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

Publication year range
1.
Am J Respir Cell Mol Biol ; 70(2): 94-109, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37874230

ABSTRACT

Oxidative stress, inflammation, and endoplasmic reticulum (ER) stress sequentially occur in bronchopulmonary dysplasia (BPD), and all result in DNA damage. When DNA damage becomes irreparable, tumor suppressors increase, followed by apoptosis or senescence. Although cellular senescence contributes to wound healing, its persistence inhibits growth. Therefore, we hypothesized that cellular senescence contributes to BPD progression. Human autopsy lungs were obtained. Sprague-Dawley rat pups exposed to 95% oxygen between Postnatal Day 1 (P1) and P10 were used as the BPD phenotype. N-acetyl-lysyltyrosylcysteine-amide (KYC), tauroursodeoxycholic acid (TUDCA), and Foxo4 dri were administered intraperitoneally to mitigate myeloperoxidase oxidant generation, ER stress, and cellular senescence, respectively. Lungs were examined by histology, transcriptomics, and immunoblotting. Cellular senescence increased in rat and human BPD lungs, as evidenced by increased oxidative DNA damage, tumor suppressors, GL-13 stain, and inflammatory cytokines with decreased cell proliferation and lamin B expression. Cellular senescence-related transcripts in BPD rat lungs were enriched at P10 and P21. Single-cell RNA sequencing showed increased cellular senescence in several cell types, including type 2 alveolar cells. In addition, Foxo4-p53 binding increased in BPD rat lungs. Daily TUDCA or KYC, administered intraperitoneally, effectively decreased cellular senescence, improved alveolar complexity, and partially maintained the numbers of type 2 alveolar cells. Foxo4 dri administered at P4, P6, P8, and P10 led to outcomes similar to TUDCA and KYC. Our data suggest that cellular senescence plays an essential role in BPD after initial inducement by hyperoxia. Reducing myeloperoxidase toxic oxidant production, ER stress, and attenuating cellular senescence are potential therapeutic strategies for halting BPD progression.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Taurochenodeoxycholic Acid , Infant, Newborn , Animals , Rats , Humans , Bronchopulmonary Dysplasia/pathology , Hyperoxia/metabolism , Rats, Sprague-Dawley , Lung/pathology , Cellular Senescence , Peroxidase/metabolism , Oxidants , Animals, Newborn , Disease Models, Animal
2.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39337630

ABSTRACT

Bronchopulmonary dysplasia (BPD) is the most common lung complication of prematurity. Despite extensive research, our understanding of its pathophysiology remains limited, as reflected by the stable prevalence of BPD. Prematurity is the primary risk factor for BPD, with oxidative stress (OS) and inflammation playing significant roles and being closely linked to premature birth. Understanding the interplay and temporal relationship between OS and inflammation is crucial for developing new treatments for BPD. Animal studies suggest that OS and inflammation can exacerbate each other. Clinical trials focusing solely on antioxidants or anti-inflammatory therapies have been unsuccessful. In contrast, vitamin A and caffeine, with antioxidant and anti-inflammatory properties, have shown some efficacy, reducing BPD by about 10%. However, more than one-third of very preterm infants still suffer from BPD. New therapeutic agents are needed. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), is a reversible myeloperoxidase inhibitor and a systems pharmacology agent. It reduces BPD severity by inhibiting MPO, enhancing antioxidative proteins, and alleviating endoplasmic reticulum stress and cellular senescence in a hyperoxia rat model. KYC represents a promising new approach to BPD treatment.


Subject(s)
Bronchopulmonary Dysplasia , Inflammation , Oxidative Stress , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/drug therapy , Oxidative Stress/drug effects , Humans , Animals , Inflammation/metabolism , Inflammation/drug therapy , Antioxidants/pharmacology , Antioxidants/therapeutic use , Infant, Newborn , Infant, Premature , Rats , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology
3.
Arch Toxicol ; 96(1): 79-103, 2022 01.
Article in English | MEDLINE | ID: mdl-34773488

ABSTRACT

Chemicals are measured regularly in air, food, the environment, and the workplace. Biomonitoring of chemicals in biological fluids is a tool to determine the individual exposure. Blood protein adducts of xenobiotics are a marker of both exposure and the biologically effective dose. Urinary metabolites and blood metabolites are short term exposure markers. Stable hemoglobin adducts are exposure markers of up to 120 days. Blood protein adducts are formed with many xenobiotics at different sites of the blood proteins. Newer methods apply the techniques developed in the field of proteomics. Larger adducted peptides with 20 amino acids are used for quantitation. Unfortunately, at present the methods do not reach the limits of detection obtained with the methods looking at single amino acid adducts or at chemically cleaved adducts. Therefore, to progress in the field new approaches are needed.


