Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 556(7699): 103-107, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29590091

ABSTRACT

A challenge in the treatment of Staphylococcus aureus infections is the high prevalence of methicillin-resistant S. aureus (MRSA) strains and the formation of non-growing, dormant 'persister' subpopulations that exhibit high levels of tolerance to antibiotics and have a role in chronic or recurrent infections. As conventional antibiotics are not effective in the treatment of infections caused by such bacteria, novel antibacterial therapeutics are urgently required. Here we used a Caenorhabditis elegans-MRSA infection screen to identify two synthetic retinoids, CD437 and CD1530, which kill both growing and persister MRSA cells by disrupting lipid bilayers. CD437 and CD1530 exhibit high killing rates, synergism with gentamicin, and a low probability of resistance selection. All-atom molecular dynamics simulations demonstrated that the ability of retinoids to penetrate and embed in lipid bilayers correlates with their bactericidal ability. An analogue of CD437 was found to retain anti-persister activity and show an improved cytotoxicity profile. Both CD437 and this analogue, alone or in combination with gentamicin, exhibit considerable efficacy in a mouse model of chronic MRSA infection. With further development and optimization, synthetic retinoids have the potential to become a new class of antimicrobials for the treatment of Gram-positive bacterial infections that are currently difficult to cure.


Subject(s)
Anti-Bacterial Agents/classification , Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Retinoids/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Benzoates/chemistry , Benzoates/pharmacology , Benzoates/therapeutic use , Benzoates/toxicity , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Cell Death/drug effects , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Gentamicins/pharmacology , Gentamicins/therapeutic use , Humans , Lipid Bilayers/chemistry , Methicillin-Resistant Staphylococcus aureus/cytology , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Mutation , Naphthols/chemistry , Naphthols/pharmacology , Naphthols/therapeutic use , Naphthols/toxicity , Retinoids/chemistry , Retinoids/therapeutic use , Retinoids/toxicity
2.
Cell Commun Signal ; 17(1): 31, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30971268

ABSTRACT

BACKGROUND: p21-activated kinase 1 (PAK1) plays a fundamental role in promoting the development and progression of several cancers and is a potential therapeutic target. However, the biological function and underlying mechanism of PAK1 in esophageal squamous cell carcinoma (ESCC) remain unclear. METHODS: The expression of PAK1 was detected in both ESCC cell lines and clinical samples. Cell growth was measured by MTT, focus formation and soft agar assays. Cell migration and invasion were detected by wound healing and transwell assays. Animal models of subcutaneous tumourigenicity and tail vein metastasis were performed to determine the inhibitory effect of pharmacological inhibitor IPA-3 on tumor growth and metastasis of ESCC cells. RESULTS: We found that PAK1 was frequently overexpressed in ESCC. Ectopic expression of PAK1 promoted cellular growth, colony formation and anchorage-independent growth. Overexpressing PAK1 also enhanced migration, invasion and the expression of MMP-2 and MMP-9 in ESCC cells. In contrast, silencing PAK1 by lentiviral knockdown or a specific inhibitor IPA-3 resulted in a contrary effect. Subsequent investigations revealed that Raf1/MEK1/ERK signaling pathway was involved in PAK1-mediated effect. Enhanced expression of Raf1 attenuated the inhibitory functions of PAK1 shRNA. Whereas blocking of Raf1 by shRNA or specific inhibition of MEK1 by U0126 antagonized the oncogenetic effect of PAK1 on ESCC cells. More importantly, Pharmacological inhibition of PAK1 by IPA-3 significantly suppressed tumor growth and lung metastasis of ESCC cells in vivo. CONCLUSIONS: These data support that PAK1 is an ideal target for the development of potential therapeutic drugs for ESCC patients even with metastasis.


Subject(s)
Disulfides/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Naphthols/pharmacology , Protein Kinase Inhibitors/pharmacology , p21-Activated Kinases/antagonists & inhibitors , Aged , Aged, 80 and over , Animals , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Disulfides/therapeutic use , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/secondary , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Middle Aged , Naphthols/therapeutic use , Neoplasm Metastasis , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA, Small Interfering/metabolism
3.
Pharmacol Res ; 123: 27-39, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28648739

ABSTRACT

G protein-coupled receptor 35 (GPR35), a receptor for lysophosphatidic acid, is highly expressed in the gastrointestinal tract. Recently, GPR35 has been implicated in the onset of inflammatory bowel disease (IBD), but its role in physiological and pathological processes in the colon remains undefined. In this study, we investigated the contribution of GPR35-mediated signalling to mucosal repair of colonic epithelium in IBD. GPR35 function was examined in a wound healing model, using young adult mouse colon epithelium (YAMC) cells, and in a dextran sulphate sodium (DSS)-induced mouse model of colitis. Cell proliferation, mRNA expression, extracellular signal-regulated kinase (ERK) activation, and protein localization were determined by MTT assay, quantitative RT-PCR, western blotting, and immunohistochemistry, respectively. GPR35 agonists (YE120, zaprinast, and pamoic acid) promoted wound repair in a concentration-dependent manner independently of cell proliferation, whereas a specific GPR35 antagonist CID2745687, forskolin, and pertussis toxin reversed the YE120-induced effect. YE120 increased the mRNA expression of fibronectin and its receptor integrin α5, and ERK1/2 phosphorylation, but these responses were attenuated by CID2745687 and forskolin. Furthermore, the severity of DSS-induced colitis was significantly reduced by daily injections of pamoic acid via upregulation of fibronectin and integrin α5 in the colonic epithelium. GPR35 signalling promotes mucosal repair by inducing fibronectin and integrin α5 expression, coupling to Gi protein, and activating ERK1/2 in colonic epithelial cells. These findings define GPR35 as a candidate therapeutic target in IBD.


Subject(s)
Cell Movement/physiology , Colon/cytology , Epithelial Cells/physiology , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/agonists , Wound Healing/physiology , Animals , Cell Line , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Cytokines/genetics , Dextran Sulfate , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/metabolism , Furans/pharmacology , Integrins/metabolism , Male , Mice, Inbred C57BL , Naphthols/therapeutic use , Nitriles/pharmacology , Peroxidase/metabolism , Purinones/pharmacology , RNA, Messenger/metabolism , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism
4.
Bioorg Med Chem Lett ; 23(14): 4225-9, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23735745

ABSTRACT

Resveratrol is known to exert a cardioprotective effect against hypoxia/reoxygenation (H/R) injury. HS-1793 is a novel, more stable resveratrol analog, but its cardioprotective effects were unknown. The present study aimed to test the cardioprotective effect of HS-1793 against H/R injury and investigate the role of mitochondria in Sprague Dawley rat heart damage using an ex vivo Langendorff system. HS-1793 ameliorated H/R-induced mitochondrial dysfunction by reducing mitochondrial reactive oxygen species production, improving mitochondrial oxygen consumption and suppressing mitochondrial calcium (Ca(2+)) overload during reperfusion. Moreover, HS-1793-treated rat heart showed reduced infarct size. Our data suggest that HS-1793 can protect cardiac against mitochondrial damage following H/R, thereby suppressing injury.


Subject(s)
Naphthols/chemistry , Resorcinols/chemistry , Stilbenes/chemistry , Animals , Calcium/metabolism , Heart/physiopathology , Hypoxia , Mitochondria/drug effects , Mitochondria/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Naphthols/pharmacology , Naphthols/therapeutic use , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Resorcinols/pharmacology , Resorcinols/therapeutic use , Resveratrol
5.
Eur J Med Chem ; 249: 115125, 2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36682292

ABSTRACT

The electron transport chain (ETC) in the cell membrane consists of a series of redox complexes that transfer electrons from electron donors to acceptors and couples this electron transfer with the transfer of protons (H+) across a membrane. This process generates proton motive force which is used to produce ATP and a myriad of other functions and is essential for the long-term survival of Mycobacterium tuberculosis (Mtb), the causative organism of tuberculosis (TB), under the hypoxic conditions present within infected granulomas. Menaquinone (MK), an important carrier molecule within the mycobacterial ETC, is synthesized de novo by a cluster of enzymes known as the classic/canonical MK biosynthetic pathway. MenA (1,4-dihydroxy-2-naphthoate prenyltransferase), the antepenultimate enzyme in this pathway, is a verified target for TB therapy. In this study, we explored structure-activity relationships of a previously discovered MenA inhibitor scaffold, seeking to improve potency and drug disposition properties. Focusing our campaign upon three molecular regions, we identified two novel inhibitors with potent activity against MenA and Mtb (IC50 = 13-22 µM, GIC50 = 8-10 µM). These analogs also displayed substantially improved pharmacokinetic parameters and potent synergy with other ETC-targeting agents, achieving nearly complete sterilization of Mtb in combination therapy within two weeks in vivo. These new inhibitors of MK biosynthesis present a promising new strategy to curb the continued spread of TB.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Naphthols/metabolism , Naphthols/therapeutic use , Tuberculosis/drug therapy , Tuberculosis/microbiology , Electron Transport , Antitubercular Agents/metabolism
6.
Mol Med ; 18: 209-14, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22113495

ABSTRACT

Mitochondria play a critical role in metabolic homeostasis of a cell. Our recent studies, based on the reported interrelationship between c-Myc and Sirt1 (mammalian orthologue of yeast sir2 [silent information regulator 2]) expression and their role in mitochondrial biogenesis and function, demonstrated a significant downregulation of Sirt1 protein expression and an upregulation of c-Myc following trauma-hemorrhage (T-H). Activators of Sirt1 are known to improve mitochondrial function and the naturally occurring polyphenol resveratrol (RSV) has been shown to significantly increase Sirt1 activity by increasing its affinity to both NAD+ and the acetylated substrate. In this study we tested the salutary effect of RSV following T-H and its influence on Sirt1 expression. Rats were subjected to T-H or sham operation. RSV (8 mg/kg body weight, intravenously) or vehicle was administered 10 min after the onset of resuscitation, and the rats were killed 2 h following resuscitation. Sirtinol, a Sirt1 inhibitor, was administered 5 min prior to RSV administration. Cardiac contractility (±dP/dt) was measured and heart tissue was tested for Sirt1, Pgc-1α, c-Myc, cytosolic cytochrome C expression and ATP level. Left ventricular function, after T-H, was improved (P < 0.05) following RSV treatment, with significantly elevated expression of Sirt1 (P < 0.05) and Pgc-1α (P < 0.05), and decreased c-Myc (P < 0.05). We also observed significantly higher cardiac ATP content, declined cytosolic cytochrome C and decreased plasma tumor necrosis factor-α in the T-H-RSV group. The salutary effect due to RSV was abolished by sirtinol, indicating a Sirt1-mediated effect. We conclude that RSV may be a useful adjunct to resuscitation fluid following T-H.


Subject(s)
Hemorrhage/drug therapy , Myocardial Contraction/drug effects , Sirtuin 1/metabolism , Stilbenes/therapeutic use , Wounds and Injuries/complications , Animals , Benzamides/therapeutic use , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Male , Naphthols/therapeutic use , Rats , Rats, Sprague-Dawley , Resveratrol
7.
Int J Gynecol Cancer ; 22(3): 360-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22228428

ABSTRACT

OBJECTIVE: In our previous study, we had isolated a series of lignan compounds, termed vitexins, from the seed of Chinese herb Vitex negundo and found broad antitumor activities of these compounds in many cancer xenograft models and cell lines. This study was aimed to determine the antitumor effect of purified vitexin compound 1 (VB1) on choriocarcinoma in vitro and in vivo. MATERIALS AND METHODS: The severe combined immunodeficiency mouse model of choriocarcinoma was established to investigate the in vivo effect of VB1. Its effect on proliferation and apoptosis in JEG-3 cell line was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, colony formation assay and flow cytometry, respectively. The expression of caspase-3, Bcl-2, and some molecules involved in the mammalian target of rapamycin (mTOR) signaling was detected by Western blot. RESULTS: Vitexin compound 1 significantly inhibited the growth of choriocarcinoma in severe combined immunodeficient mice and reduced the serum ß-human chorionic gonadotropin level. Vitexin compound 1 inhibited cell proliferation, induced apoptosis, and inhibited the mTOR signaling in JEG-3 cell line. CONCLUSION: Vitexin compound 1 could inhibit choriocarcinoma via inducing cell apoptosis and suppressing the mTOR pathway.


Subject(s)
Apigenin/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Choriocarcinoma/drug therapy , Uterine Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Apigenin/chemistry , Apigenin/isolation & purification , Cell Line, Tumor , Choriocarcinoma/pathology , Down-Regulation/drug effects , Female , Guaiacol/analogs & derivatives , Guaiacol/isolation & purification , Guaiacol/therapeutic use , Humans , Mice , Mice, Nude , Mice, SCID , Models, Biological , Naphthols/isolation & purification , Naphthols/therapeutic use , Pregnancy , Tumor Burden/drug effects , Up-Regulation/drug effects , Uterine Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Bioorg Med Chem Lett ; 21(11): 3354-7, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21531136

ABSTRACT

We have identified naphthol derivatives as inhibitors of the vanilloid receptor TRPV1 by high throughput screening. The initial lead showed high clearance in rats and has been optimized by enhancing the acidity of the phenol group. Compound 6b has reduced clearance, improved potency and is active in rat cystometry models of urinary incontinence after intravenous administration.


Subject(s)
Naphthols/chemistry , TRPV Cation Channels/antagonists & inhibitors , Urinary Incontinence/drug therapy , Animals , Disease Models, Animal , Female , Inhibitory Concentration 50 , Molecular Structure , Naphthols/chemical synthesis , Naphthols/therapeutic use , Parenteral Nutrition , Rats , Structure-Activity Relationship
9.
J Infect Dis ; 201(8): 1250-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20205571

ABSTRACT

BACKGROUND: Nonsteroidal agonists have been developed that selectively bind to and activate estrogen receptor beta (ERbeta) rather than estrogen receptor alpha (ERalpha). ERbeta is expressed equally in both male and female mammals in multiple extragonadal tissues. Work reported elsewhere has demonstrated that ERbeta agonists have beneficial effects in multiple (but not all) models of inflammatory diseases and also increase survival in experimentally induced sepsis. METHODS: In these experiments, ERbeta agonists (ERB-041 or WAY-202196) were compared with vehicle control in the murine cecal ligation and puncture (CLP) model and in the pneumococcal pneumonia model of sepsis. The effect of WAY-202196 on the gene expression profile in the CLP model was further studied by transcriptome analysis of lung and small intestine tissue samples. RESULTS: ERbeta agonists provided a significant survival benefit in both experimental models of bacterial sepsis. This survival advantage was accompanied by reduced histologic evidence of tissue damage, reduced transcription of multiple proinflammatory proteins by transcriptome analysis and was not associated with increased bacterial outgrowth. CONCLUSIONS: ERbeta agonist administration provided a survival advantage in septic animals and appears to be a promising therapeutic modality in sepsis.


Subject(s)
Estrogen Receptor beta/agonists , Naphthols/therapeutic use , Oxazoles/therapeutic use , Sepsis/drug therapy , Animals , Disease Models, Animal , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/physiology , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred BALB C , Pneumonia, Pneumococcal/drug therapy , Sepsis/physiopathology , Transcription, Genetic/drug effects
10.
Biochem Pharmacol ; 192: 114688, 2021 10.
Article in English | MEDLINE | ID: mdl-34274354

ABSTRACT

Signal Transducer and Activator of Transcription (STAT) 3 emerged rapidly as a high-value target for treatment of cancer. However, small-molecule STAT3 inhibitors have been slow to enter the clinic due, in part, to serious adverse events (SAE), including lactic acidosis and peripheral neuropathy, which have been attributed to inhibition of STAT3's mitochondrial function. Our group developed TTI-101, a competitive inhibitor of STAT3 that targets the receptor pY705-peptide binding site within the Src homology 2 (SH2) domain to block its recruitment and activation. TTI-101 has shown target engagement, no toxicity, and evidence of clinical benefit in a Phase I study in patients with solid tumors. Here we report that TTI-101 did not affect mitochondrial function, nor did it cause STAT3 aggregation, chemically modify STAT3 or cause neuropathic pain. Instead, TTI-101 unexpectedly suppressed neuropathic pain induced by chemotherapy or in a spared nerve injury model. Thus, in addition to its direct anti-tumor effect, TTI-101 may be of benefit when administered to cancer patients at risk of developing chemotherapy-induced peripheral neuropathy (CIPN).


Subject(s)
Hyperalgesia/drug therapy , Naphthols/therapeutic use , Neuralgia/drug therapy , Oxidative Phosphorylation/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , Sulfonamides/therapeutic use , Touch , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Naphthols/pharmacology , Neuralgia/metabolism , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology
11.
Cancer Treat Res Commun ; 26: 100271, 2021.
Article in English | MEDLINE | ID: mdl-33341453

ABSTRACT

Worldwide prevalence of esophageal adenocarcinomas with high rates of mortality coupled with increased mutations in esophageal cells warrants investigation to understand deregulation of cell signaling pathways leading to cancer. To this end, the current study was undertaken to unravel the cell death signatures using the model human esophageal adenocarcinoma cell line-OE33. The strategy involved targeting the key epigenetic modulator SIRT1, a histone deacetylase by a small molecule inhibitor - sirtinol. Sirtinol induced a dose-dependent inhibition of cell viability under both normoxic and hypoxic conditions with long term impact on proliferation as shown by clonogenic assays. Signature apoptotic signaling pathways including caspase activation and decreased Bcl-2 were observed. Proteomic analysis highlighted an array of entities affected including molecules involved in replication, transcription, protein synthesis, cell division control, stress-related proteins, spliceosome components, protein processing and cell detoxification/degradation systems. Importantly, the stoichiometry of the fold changes of the affected proteins per se could govern the cell death phenotype by sirtinol. Sirtinol could also potentially curb resistant and recurrent tumors that reside in hypoxic environments. Overall, in addition to unraveling the cellular, molecular and proteomics basis of SIRT1 inhibition, the findings open up avenues for designing novel strategies against esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Benzamides/pharmacology , Esophageal Neoplasms/genetics , Naphthols/pharmacology , Sirtuin 1/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Apoptosis/drug effects , Apoptosis/genetics , Benzamides/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Screening Assays, Antitumor , Epigenesis, Genetic/drug effects , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Naphthols/therapeutic use , Proteomics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirtuin 1/metabolism
12.
Osteoporos Int ; 21(8): 1437-47, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19813044

ABSTRACT

SUMMARY: The main component of the metabolic by-products of fermentation by Propionibacterium freudenreichii ET-3 is 1,4-dihydroxy-2-naphthoic acid (DHNA), which has a naphthoquinone skeleton, as in vitamin K2. This study showed that DHNA improved bone mass reduction with osteoporosis model mice caused by FK506. INTRODUCTION: Growth of the intestinal bacterium Lactobacillus bifidus is specifically facilitated by DHNA. The present study used osteoporosis model mice to investigate the effects of DHNA on bone remodeling. METHODS: FK506, an immunosuppressant, was used to prepare osteoporosis model mice. Thirty mice were divided into three groups: FK group, FK+DHNA group, and control group. In the FK group, FK506 was administered to induce bone mass reduction. In the FK-DHNA group, FK506 and DHNA were administered concurrently to observe improvements in bone mass reduction. To ascertain systemic and local effects of DHNA, we investigated systemic pathological changes in colon, kidney function and cytokine dynamics, and morphological and organic changes in bone and osteoclast dynamics as assessed by culture experiments. RESULTS: Compared to the FK group without DHNA, colon damage and kidney dysfunction were milder for FK+DHNA group, and production of inflammatory cytokines (interleukin (IL)-1beta, IL-6 and tumor necrosis factor (TNF)-alpha) was more suppressed. Furthermore, compared to the group without DHNA, histological analyses and radiography showed that bone resorption was suppressed for the DHNA group. Culture experiments using osteoclasts from murine bone marrow showed osteoclast suppression for the DHNA group compared to the group without DHNA. CONCLUSION: These results show that DHNA has some effects for improving bone mass reduction caused by FK506.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Naphthols/therapeutic use , Osteoporosis/prevention & control , Animals , Body Weight/drug effects , Bone Density/drug effects , Cells, Cultured , Colon/drug effects , Colon/pathology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Eating/drug effects , Femur/drug effects , Femur/pathology , Femur/physiopathology , Immunosuppressive Agents , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/physiopathology , Male , Mice , Mice, Inbred ICR , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoporosis/chemically induced , Tacrolimus
13.
Biochem Pharmacol ; 177: 113943, 2020 07.
Article in English | MEDLINE | ID: mdl-32240651

ABSTRACT

Metastatic prostate cancer (PCa) has high mortality and a poor 5-year survival rate primarily due to the lack of effective treatments. Bone is the primary site of PCa metastasis in humans and the development of reliable therapeutic options for bone metastatic PCa will make a huge impact in reducing the mortality among these patients. Although P21 activated kinases (PAKs) have been studied in the past for their role in cancer, the efficacy of targeting PAKs to treat lung and bone metastatic PCa has not been tested yet. In the current study, we report that targeting PAK1 using IPA-3, an allosteric inhibitor of PAK1 kinase activity, significantly inhibits the murine metastatic PCa (RM1) cell proliferation and motility in vitro, and metastasis to the lungs in vivo. More importantly, we demonstrate for the first time that treatment with IPA-3 can blunt metastatic PCa-induced bone remodeling in vivo as analyzed by the 3-dimensional microcomputer tomography analysis. Our study has identified IPA-3 as a potential drug to treat bone metastatic PCa.


Subject(s)
Bone Remodeling/drug effects , Disulfides/pharmacology , Disulfides/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Naphthols/pharmacology , Naphthols/therapeutic use , Prostatic Neoplasms/pathology , p21-Activated Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Tibia/pathology
14.
Arch Virol ; 154(9): 1539-44, 2009.
Article in English | MEDLINE | ID: mdl-19685199

ABSTRACT

On the basis of the structural homologies between ST1859 (1[(2-hydroxy-1-naphtyl)methyl]-2-naphthol) and the anti-prion agents and its anti-amyloidogenic activity, we tested whether this molecule altered the biochemical properties of aggregates formed in vitro by synthetic prion peptides and affected prion infectivity in experimental scrapie. Co-incubation of ST1859 with the peptides PrP 106-126 and PrP 82-146 reduced their fibrillogenic capacity and their resistance to digestion with protease K. Hamsters inoculated with the ST1859-treated homogenate showed a significant delay in the onset of clinical signs of disease and longer survival. Survival was also significantly longer in infected hamsters treated peripherally with ST1859 for the whole post-inoculation period until the onset of clinical symptoms. Similar results were found with the analogue ST1745. Our data indicate that ST1859 reduces prion infectivity and can exert a therapeutic effect in experimental scrapie.


Subject(s)
Naphthols/therapeutic use , PrP 27-30 Protein/antagonists & inhibitors , Scrapie/drug therapy , gamma-Aminobutyric Acid/analogs & derivatives , Animals , Cricetinae , Endopeptidase K/metabolism , Injections, Intraperitoneal , Male , Mesocricetus , Naphthols/administration & dosage , Naphthols/chemistry , PrP 27-30 Protein/metabolism , gamma-Aminobutyric Acid/administration & dosage , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/therapeutic use
15.
Anesth Analg ; 108(6): 1855-61, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19448212

ABSTRACT

BACKGROUND: Hemeoxygenase-1 induction in response to adverse circulatory conditions is protective. Our recent study has shown that administration of sirtinol attenuates hepatic injury in male Sprague-Dawley rats after trauma-hemorrhage; however, the mechanism by which sirtinol produces the salutary effects remains unknown. We hypothesized that sirtinol administration in male Sprague-Dawley rats after trauma-hemorrhage decreases cytokine production and protects against lung injury through a hemeoxygenase-1 related pathway. METHODS: Male Sprague-Dawley rats (n = 8 per group) underwent trauma-hemorrhage (mean arterial blood pressure 40 mm Hg for 90 min, then resuscitation). A single dose of sirtinol (1 mg/kg of body weight) with or without a hemeoxygenase enzyme inhibitor (chromium-mesoporphyrin) or vehicle was administered IV during resuscitation. Twenty-four hours thereafter, myeloperoxidase activity (a marker of neutrophil sequestration) and tumor necrosis factor alpha, interleukin-6, and interleukin-10 levels in the lung, protein concentrations in bronchoalveolar lavage fluid and tissue histology were measured. Lung hemeoxygenase-1 protein level was also determined. RESULTS: In the sirtinol-treated rats subjected to trauma-hemorrhage, there were significant improvements in lung myeloperoxidase activity (4.68 +/- 0.31 vs 9.36 +/- 1.03 U/mg protein, P < 0.05), tumor necrosis factor alpha levels (710.7 +/- 28 vs 1288 +/- 40.69 pg/mg protein, P < 0.05), interleukin-6 levels (343.6 +/- 18.41 vs 592.7 +/- 22.3 pg/mg protein, P < 0.05), and protein concentrations (303.8 +/- 24.54 vs 569.6 +/- 34.82 microg/mL, P < 0.05) and lesser damage in histology. There was no statistically significant difference in interleukin-10 levels in the lung between sirtinol-treated trauma-hemorrhaged rats and vehicle-treated trauma-hemorrhaged rats (842.5 +/- 54.18 vs 756.2 +/- 41.34 pg/mg protein, respectively). Lung hemeoxygenase-1 protein levels were increased in rats receiving sirtinol treatment as compared with vehicle-treated trauma-hemorrhaged rats (5.18 +/- 0.25 vs 2.70 +/- 0.16, P < 0.05). Administration of the hemeoxygenase inhibitor chromium-mesoporphyrin prevented the sirtinol-induced attenuation of shock-induced lung damage. CONCLUSION: The salutary effects of sirtinol administration on attenuation of lung inflammation after trauma-hemorrhage are mediated via upregulation of hemeoxygenase-1 expression.


Subject(s)
Benzamides/therapeutic use , Heme Oxygenase-1/biosynthesis , Hemorrhage/complications , Lung Diseases/drug therapy , Naphthols/therapeutic use , Sirtuins/antagonists & inhibitors , Wounds and Injuries/complications , Animals , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Hemorrhage/pathology , Interleukin-10/biosynthesis , Interleukin-6/biosynthesis , Lung Diseases/pathology , Male , Peroxidase/metabolism , Proteins/analysis , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/drug effects , Up-Regulation/physiology , Wounds and Injuries/pathology
16.
Article in English | MEDLINE | ID: mdl-31527426

ABSTRACT

Noise is one of the most common causes of hearing loss in industrial countries. There are many studies about chemical agents to prevent noise-induced hearing loss (NIHL). However, there is no commercially available drug yet. Retinoic acid is an active metabolite of Vitamin A; it has an anti-apoptic role in NIHL. This study aims to verify the differences among selective agonists of retinoic acid receptors (RARs) in NIHL. All-trans retinoic acid (ATRA), AM80 (selective retinoic acid receptor α agonist), AC261066 (Selective retinoic acid receptor ß1 agonist), and CD1530 (Selective retinoic acid λ agonist) were injected to 6-7 weeks old CJ5BL/6 mice before noise (110 dB for 3 h) exposure. In the auditory brainstem response test pre-, post 1, 3, and 7 days after noise exposure, not only ATRA but all kinds of selective RAR agonists showed protective effects in hearing threshold and wave I amplitude. Though there was no significant difference in the level of protective effects between agonists, α agonist showed the most prominent effect in preserving hearing function as well as outer hair cells after noise exposure. In conclusion, selective agonists of RAR demonstrate comparable protective effects against NIHL to retinoic acid. Given that these selective RAR agonists have less side effects than retinoic acid, they may be promising potential drugs against NIHL.


Subject(s)
Benzoates/therapeutic use , Hearing Loss, Noise-Induced/prevention & control , Naphthols/therapeutic use , Receptors, Retinoic Acid/agonists , Tetrahydronaphthalenes/therapeutic use , Thiazoles/therapeutic use , Tretinoin/therapeutic use , Animals , Mice, Inbred C57BL , Noise/adverse effects
17.
Biochem Pharmacol ; 154: 464-473, 2018 08.
Article in English | MEDLINE | ID: mdl-29906467

ABSTRACT

Dendritic cell (DC) trafficking from lung to the draining mediastinal lymph nodes (MLNs) is a key step for initiation of T cell responses in allergic asthma. In the present study, we investigate the role of DC-mediated airway inflammation after inhibition of p21-activated kinase-1 (PAK1), an effector of Rac and Cdc42 small GTPases, in the allergen-induced mouse models of asthma. Systemic administration of PAK1 specific inhibitor IPA-3 significantly attenuates not only the airway inflammation but also the airway hyperresponsiveness in a mouse model of ovalbumin-induced asthma. Specifically, intratracheal administration of low dosage of IPA-3 consistently decreases not only the airway inflammation but also the DC trafficking from lung to the MLNs. Importantly, intratracheal instillation of IPA-3-treated and ovalbumin-pulsed DCs behaves largely the same as that of either Rac inhibitor-treated and ovalbumin-pulsed DCs or Cdc42 inhibitor-treated and ovalbumin-pulsed DCs in attenuation of the airway inflammation in ovalbumin-challenged mice. Mechanistically, PAK1 is not involved in the maturation, apoptosis, antigen uptake, and T cell activation of cultured DCs, but PAK1 dose lie on the downstream of Rac and Cdc42 to regulate the DC migration toward the chemokine C-C motif chemokine ligand 19. Taken together, this study demonstrates that inhibition of PAK1 attenuates the cardinal features of asthma through suppressing the DC trafficking from lung to the MLN, and that interfere with DC trafficking by a PAK1 inhibitor thus holds great promise for the therapeutic intervention of allergic diseases.


Subject(s)
Asthma/metabolism , Dendritic Cells/metabolism , Lymph Nodes/metabolism , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Animals , Asthma/chemically induced , Asthma/drug therapy , Bronchoconstrictor Agents/toxicity , Cells, Cultured , Dendritic Cells/drug effects , Disulfides/pharmacology , Disulfides/therapeutic use , Dose-Response Relationship, Drug , Lymph Nodes/drug effects , Mice , Mice, Inbred BALB C , Mice, Transgenic , Naphthols/pharmacology , Naphthols/therapeutic use , Ovalbumin/toxicity
18.
Sci Rep ; 7(1): 17949, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263373

ABSTRACT

MiR-34a acts as tumor suppressor microRNA (miRNA) in several cancers, including multiple myeloma (MM), by controlling the expression of target proteins involved in cell cycle, differentiation and apoptosis. Here, we have investigated the combination between miR-34a and γ-secretase inhibitor (γSI), Sirtinol or zoledronic acid (ZOL) in order to enhance the inhibitory action of this miRNA on its canonical targets such as Notch1 and SIRT1, and on Ras/MAPK-dependent pathways. Our data demonstrate that miR-34a synthetic mimics significantly enhance the anti-tumor activity of all the above-mentioned anti-cancer agents in RPMI 8226 MM cells. We found that γSI enhanced miR-34a-dependent anti-tumor effects by activating the extrinsic apoptotic pathway which could overcome the cytoprotective autophagic mechanism. Moreover, the combination between miR-34a and γSI increased the cell surface calreticulin (CRT) expression, that is well known for triggering anti-tumor immunological response. The combination between miR-34a and Sirtinol induced the activation of an intrinsic apoptotic pathway along with increased surface expression of CRT. Regarding ZOL, we found a powerful growth inhibition after enforced miR-34a expression, which was not likely attributable to neither apoptosis nor autophagy modulation. Based on our data, the combination of miR-34a with other anti-cancer agents appears a promising anti-MM strategy deserving further investigation.


Subject(s)
MicroRNAs/therapeutic use , Multiple Myeloma/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Benzamides/therapeutic use , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Naphthols/therapeutic use , Zoledronic Acid/therapeutic use
19.
Clin Pharmacol Ther ; 80(3): 216-27, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16952488

ABSTRACT

This work describes a microdosing study with an investigational, carbon 11-labeled antiamyloid drug, 1,1'-methylene-di-(2-naphthol) (ST1859), and positron emission tomography (PET) in healthy volunteers (n = 3) and patients with Alzheimer's disease (n = 6). The study aimed to assess the distribution and local tissue pharmacokinetics of the study drug in its target organ, the human brain. Before PET studies were performed in humans, the toxicologic characteristics of ST1859 were investigated by an extended single-dose toxicity study according to guidelines of the Food and Drug Administration and European Medicines Agency, which are relevant for clinical trials with a single microdose. After intravenous bolus injection of 341 +/- 21 MBq [(11)C]ST1859 (containing <11.4 nmol of unlabeled ST1859), peripheral metabolism was rapid, with less than 20% of total plasma radioactivity being in the form of unchanged parent drug at 10 minutes after administration. In both the control and patient groups, uptake of radioactivity into the brain was relatively fast (time to reach maximum concentration, 9-17 minutes) and pronounced (maximum concentration [standardized uptake value], 1.3-2.2). In both healthy volunteers and patients, there was a rather uniform distribution of radioactivity in the brain, including both amyloid-beta-rich and -poor regions, with slow washout of radioactivity (half-life, 82-185 minutes). In conclusion, these data provide important information on the blood-brain barrier penetration and metabolism of an investigational antiamyloid drug and suggest that the PET microdosing approach is a useful method to describe the target-organ pharmacokinetics of radiolabeled drugs in humans.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid/antagonists & inhibitors , Naphthols/pharmacokinetics , Positron-Emission Tomography/methods , Adult , Aged , Alzheimer Disease/metabolism , Amyloid/metabolism , Area Under Curve , Biological Availability , Blood-Brain Barrier/metabolism , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Female , Humans , Injections, Intravenous , Male , Molecular Structure , Naphthols/blood , Naphthols/therapeutic use , Neuroprotective Agents/blood , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Pilot Projects , Time Factors , Tissue Distribution
20.
Spine (Phila Pa 1976) ; 41(11): 919-925, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26863260

ABSTRACT

STUDY DESIGN: Ninety-six male adult CD-1 mice were randomly divided into sham, spinal cord injury (SCI) + vehicle, and SCI + IPA-3 groups. Expression of matrix metalloproteinase (MMP)-2 and MMP-9, production of tumor necrosis factors (TNF)-α and interleukin (IL)-1ß, tissue edema, blood-spinal cord barrier penetrability, neural cell apoptosis, and neurological function recovery were measured. OBJECTIVE: The aim of the study was to evaluate the effect of specific inhibition of p21-activated kinase 1 (PAK1) by IPA-3 on SCI and the underlying mechanisms thereof. SUMMARY OF BACKGROUND DATA: SCI is a devastating clinical condition that may result in long-lasting and deteriorating functional deficits. The major goal of SCI treatment is to limit the development of secondary injury. IPA-3, a PAK1 inhibitor, exhibited neuroprotection against secondary damage after traumatic brain injury and subarachnoid hemorrhage (SAH). METHODS: MMP-2, MMP-9, and cleaved caspase-3 expression were assessed by Western blot. Inflammatory cytokines TNF-α and IL-1ß were detected by enzyme-linked immunosorbent assay (ELISA). The blood-spinal cord barrier disruption was measured by water content and Evans blue extravasation of the spinal cord. Neuronal apoptosis was evaluated by Nissl staining and Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling (TUNEL) assay. The locomotor behavior of hind limb was evaluated by Basso Mouse Scale (BMS) at 1, 3, 7, 14, and 28 days post-injury. RESULTS: Compared with SCI + vehicle mice, IPA-3 treatment showed decreased p-PAK1, MMP-2, MMP-9, cleaved caspase-3, TNF-α, and IL-1ß expression. Moreover, inhibition of PAK1 by IPA-3 reduced spinal cord water content and Evans blue extravasation, increased neuronal survival, and reduced TUNEL-positive cells at 24 hours after SCI. Furthermore, IPA-3 improved spinal cord functional recovery 7 days after SCI. CONCLUSION: Inhibition of PAK1 by IPA-3 promoted recovery of neurological function, possibly by downregulating the expression of MMP-2, MMP-9, TNF-α, and IL-1ß. Our data suggest that PAK1 may be a potential therapeutic target in patients with SCI. LEVEL OF EVIDENCE: 1.


Subject(s)
Disulfides/therapeutic use , Motor Activity/drug effects , Naphthols/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , p21-Activated Kinases/antagonists & inhibitors , Animals , Disulfides/pharmacology , Male , Mice , Motor Activity/physiology , Naphthols/pharmacology , Protein Kinase Inhibitors/pharmacology , Random Allocation , Recovery of Function/physiology , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/physiopathology , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL