage , Drinking Water/analysis , Drinking Water/metabolism , Environmental Exposure/analysis , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Mice
13.
Chem Biol Drug Des ; 96(5): 1180-1186, 2020 11.
Article in English | MEDLINE | ID: mdl-32562384

ABSTRACT

The mounting threat of multi-drug-resistant (MDR) bacteria places a tremendous strain on the antimicrobial clinical arsenal, forcing physicians to revert to near-obsolete antibiotics to treat otherwise intractable infections. Antibiotic adjuvant therapy has emerged as a viable alternative to the development of novel antimicrobial agents. This method uses combinations of an existing antibiotic and a non-antimicrobial small molecule, where the combination either breaks drug resistance or further potentiates antibiotic activity. Through a high-content screen of eukaryotic kinase inhibitors, our group previously identified two highly potent adjuvants that synergize with colistin, a cyclic, polycationic antimicrobial peptide that serves as a drug of last resort for the treatment of MDR Gram-negative bacterial infections. Cell signaling proteins implicated in colistin resistance mechanisms display both kinase and phosphatase activities. Herein, we explore the potential for eukaryotic phosphatase inhibitors to be repurposed as colistin adjuvants. From a panel of 48 unique structures, we discovered that the natural product kuwanon G breaks colistin resistance, while the non-antimicrobial macrolide ascomycin potentiates colistin in polymyxin-susceptible bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Gram-Negative Bacteria/drug effects , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Drug Resistance, Multiple, Bacterial/drug effects , Drug Synergism , Eukaryota/enzymology , Flavonoids/pharmacology , Gram-Negative Bacteria/growth & development , Microbial Sensitivity Tests , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology
14.
ChemMedChem ; 15(2): 210-218, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31756025

ABSTRACT

Infections caused by multidrug-resistant (MDR) bacteria, particularly Gram-negative bacteria, are an escalating global health threat. Often clinicians are forced to administer the last-resort antibiotic colistin; however, colistin resistance is becoming increasingly prevalent, giving rise to the potential for a situation in which there are no treatment options for MDR Gram-negative infections. The development of adjuvants that circumvent bacterial resistance mechanisms is a promising orthogonal approach to the development of new antibiotics. We recently disclosed that the known IKK-ß inhibitor IMD-0354 potently suppresses colistin resistance in several Gram-negative strains. In this study, we explore the structure-activity relationship (SAR) between the IMD-0354 scaffold and colistin resistance suppression, and identify several compounds with more potent activity than the parent against highly colistin-resistant strains of Acinetobacter baumannii and Klebsiella pneumoniae.


Subject(s)
Acinetobacter baumannii/drug effects , Adjuvants, Pharmaceutic/pharmacology , Anti-Bacterial Agents/pharmacology , Benzamides/pharmacology , Klebsiella pneumoniae/drug effects , Adjuvants, Pharmaceutic/chemical synthesis , Adjuvants, Pharmaceutic/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Benzamides/chemical synthesis , Benzamides/chemistry , Colistin/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Multiple, Bacterial/drug effects , Humans , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
15.
PLoS One ; 14(11): e0225449, 2019.
Article in English | MEDLINE | ID: mdl-31747445

ABSTRACT

Environmental and endogenous electrophiles cause tissue damage through their high reactivity with endogenous nucleophiles such as DNA, proteins, and lipids. Protection against damage is mediated by glutathione (GSH) conjugation, which can occur spontaneously or be facilitated by the glutathione S-transferase (GST) enzymes. To determine the role of GST enzymes in protection against electrophiles as well as the role of specific GST families in mediating this protection, we exposed mutant mouse lines lacking the GSTP, GSTM, and/or GSTT enzyme families to the model electrophile acrylamide, a ubiquitous dietary contaminant known to cause adverse effects in humans. An analysis of urinary metabolites after acute acrylamide exposure identified the GSTM family as the primary mediator of GSH conjugation to acrylamide. However, surprisingly, mice lacking only this enzyme family did not show increased toxicity after an acute acrylamide exposure. Therefore, GSH conjugation is not the sole mechanism by which GSTs protect against the toxicity of this substrate. Given the prevalence of null GST polymorphisms in the human population (approximately 50% for GSTM1 and 20-50% for GSTT1), a substantial portion of the population may also have impaired acrylamide metabolism. However, our study also defines a role for GSTP and/or GSTT in protection against acrylamide mediated toxicity. Thus, while the canonical detoxification function of GSTs may be impaired in GSTM null individuals, disease risk secondary to acrylamide exposure may be mitigated through non-canonical pathways involving members of the GSTP and/or GSTT families.


Subject(s)
Acrylamide/toxicity , Epoxy Compounds/toxicity , Gene Deletion , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Liver/pathology , Animals , Disease Models, Animal , Female , Glutathione/urine , Humans , Inactivation, Metabolic , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/enzymology , Male , Mice , Mutagenicity Tests
16.
J Exp Med ; 216(12): 2838-2853, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31558613

ABSTRACT

The role of lipids in inflammasome activation remains underappreciated. The phospholipid, platelet-activating factor (PAF), exerts multiple physiological functions by binding to a G protein-coupled seven-transmembrane receptor (PAFR). PAF is associated with a number of inflammatory disorders, yet the molecular mechanism underlying its proinflammatory function remains to be fully elucidated. We show that multiple PAF isoforms and PAF-like lipids can activate the inflammasome, resulting in IL-1ß and IL-18 maturation. This is dependent on NLRP3, ASC, caspase-1, and NEK7, but not on NLRC4, NLRP1, NLRP6, AIM2, caspase-11, or GSDMD. Inflammasome activation by PAF also requires potassium efflux and calcium influx but not lysosomal cathepsin or mitochondrial reactive oxygen species. PAF exacerbates peritonitis partly through inflammasome activation, but PAFR is dispensable for PAF-induced inflammasome activation in vivo or in vitro. These findings reveal that PAF represents a damage-associated signal that activates the canonical inflammasome independently of PAFR and provides an explanation for the ineffectiveness of PAFR antagonist in blocking PAF-mediated inflammation in the clinic.


Subject(s)
Inflammasomes/metabolism , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Calcium/metabolism , Caspase 1/metabolism , Furans/pharmacology , Heterocyclic Compounds, 4 or More Rings , Humans , Indenes , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Phosphate-Binding Proteins/metabolism , Potassium/metabolism , Sulfonamides/pharmacology , Sulfones
17.
ACS Infect Dis ; 5(10): 1764-1771, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31434474

ABSTRACT

Kinase inhibitors comprise a diverse cohort of chemical scaffolds that are active in multiple biological systems. Currently, thousands of eukaryotic kinase inhibitors are commercially available, have well-characterized targets, and often carry pharmaceutically favorable toxicity profiles. Recently, our group disclosed that derivatives of the natural product meridianin D, a known inhibitor of eukaryotic kinases, modulated behaviors of both Gram-positive and Gram-negative bacteria. Herein, we expand our exploration of kinase inhibitors in Gram-negative bacilli utilizing three commercially available kinase inhibitor libraries and, ultimately, identify two chemical structures that potentiate colistin (polymyxin E) in multiple strains. We report IMD-0354, an inhibitor of IKK-ß, as a markedly effective adjuvant in colistin-resistant bacteria and also describe AR-12 (OSU-03012), an inhibitor of pyruvate dehydrogenase kinase-1 (PDK-1), as a potentiator in colistin-sensitive strains. This report comprises the first description of the novel cross-reactivity of these molecules.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Colistin/pharmacology , Gram-Negative Bacteria/drug effects , Protein Kinase Inhibitors/pharmacology , Adjuvants, Pharmaceutic/chemistry , Benzamides/pharmacology , Cell Line , Colistin/chemistry , Drug Combinations , Drug Resistance, Multiple, Bacterial/drug effects , Eukaryota , Gram-Positive Bacteria/drug effects , I-kappa B Kinase/drug effects , Lipid A , Microbial Sensitivity Tests , Pyrazoles/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/drug effects , Sulfonamides/pharmacology
18.
ACS Med Chem Lett ; 10(5): 828-833, 2019 May 09.
Article in English | MEDLINE | ID: mdl-31098007

ABSTRACT

Antibiotic resistance has significantly increased since the beginning of the 21st century. Currently, the polymyxin colistin is typically viewed as the antibiotic of last resort for the treatment of multidrug resistant Gram-negative bacterial infections. However, increased colistin usage has resulted in colistin-resistant bacterial isolates becoming more common. The recent dissemination of plasmid-borne colistin resistance genes (mcr 1-8) into the human pathogen pool is further threatening to render colistin therapy ineffective. New methods to combat antibiotic resistant pathogens are needed. Herein, the utilization of a colistin-adjuvant combination that is effective against colistin-resistant bacteria is described. At 5 µM, the lead adjuvant, which is nontoxic to the bacteria alone, increases colistin efficacy 32-fold against bacteria containing the mcr-1 gene and effects a 1024-fold increase in colistin efficacy against bacteria harboring chromosomally encoded colistin resistance determinants; these combinations lower the colistin minimum inhibitory concentration (MIC) to or below clinical breakpoint levels (≤2 µg/mL).

19.
J Am Heart Assoc ; 8(4): e010745, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30764697

ABSTRACT

Background Prostaglandin E2 ( PGE 2) is a major prostanoid with multiple actions that potentially affect blood pressure ( BP ). PGE 2 acts through 4 distinct E-prostanoid ( EP ) receptor isoforms: EP 1 to EP 4. The EP 4 receptor ( EP 4R) promotes PGE 2-dependent vasodilation, but its role in the pathogenesis of hypertension is not clear. Methods and Results To address this issue, we studied mice after temporal- and cell-specific deletion of EP 4R. First, using a mouse line with loss of EP 4 expression induced universally after birth, we confirm that EP 4R mediates a major portion of the acute vasodilatory effects of infused PGE 2. In addition, EP 4 contributes to control of resting BP , which was increased by 5±1 mm Hg in animals with generalized deficiency of this receptor. We also show that EP 4 is critical for limiting elevations in BP caused by high salt feeding and long-term infusion of angiotensin II . To more precisely identify the mechanism for these actions, we generated mice in which EP 4R loss is induced after birth and is limited to smooth muscle. In these mice, acute PGE 2-dependent vasodilation was attenuated, indicating that this response is mediated by EP 4R in vascular smooth muscle cells. However, absence of EP 4R only in this vascular compartment had a paradoxical effect of lowering resting BP , whereas the protective effect of EP 4R on limiting angiotensin II-dependent hypertension was unaffected. Conclusions Taken together, our findings support a complex role for EP 4R in regulation of BP and in hypertension, which appears to involve actions of the EP 4R in tissues beyond vascular smooth muscle cells.


Subject(s)
Blood Pressure/physiology , Hypertension/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Vasodilation/physiology , Angiotensin II/administration & dosage , Animals , Dinoprostone/administration & dosage , Disease Models, Animal , Hypertension/drug therapy , Hypertension/physiopathology , Infusions, Intravenous , Mice , Mice, Transgenic , Rest , Signal Transduction , Vasoconstrictor Agents/administration & dosage , Vasodilation/drug effects
20.
Cell Rep ; 24(5): 1254-1265, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067980

ABSTRACT

Lysosomes play a critical role in maintenance of the integrity of neuronal function, and mutations in genes that contribute to lysosome formation, transport, and activity are associated with neurodegenerative disorders. Recently, the multisubunit complex, BLOC-one-related complex (BORC), has been shown to be involved in positioning lysosomes within the cytoplasm, although the consequences of altered BORC function in adult animals have not been established. We show that a spontaneous truncation mutation in the mouse Borcs7 gene, identified through whole-genome sequencing followed by genetic complementation, results in progressive axonal dystrophy with dramatic impairment of motor function. Furthermore, mice homozygous for deletion of the entire Borcs7 coding sequence die shortly after birth, and neurons cultured from these animals show impaired centrifugal transport of lysosomes. This identifies BORCS7 as a central factor in axonal transport of lysosomes and a possible target for improving disease-related disturbances in this important function.


Subject(s)
Axonal Transport , Lysosomal Membrane Proteins/genetics , Neuroaxonal Dystrophies/genetics , Animals , Female , Locomotion , Loss of Function Mutation , Lysosomes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...