Subject(s)
Biological Monitoring , Blood Proteins , Biomarkers , Hemoglobins/analysis , Proteomics , Xenobiotics/toxicity
4.
Chem Res Toxicol ; 33(8): 2010-2021, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32672951

ABSTRACT

Biomonitoring of xenobiotics has been performed for many years in occupational and environmental medicine. It has revealed hidden exposures and the exposure of workers could be reduced. Although most of the toxic effects of chemicals on humans were discovered in workers, the scientific community has more recently focused on environmental samples. In several countries, urinary and blood samples have been collected and analyzed for xenobiotics. Health, biochemical, and clinical parameters were measured in the biomonitoring program of the Unites States. The data were collected and evaluated as group values, comparing races, ages, and gender. The term exposome was created in order to relate chemical exposure to health effects together with the terms genome, proteome, and transcriptome. Internal exposures were mostly established with snapshot measurements, which can lead to an obvious misclassification of the individual exposures. Albumin and hemoglobin adducts of xenobiotics reflect the exposure of a larger time frame, up to 120 days. It is likely that only a small fraction of xenobiotics form such adducts. In addition, adduct analyses are more work intensive than the measurement of xenobiotics and metabolites in urine and/or blood. New technology, such as high-resolution mass spectrometry, will enable the discovery of new compounds that have been overlooked in the past, since over 300,000 chemicals are commercially available and most likely also present in the environment. Yet, quantification will be challenging, as it was for the older methods. At this stage, determination of a lifetime internal exposome is very unrealistic. Instead of an experimental approach with a large number of people, which is economically and scientifically not feasible, in silico methods should be developed further to predict exposure, toxicity, and potential health effects of mixtures. The computer models will help to focus internal exposure investigations on smaller groups of people and smaller number of chemicals.


Subject(s)
Environmental Exposure/analysis , Environmental Monitoring , Xenobiotics/analysis , Humans
5.
J Pharmacol Exp Ther ; 361(1): 39-50, 2017 04.
Article in English | MEDLINE | ID: mdl-28154014

ABSTRACT

Dual specificity mitogen-activated protein kinase (MAPK) phosphatases [dual specificity phosphatase/MAP kinase phosphatase (DUSP-MKP)] have been hypothesized to maintain cancer cell survival by buffering excessive MAPK signaling caused by upstream activating oncogenic products. A large and diverse body of literature suggests that genetic depletion of DUSP-MKPs can reduce tumorigenicity, suggesting that hyperactivating MAPK signaling by DUSP-MKP inhibitors could be a novel strategy to selectively affect the transformed phenotype. Through in vivo structure-activity relationship studies in transgenic zebrafish we recently identified a hyperactivator of fibroblast growth factor signaling [(E)-2-benzylidene-5-bromo-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI-215)] that is devoid of developmental toxicity and restores defective MAPK activity caused by overexpression of DUSP1 and DUSP6 in mammalian cells. Here, we hypothesized that BCI-215 could selectively affect survival of transformed cells. In MDA-MB-231 human breast cancer cells, BCI-215 inhibited cell motility, caused apoptosis but not primary necrosis, and sensitized cells to lymphokine-activated killer cell activity. Mechanistically, BCI-215 induced rapid and sustained phosphorylation of extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK) in the absence of reactive oxygen species, and its toxicity was partially rescued by inhibition of p38 but not JNK or ERK. BCI-215 also hyperactivated MKK4/SEK1, suggesting activation of stress responses. Kinase phosphorylation profiling documented BCI-215 selectively activated MAPKs and their downstream substrates, but not receptor tyrosine kinases, SRC family kinases, AKT, mTOR, or DNA damage pathways. Our findings support the hypothesis that BCI-215 causes selective cancer cell cytotoxicity in part through non-redox-mediated activation of MAPK signaling, and the findings also identify an intersection with immune cell killing that is worthy of further exploration.


Subject(s)
Breast Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/metabolism , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Female , HeLa Cells , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Killer Cells, Lymphokine-Activated/immunology , Mitogen-Activated Protein Kinase Phosphatases/immunology , Rats , Zebrafish
6.
J Org Chem ; 81(21): 10302-10320, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27447195

ABSTRACT

We report a second-generation synthesis of the exceedingly potent antimitotic agent N14-desacetoxytubulysin H (1) as well as the preparation of nine analogues of this lead structure. Highlights of our synthetic efforts include an efficient late-stage functionalization that allows for the preparation of new side-chain- and backbone-modified analogues. We also discovered C-terminal modifications that preserve the exquisite biological activity of acid 1 and offer the opportunity for effective conjugation to cell type-targeting moieties. All analogues had antiproliferative activities in the high picomolar to low nanomolar range and caused apoptosis and mitotic arrest as measured in a high content nuclear morphology assay. The ten synthetic agents described herein spanned a range of almost 4 orders of magnitude in biological activity and illustrate the continued potential to discover extraordinarily potent antiproliferative compounds based on natural product leads.


Subject(s)
Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Apoptosis/drug effects , Carbon-13 Magnetic Resonance Spectroscopy , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Mitosis/drug effects , Oligopeptides/chemistry , Proton Magnetic Resonance Spectroscopy
7.
Bioorg Med Chem Lett ; 26(5): 1480-1484, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26852364

ABSTRACT

The HIV-1 Nef accessory factor enhances viral replication and promotes immune system evasion of HIV-infected cells, making it an attractive target for drug discovery. Recently we described a novel class of diphenylpyrazolodiazene compounds that bind directly to Nef in vitro and inhibit Nef-dependent HIV-1 infectivity and replication in cell culture. However, these first-generation Nef antagonists have several structural liabilities, including an azo linkage that led to poor oral bioavailability. The azo group was therefore replaced with either a one- or two-carbon linker. The resulting set of non-azo analogs retained nanomolar binding affinity for Nef by surface plasmon resonance, while inhibiting HIV-1 replication with micromolar potency in cell-based assays without cytotoxicity. Computational docking studies show that these non-azo analogs occupy the same predicted binding site within the HIV-1 Nef dimer interface as the original azo compound. Computational methods also identified a hot spot for inhibitor binding within this site that is defined by conserved HIV-1 Nef residues Asp108, Leu112, and Pro122. Pharmacokinetic evaluation of the non-azo B9 analogs in mice showed that replacement of the azo linkage dramatically enhanced oral bioavailability without substantially affecting plasma half-life or clearance. The improved oral bioavailability of non-azo diphenylpyrazolo Nef antagonists provides a starting point for further drug lead optimization in support of future efficacy testing in animal models of HIV/AIDS.


Subject(s)
Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Small Molecule Libraries/pharmacology , nef Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Administration, Oral , Animals , Anti-HIV Agents/administration & dosage , Binding Sites/drug effects , Dose-Response Relationship, Drug , HIV-1/drug effects , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship , Tumor Cells, Cultured , Virus Replication/drug effects
8.
Mol Cell ; 32(5): 652-61, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-19061640

ABSTRACT

Ubiquitin-dependent proteolysis is an important mechanism that suppresses the beta-catenin transcription factor in cells without Wnt stimulation. A critical step in this regulatory pathway is to create a SCF(beta-TrCP) E3 ubiquitin ligase binding site for beta-catenin. Here we show that the SCF(beta-TrCP) binding site created by phosphorylation of beta-catenin is highly vulnerable to protein phosphatase 2A (PP2A) and must be protected by the adenomatous polyposis coli (APC) tumor suppressor protein. Specifically, phosphorylated beta-catenin associated with the wild-type APC protein is recruited to the SCF(beta-TrCP) complex, ubiquitin conjugated, and degraded. A mutation in APC that deprives this protective function exposes the N-terminal phosphorylated serine/threonine residues of beta-catenin to PP2A. Dephosphorylation at these residues by PP2A eliminates the SCF(beta-TrCP) recognition site and blocks beta-catenin ubiquitin conjugation. Thus, by acting to protect the E3 ligase binding site, APC ensures the ubiquitin conjugation of phosphorylated beta-catenin.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , beta Catenin/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Adenomatous Polyposis Coli Protein/chemistry , Animals , Binding Sites , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Mutation/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Phosphatase 2/deficiency , Protein Phosphatase 2/metabolism , Protein Processing, Post-Translational , Protein Transport , Substrate Specificity , Ubiquitin/metabolism , Ubiquitination , Xenopus
9.
Chembiochem ; 15(10): 1436-45, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24909879

ABSTRACT

Dual specificity phosphatase 6 (DUSP6) functions as a feedback attenuator of fibroblast growth factor signaling during development. In vitro high throughput chemical screening attempts to discover DUSP6 inhibitors have yielded limited success. However, in vivo whole-organism screens of zebrafish identified compound 1 (BCI) as an allosteric inhibitor of DUSP6. Here we designed and synthesized a panel of analogues to define the structure-activity relationship (SAR) of DUSP6 inhibition. In vivo high-content analysis in transgenic zebrafish, coupled with cell-based chemical complementation assays, identified structural features of the pharmacophore of 1 that were essential for biological activity. In vitro assays of DUSP hyperactivation corroborated the results from in vivo and cellular SAR. The results reinforce the notion that DUSPs are druggable through allosteric mechanisms and illustrate the utility of zebrafish as a model organism for in vivo SAR analyses.


Subject(s)
Dual Specificity Phosphatase 6/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indenes/chemistry , Indenes/pharmacology , Allosteric Regulation , Animals , Drug Design , Dual Specificity Phosphatase 6/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/metabolism , Models, Molecular , Signal Transduction/drug effects , Structure-Activity Relationship , Zebrafish/embryology
10.
Bioorg Med Chem Lett ; 24(7): 1702-6, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24650642

ABSTRACT

HIV-1 Nef is a critical AIDS progression factor yet underexplored target for antiretroviral drug discovery. A recent high-throughput screen for pharmacological inhibitors of Nef-dependent Src-family kinase activation identified a diphenylpyrazolodiazene hit compound with submicromolar potency in HIV-1 replication assays against a broad range of primary Nef variants. This compound, known as 'B9', binds directly to Nef and inhibits its dimerization in cells as a possible mechanism of action. Here were synthesized a diverse set of B9 analogs and identified structural features essential to antiretroviral activity. Chemical modifications to each of the three rings present in the parent compound were identified that did not compromise antiviral action. These analogs will guide the development of next-generation compounds with appropriate pharmacological profiles for assessment of antiretroviral activity in vivo.


Subject(s)
Anti-HIV Agents/pharmacology , Azo Compounds/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Pyrazoles/pharmacology , nef Gene Products, Human Immunodeficiency Virus/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Azo Compounds/chemical synthesis , Azo Compounds/chemistry , Cell Line , Dose-Response Relationship, Drug , HIV Infections/virology , HIV-1/genetics , Humans , Microbial Sensitivity Tests , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , nef Gene Products, Human Immunodeficiency Virus/genetics
11.
J Nat Prod ; 77(7): 1572-8, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24964362

ABSTRACT

The heterocyclic alkaloids, ceratamines A and B, are isolates from a marine Pseudoceratina sp. sponge. They behave as antimitotic agents, with IC50 values in the low micromolar range. The mechanism of this activity involves the disruption of microtubule dynamics; therefore, the ceratamines are of great interest in cancer drug discovery. Studies of in vitro metabolism were performed using rat liver microsomes to begin to understand the pharmacokinetics of these unique natural products. A total of eight metabolites were identified using UV and LC-MS/MS techniques. The majority of metabolites were formed as a result of various demethylation reactions. The formation of two metabolites, M1 and M3, involved monooxygenation, most likely on the aromatic ring, however the exact structure has not been determined. UV absorbance revealed a hypsochromic shift as a result of monooxygenation, an observation that may suggest the loss of aromaticity; however, further investigation is required. The structures of two major metabolites of ceratamine B, M4 and M6, were confirmed by (1)H NMR spectroscopy. These metabolites formed as a result of demethylation at the methoxy and aminoimidazole, respectively.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Azepines/isolation & purification , Azepines/pharmacology , Hydrocarbons, Brominated/isolation & purification , Hydrocarbons, Brominated/pharmacology , Imidazoles/isolation & purification , Imidazoles/pharmacology , Porifera/chemistry , Alkaloids/biosynthesis , Alkaloids/chemistry , Alkaloids/isolation & purification , Alzheimer Disease/drug therapy , Animals , Antineoplastic Agents/chemistry , Azepines/chemistry , Brain/drug effects , Hydrocarbons, Brominated/chemistry , Imidazoles/chemistry , Inhibitory Concentration 50 , Marine Biology , Microsomes, Liver/drug effects , Microtubules/drug effects , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Rats
12.
Planta Med ; 80(6): 473-81, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24710897

ABSTRACT

As a result of a program to find antitumor compounds of endophytes from medicinal Asteraceae, the steroid (22E,24R)-8,14-epoxyergosta-4,22-diene-3,6-dione (a) and the diterpene aphidicolin (b) were isolated from the filamentous fungi Papulaspora immersa and Nigrospora sphaerica, respectively, and exhibited strong cytotoxicity against HL-60 cells. A proteomic approach was used in an attempt to identify the drugs' molecular targets and their respective antiproliferative mode of action. Results suggested that the (a) growth inhibition effect occurs by G2/M cell cycle arrest via reduction of tubulin alpha and beta isomers and 14-3-3 protein gamma expression, followed by a decrease of apoptotic and inflammatory proteins, culminating in mitochondrial oxidative damage that triggered autophagy-associated cell death. Moreover, the decrease observed in the expression levels of several types of histones indicated that (a) might be disarming oncogenic pathways via direct modulation of the epigenetic machinery. Effects on cell cycle progression and induction of apoptosis caused by (b) were confirmed. In addition, protein expression profiles also revealed that aphidicolin is able to influence microtubule dynamics, modulate proteasome activator complex expression, and control the inflammatory cascade through overexpression of thymosin beta 4, RhoGDI2, and 14-3-3 proteins. Transmission electron micrographs of (b)-treated cells unveiled dose-dependent morphological characteristics of autophagy- or oncosis-like cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Aphidicolin/pharmacology , Endophytes/chemistry , Ergosterol/analogs & derivatives , Fungi/chemistry , Leukemia, Promyelocytic, Acute/metabolism , Proteome/metabolism , 14-3-3 Proteins/metabolism , Antineoplastic Agents/therapeutic use , Aphidicolin/therapeutic use , Asteraceae/chemistry , Biological Products/pharmacology , Biological Products/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Death , Ergosterol/pharmacology , Ergosterol/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Inflammation/metabolism , Inflammation/prevention & control , Leukemia, Promyelocytic, Acute/drug therapy , Microtubules/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress , Proteomics , Thymosin/metabolism , Tubulin/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
13.
J Am Soc Nephrol ; 24(6): 943-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23620402

ABSTRACT

At present, there are no effective therapies to ameliorate injury, accelerate recovery, or prevent postinjury fibrosis after AKI. Here, we sought to identify candidate compounds that accelerate recovery after AKI by screening for small molecules that increase proliferation of renal progenitor cells in zebrafish embryos. One compound identified from this screen was the histone deacetylase inhibitor methyl-4-(phenylthio)butanoate, which we subsequently administered to zebrafish larvae and mice 24-48 hours after inducing AKI. In zebrafish, treatment with the compound increased larval survival and proliferation of renal tubular epithelial cells. In mice, treatment accelerated recovery, reduced postinjury tubular atrophy and interstitial fibrosis, and increased the regenerative capacity of actively cycling renal tubular cells by decreasing the number of cells in G2/M arrest. These data suggest that accelerating recovery may be a viable approach to treating AKI and provide proof of concept that a screen in zebrafish embryos can identify therapeutic candidates for kidney injury.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/enzymology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Phenylbutyrates/pharmacology , Zebrafish Proteins/antagonists & inhibitors , Acute Kidney Injury/pathology , Animals , Disease Models, Animal , Fibrosis , Gentamicins/toxicity , Histone Deacetylase 1/metabolism , Ischemia/drug therapy , Ischemia/enzymology , Ischemia/pathology , Kidney/drug effects , Kidney/enzymology , Kidney/pathology , Male , Mice , Mice, Inbred BALB C , Protein Synthesis Inhibitors/toxicity , Recovery of Function/drug effects , Zebrafish , Zebrafish Proteins/metabolism
14.
Antioxidants (Basel) ; 13(8)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39199135

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a lung complication of premature births. The leading causes of BPD are oxidative stress (OS) from oxygen treatment, infection or inflammation, and mechanical ventilation. OS activates alveolar myeloid cells with subsequent myeloperoxidase (MPO)-mediated OS. Premature human neonates lack sufficient antioxidative capacity and are susceptible to OS. Unopposed OS elicits inflammation, endoplasmic reticulum (ER) stress, and cellular senescence, culminating in a BPD phenotype. Poor nutrition, patent ductus arteriosus, and infection further aggravate OS. BPD survivors frequently suffer from reactive airway disease, neurodevelopmental deficits, and inadequate exercise performance and are prone to developing early-onset chronic obstructive pulmonary disease. Rats and mice are commonly used to study BPD, as they are born at the saccular stage, comparable to human neonates at 22-36 weeks of gestation. The alveolar stage in rats and mice starts at the postnatal age of 5 days. Because of their well-established antioxidative capacities, a higher oxygen concentration (hyperoxia, HOX) is required to elicit OS lung damage in rats and mice. Neutrophil infiltration and ER stress occur shortly after HOX, while cellular senescence is seen later. Studies have shown that MPO plays a critical role in the process. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), a reversible MPO inhibitor, attenuates BPD effectively. In contrast, the irreversible MPO inhibitor-AZD4831-failed to provide similar efficacy. Interestingly, KYC cannot offer its effectiveness without the existence of MPO. We review the mechanisms by which this anti-MPO agent attenuates BPD.

15.
Front Oncol ; 14: 1406946, 2024.
Article in English | MEDLINE | ID: mdl-39165691

ABSTRACT

Introduction: Breast cancer (BC) is the most common cancer affecting women in the United States. Ductal carcinoma in situ (DCIS) is the earliest identifiable pre-invasive BC lesion. Estimates show that 14 to 50% of DCIS cases progress to invasive BC. Methods: Our objective was to identify nuclear matrix proteins (NMP) with specifically altered expression in DCIS and later stages of BC compared to non-diseased breast reduction mammoplasty and a contralateral breast explant culture using mass spectrometry and RNA sequencing to accurately identify aggressive DCIS. Results: Sixty NMPs were significantly differentially expressed between the DCIS and non-diseased breast epithelium in an isogenic contralateral pair of patient-derived extended explants. Ten of the sixty showed significant mRNA expression level differences that matched the protein expression. These 10 proteins were similarly expressed in non-diseased breast reduction cells. Three NMPs (RPL7A, RPL11, RPL31) were significantly upregulated in DCIS and all other BC stages compared to the matching contralateral breast culture and an unrelated non-diseased breast reduction culture. RNA sequencing analyses showed that these three genes were increasingly upregulated with BC progression. Finally, we identified three NMPs (AHNAK, CDC37 and DNAJB1) that were significantly downregulated in DCIS and all other BC stages compared to the isogenically matched contralateral culture and the non-diseased breast reduction culture using both proteomics and RNA sequencing techniques. Discussion: These genes should form the basis of, or contribute to, a molecular diagnostic panel that could identify DCIS lesions likely to be indolent and therefore not requiring aggressive treatment.

16.
bioRxiv ; 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38405693

ABSTRACT

Breast cancer (BC) is the most common cancer affecting women in the United States. Ductal carcinoma in situ (DCIS) is the earliest identifiable pre-invasive BC lesion. Estimates show that 14 to 50% of DCIS cases progress to invasive BC. Our objective was to identify nuclear matrix proteins (NMP) with specifically altered expression in DCIS and later stages of BC compared to non-diseased breast reduction mammoplasty and a contralateral breast explant using mass spectrometry and RNA sequencing to accurately identify aggressive DCIS. Sixty NMPs were significantly differentially expressed between the DCIS and non-diseased breast epithelium in an isogenic contralateral pair of patient-derived extended explants. Ten of the sixty showed significant mRNA expression level differences that matched the protein expression. These 10 proteins were similarly expressed in non-diseased breast reduction cells. Three NMPs (RPL7A, RPL11, RPL31) were significantly upregulated in DCIS and all other BC stages compared to the matching contralateral breast culture and an unrelated non-diseased breast reduction culture. RNA sequencing analyses showed that these three genes were upregulated increasingly with BC progression. Finally, we identified three NMPs (AHNAK, CDC37 and DNAJB1) that were significantly downregulated in DCIS and all other BC stages compared to the isogenically matched contralateral culture and the non-diseased breast reduction culture using both proteomics and RNA sequencing techniques.

17.
Free Radic Biol Med ; 215: 112-126, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336101

ABSTRACT

Murine sickle cell disease (SCD) results in damage to multiple organs, likely mediated first by vasculopathy. While the mechanisms inducing vascular damage remain to be determined, nitric oxide bioavailability and sterile inflammation are both considered to play major roles in vasculopathy. Here, we investigate the effects of high mobility group box-1 (HMGB1), a pro-inflammatory damage-associated molecular pattern (DAMP) molecule on endothelial-dependent vasodilation and lung morphometrics, a structural index of damage in sickle (SS) mice. SS mice were treated with either phosphate-buffered saline (PBS), hE-HMGB1-BP, an hE dual-domain peptide that binds and removes HMGB1 from the circulation via the liver, 1-[4-(aminocarbonyl)-2-methylphenyl]-5-[4-(1H-imidazol-1-yl)phenyl]-1H-pyrrole-2-propanoic acid (N6022) or N-acetyl-lysyltyrosylcysteine amide (KYC) for three weeks. Human umbilical vein endothelial cells (HUVEC) were treated with recombinant HMGB1 (r-HMGB1), which increases S-nitrosoglutathione reductase (GSNOR) expression by ∼80%, demonstrating a direct effect of HMGB1 to increase GSNOR. Treatment of SS mice with hE-HMGB1-BP reduced plasma HMGB1 in SS mice to control levels and reduced GSNOR expression in facialis arteries isolated from SS mice by ∼20%. These changes were associated with improved endothelial-dependent vasodilation. Treatment of SS mice with N6022 also improved vasodilation in SS mice suggesting that targeting GSNOR also improves vasodilation. SCD decreased protein nitrosothiols (SNOs) and radial alveolar counts (RAC) and increased GSNOR expression and mean linear intercepts (MLI) in lungs from SS mice. The marked changes in pulmonary morphometrics and GSNOR expression throughout the lung parenchyma in SS mice were improved by treating with either hE-HMGB1-BP or KYC. These data demonstrate that murine SCD induces vasculopathy and chronic lung disease by an HMGB1- and GSNOR-dependent mechanism and suggest that HMGB1 and GSNOR might be effective therapeutic targets for reducing vasculopathy and chronic lung disease in humans with SCD.


Subject(s)
Anemia, Sickle Cell , Benzamides , HMGB1 Protein , Lung Diseases , Lung Injury , Pyrroles , Vascular Diseases , Humans , Animals , Mice , Lung Injury/etiology , HMGB1 Protein/genetics , Endothelial Cells/metabolism , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Inflammation , Vascular Diseases/etiology
18.
Retrovirology ; 10: 135, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24229420

ABSTRACT

BACKGROUND: HIV-1 Nef is a viral accessory protein critical for AIDS progression. Nef lacks intrinsic catalytic activity and binds multiple host cell signaling proteins, including Hck and other Src-family tyrosine kinases. Nef binding induces constitutive Hck activation that may contribute to HIV pathogenesis by promoting viral infectivity, replication and downregulation of cell-surface MHC-I molecules. In this study, we developed a yeast-based phenotypic screen to identify small molecules that inhibit the Nef-Hck complex. RESULTS: Nef-Hck interaction was faithfully reconstituted in yeast cells, resulting in kinase activation and growth arrest. Yeast cells expressing the Nef-Hck complex were used to screen a library of small heterocyclic compounds for their ability to rescue growth inhibition. The screen identified a dihydrobenzo-1,4-dioxin-substituted analog of 2-quinoxalinyl-3-aminobenzene-sulfonamide (DQBS) as a potent inhibitor of Nef-dependent HIV-1 replication and MHC-I downregulation in T-cells. Docking studies predicted direct binding of DQBS to Nef which was confirmed in differential scanning fluorimetry assays with recombinant purified Nef protein. DQBS also potently inhibited the replication of HIV-1 NL4-3 chimeras expressing Nef alleles representative of all M-group HIV-1 clades. CONCLUSIONS: Our findings demonstrate the utility of a yeast-based growth reversion assay for the identification of small molecule Nef antagonists. Inhibitors of Nef function discovered with this assay, such as DQBS, may complement the activity of current antiretroviral therapies by enabling immune recognition of HIV-infected cells through the rescue of cell surface MHC-I.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Evaluation, Preclinical/methods , Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Quinoxalines/pharmacology , Saccharomyces cerevisiae/drug effects , Sulfonamides/pharmacology , nef Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/isolation & purification , Humans , Molecular Docking Simulation , Protein Binding/drug effects , Proto-Oncogene Proteins c-hck/genetics , Quinoxalines/isolation & purification , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sulfonamides/isolation & purification , nef Gene Products, Human Immunodeficiency Virus/genetics , Benzenesulfonamides
19.
Biometals ; 26(1): 151-65, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23344786

ABSTRACT

Complexes [Ga(2Ac4pFPh)(2)]NO(3) (1), [Ga(2Ac4pClPh)(2)]NO(3) (2), [Ga(2Ac4pIPh)(2)]NO(3) (3), [Ga(2Ac4pNO(2)Ph)(2)]NO(3)·3H(2)O (4) and [Ga(2Ac4pT)(2)]NO(3) (5) were obtained with 2-acetylpyridine N(4)-para-fluorophenyl-(H2Ac4pFPh), 2-acetylpyridine N(4)-para-chlorophenyl-(H2Ac4pClPh), 2-acetylpyridine N(4)-para-iodophenyl-(H2Ac4pIPh), 2-acetylpyridine N(4)-para-nitrophenyl-(H2Ac4pNO(2)Ph) and 2-acetylpyridine N(4)-para-tolyl-(H2Ac4pT) thiosemicarbazone. 1-5 presented antimicrobial and cytotoxic properties. Coordination to gallium(III) proved to be an effective strategy for activity improvement against Pseudomonas aeruginosa and Candida albicans. The complexes were highly cytotoxic against malignant glioblastoma and breast cancer cells at nanomolar concentrations. The compounds induced morphological changes characteristic of apoptotic death in tumor cells and showed no toxicity against erythrocytes. 2 partially inhibited tubulin assembly at high concentrations and induced cellular microtubule disorganization, but this does not appear to be the main mechanism of cytotoxic activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Gallium/chemistry , Thiosemicarbazones/chemistry , Tubulin/chemistry , Anti-Bacterial Agents/chemistry , Antifungal Agents/chemistry , Antineoplastic Agents/chemistry , Candida albicans/drug effects , Cell Cycle Checkpoints/drug effects , Cell Shape/drug effects , Coordination Complexes/chemistry , Crystallography, X-Ray , Erythrocytes/drug effects , HeLa Cells , Humans , Inhibitory Concentration 50 , Kinetics , MCF-7 Cells , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Protein Multimerization/drug effects , Pseudomonas aeruginosa/drug effects , Pyridines/chemistry , Staphylococcus aureus/drug effects , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
20.
J Org Chem ; 77(5): 2225-35, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22329423

ABSTRACT

The synthesis of neopeltolide analogues that contain variations in the oxazole-containing side chain and in the macrolide core are reported along with the GI(50) values for these compounds against MCF-7, HCT-116, and p53 knockout HCT-116 cell lines. Although biological activity is sensitive to changes in the macrocycle and the side chain, several analogues displayed GI(50) values of <25 nM. Neopeltolide and several of the more potent analogues were significantly less potent against p53 knockout cells, suggesting that p53 plays an auxiliary role in the activity of these compounds.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Macrolides/chemical synthesis , Macrolides/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Macrolides/chemistry , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL