Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Life Sci ; 282: 119762, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34186047

ABSTRACT

AIMS: Radiofrequency ablation (RFA) is the first-line option for early-stage hepatocellular carcinoma (HCC). However, the residual tumor attributed to insufficient RFA (iRFA) led to tumor recurrence and metastasis. Novel combination strategies are urgently needed to enhance efficiency of RFA. MAIN METHODS: For in vitro iRFA models, HCC cells were placed in a water bath at 46 °C for 10 min and then returned to the original incubator. For in vivo models, HCC cells were implanted subcutaneously into nude mice. The nude mice were then randomly assigned into 4 groups: control group, XL888 group, iRFA group, combination of XL888 and iRFA group. CCK8 was performed to detect cell viability; Hoechst 33258 was used to explore nuclear morphology; The expression levels of proteins were demonstrated by western blotting; Co-localization of HSP90 and STAT3 was elucidated by immunofluorescence confocal microscopy; Immunohistochemistry was used to explore expression levels of proteins at tissue level. KEY FINDINGS: XL888 promoted apoptosis of HCC cells induced by heat via inhibiting expression levels of Mcl-1 and cleaved-caspase 3 in vivo and in vitro. XL888 attenuated the complex formation of HSP90 and STAT3, leading to decreased expression levels of STAT3 and p-STAT3. In human HCC tissues, IHC scores of HSP90 were positively correlated with those of STAT3. Overexpression of STAT3 rescued cell apoptosis induced by co-treatment of XL888 and heat. SIGNIFICANCE: We implied that XL888 promoted apoptosis of HCC cells induced by heat via disrupting the binding of HSP90 and STAT3, providing theoretical basis for a novel combination strategy for HCC.


Subject(s)
Apoptosis/drug effects , Azabicyclo Compounds/pharmacology , Carcinoma, Hepatocellular/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Liver Neoplasms/drug therapy , Phthalic Acids/pharmacology , STAT3 Transcription Factor/genetics , Animals , Azabicyclo Compounds/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Down-Regulation/drug effects , HSP90 Heat-Shock Proteins/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Mice, Inbred BALB C , Mice, Nude , Phthalic Acids/therapeutic use , Radiofrequency Ablation
2.
Cell Death Dis ; 12(5): 483, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986262

ABSTRACT

Osteoarthritis (OA) is a common articular degenerative disease characterized by loss of cartilage matrix and subchondral bone sclerosis. Kartogenin (KGN) has been reported to improve chondrogenic differentiation of mesenchymal stem cells. However, the therapeutic effect of KGN on OA-induced cartilage degeneration was still unclear. This study aimed to explore the protective effects and underlying mechanisms of KGN on articular cartilage degradation using mice with post-traumatic OA. To mimic the in vivo arthritic environment, in vitro cultured chondrocytes were exposed to interleukin-1ß (IL-1ß). We found that KGN barely affected the cell proliferation of chondrocytes; however, KGN significantly enhanced the synthesis of cartilage matrix components such as type II collagen and aggrecan in a dose-dependent manner. Meanwhile, KGN markedly suppressed the expression of matrix degradation enzymes such as MMP13 and ADAMTS5. In vivo experiments showed that intra-articular administration of KGN ameliorated cartilage degeneration and inhibited subchondral bone sclerosis in an experimental OA mouse model. Molecular biology experiments revealed that KGN modulated intracellular reactive oxygen species in IL-1ß-stimulated chondrocytes by up-regulating nuclear factor erythroid 2-related factor 2 (NRF2), while barely affecting its mRNA expression. Microarray analysis further revealed that IL-1ß significantly up-regulated miR-146a that played a critical role in regulating the protein levels of NRF2. KGN treatment showed a strong inhibitory effect on the expression of miR-146a in IL-1ß-stimulated chondrocytes. Over-expression of miR-146a abolished the anti-arthritic effects of KGN not only by down-regulating the protein levels of NRF2 but also by up-regulating the expression of matrix degradation enzymes. Our findings demonstrate, for the first time, that KGN exerts anti-arthritic effects via activation of the miR-146a-NRF2 axis and KGN is a promising heterocyclic molecule to prevent OA-induced cartilage degeneration.


Subject(s)
Anilides/therapeutic use , MicroRNAs/metabolism , Osteoarthritis/drug therapy , Phthalic Acids/therapeutic use , Anilides/pharmacology , Animals , Cell Differentiation , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Phthalic Acids/pharmacology
3.
J Mater Chem B ; 8(38): 8884-8893, 2020 10 07.
Article in English | MEDLINE | ID: mdl-33026410

ABSTRACT

Cartilage injury is very common and results in considerable pain and osteoarthritis. Owing to its low self-renewal capability, cartilage regeneration is still a great challenge for clinicians. Stem cell therapy has been treated as the most promising treatment for cartilage regeneration in recent decades. However, increasing concerns about the potential biosafety of stem cell products such as immune rejection and neoplastic transformation restrict their further application in clinic. Herein, biomimetic stem cell membrane-disguised nanovehicles without biosafety risks are designed and prepared for cartilage regeneration. In this study, based on the disguise of the natural bone marrow mesenchymal stem cell (BMSC) membrane, Kartogenin (KGN) as a drug for cartilage regeneration was encapsulated into Fe3O4 nanoparticles as the core of biomimetic stem cell nanovehicles. In the core-shell structure of biomimetic stem cell nanovehicles, the fabricated KGN-loaded BMSC membrane-disguised Fe3O4 nanoparticles (KGN-MNPs) showed a stable hybrid structure with a uniform size and morphology in the physiological environments. Moreover, the prepared KGN-MNPs exhibited excellent biocompatibility when disguised with the natural membrane of BMSCs and good biosafety by eliminating the nuclei of BMSCs. In a cartilage defect rat model, compared with pure KGN, the intra-articularly injected KGN-MNPs were capable of regenerating an integrated organized structure with a layer of hyaline-like cartilage in a shorter time due to the retained natural activities of the BMSC membrane. In a word, KGN-MNPs as one kind of our designed biomimetic stem cell nanovehicles enable rapid and high quality cartilage regeneration, and provide a novel and standardized strategy for stem cell therapy in the future.


Subject(s)
Anilides/therapeutic use , Cartilage/metabolism , Cell Membrane/chemistry , Drug Carriers/chemistry , Magnetite Nanoparticles/chemistry , Phthalic Acids/therapeutic use , Regeneration/drug effects , Animals , Cartilage/physiology , Chondrocytes/drug effects , Chondrogenesis/drug effects , Drug Carriers/toxicity , Knee Joint/metabolism , Magnetite Nanoparticles/toxicity , Male , Mesenchymal Stem Cells/chemistry , Osteoblasts/drug effects , Rats, Sprague-Dawley
4.
Curr Med Sci ; 39(1): 16-20, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30868486

ABSTRACT

Regenerative medicine refers to the possibility of replacing aged/damaged cells with genetically similar young and functional cells to restore or establish normal function. Kartogenin (KGN), a small heterocyclic, drug-like compound was discovered in 2012, which is strongly associated with regenerative medicine. KGN has been applied in many regenerative fields, including cartilage regeneration and protection, tendon-bone healing, wound healing, and limb development. KGN could facilitate cartilage repair, promote formation of cartilage-like transition zone in tendon-bone junctions, stimulate collagen synthesis for wound healing, and regulate limb development in a coordinated manner. Considering the related mechanism, filamin A/CBFß/RUNX1, Ihh, and TGFß/Smad pathways have been reported to involve KGN. Therefore, KGN is proven a promising agent in regenerative medicine; however, studies conducted on the effect of KGN are limited to date and not convictive for long-term use. Further studies are recommended to explore the long-term effect and potential molecular mechanisms of KGN. Our investigations may motivate researchers to expand its applications in different forms and fields.


Subject(s)
Anilides/pharmacology , Anilides/therapeutic use , Phthalic Acids/pharmacology , Phthalic Acids/therapeutic use , Regenerative Medicine/methods , Signal Transduction , Animals , Cartilage/cytology , Cartilage/drug effects , Collagen/metabolism , Extremities/growth & development , Humans , Osteogenesis/drug effects , Tendon Injuries/drug therapy , Tendon Injuries/metabolism , Tissue Engineering , Wound Healing/drug effects
5.
Oxid Med Cell Longev ; 2018: 1861984, 2018.
Article in English | MEDLINE | ID: mdl-29887939

ABSTRACT

OBJECTIVE: This study evaluated the effects of di-(2-ethylhexyl) phthalate (DEHP) and obesity on male reproductive organ function in male mice and the potential mechanism of male secondary hypogonadism (SH) in such mice. METHODS: 140 mice were assigned to six groups for 12 weeks: normal, DEHP, DIO, DIO + DEHP low, DIO + DEHP middle, and DIO + DEHP high. The effects of DEHP and obesity upon the reproductive organs were determined by measuring sperm count and motility, relative testis and epididymis weight, hormone level, and pathological changes. Oxidative stress was evaluated by determining malondialdehyde, T-AOC, SOD, GSH, H2O2, CAT, and GSH-PX in testicular tissues. Nrf2 and Keap1 protein were measured by Western blotting. RESULTS: DEHP and obesity reduced sperm count and motility, relative testis and epididymis weight, and testosterone level but increased the levels of MDA, H2O2, leptin, and estradiol. Pathological injury was observed in the testicular Leydig cells. Moreover, the activity of CAT, SOD, and GSH-Px enzymes was inhibited. Nrf2 protein expression was reduced but that of Keap1 was increased. CONCLUSIONS: DEHP and obesity jointly caused damage to male productive function. Oxidative stress in testicular tissue, and a high level of leptin, may provide some evidence to clarify the mechanisms of male SH with DEHP and obesity.


Subject(s)
Genitalia, Male/pathology , Obesity/pathology , Phthalic Acids/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL , Phthalic Acids/pharmacology
6.
Environ Health ; 17(1): 15, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29444690

ABSTRACT

BACKGROUND: Chronic mercury intoxication is a severe health issue and occurs especially in gold mining communities. Common chelators used for improving mercury elimination are not everywhere available and challenged by poor cell wall penetration. This study is part of a feasibility trial and the aim was to gather first information about the efficacy of the newly developed chelator N,N'bis-(2-mercaptoethyl) isophthalamide (NBMI) on chronic mercury intoxication. METHODS: In this three-armed, placebo-controlled randomized trial, 36 miners with mercury urine levels exceeding 15 µg/l were administered 100 mg NBMI, 300 mg NBMI or placebo for 14 days. Levels of mercury in urine [µg/l and µg/g creatinine] and plasma l were analyzed. Therapeutic effect was assessed using the medical intoxication score (MIS) and its single health outcomes (e.g. excessive salivation, sleeping problems), fatigue scores, a neuromotoric test battery (CATSYS) and a neurological outcome (Finger to nose test). RESULTS: Physical fatigue was significantly decreased in the 300 mg NBMI group compared to the control. Mercury concentration in urine following 300 mg NBMI treatment was significantly lowered compared to control, however, this effect was less distinct with adjustment for creatinine. CONCLUSION: NBMI showed an effect on physical fatigue and there were indications to positive effects on other symptoms as well. More comprehensive studies are mandatory to verify the effects of NBMI as a novel tool for treating mercury intoxications. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02486289 . Date of registration: June 24, 2015.


Subject(s)
Chelating Agents/therapeutic use , Cysteamine/analogs & derivatives , Environmental Pollutants/urine , Mercury Poisoning/drug therapy , Mercury/urine , Occupational Exposure , Phthalic Acids/therapeutic use , Adult , Cysteamine/therapeutic use , Dose-Response Relationship, Drug , Gold , Hispanic or Latino , Humans , Male , Middle Aged , Mining , Young Adult
7.
Small ; 14(8)2018 02.
Article in English | MEDLINE | ID: mdl-29327460

ABSTRACT

An efficient treatment for osteoarthritis (OA) can benefit from the local release of a high therapeutic dose over an extended period of time. Such a treatment will minimize systemic side effects and avoid the inconvenience of frequent injections. To this aim, nanocrystal-polymer particles (NPPs) are developed by combining the advantages of nanotechnology and microparticles. Nanocrystals are produced by wet milling kartogenin (KGN), which is known to promote chondrogenesis and to foster chondroprotection. A fluorescent biodegradable polymer is synthesized for intravital particle tracking. Polymer microparticles with 320 nm embedded KGN nanocrystals (KGN-NPPs) show a high drug loading of 31.5% (w/w) and an extended drug release of 62% over 3 months. In vitro, these particles do not alter mitochondrial activity in cultured human OA synoviocytes. In vivo, KGN-NPPs demonstrate higher bioactivity than a KGN solution in a murine mechanistic OA model based on histological assessment (Osteoarthritis Research Society International score), epiphyseal thickness (microcomputed tomography), OA biomarkers (e.g., vascular endothelial growth factor, Adamts5), and prolonged intra-articular persistence (fluorescence analysis). This work provides proof-of-concept of a novel and innovative extended drug delivery system with the potential to treat human OA.


Subject(s)
Anilides/therapeutic use , Nanoparticles/chemistry , Osteoarthritis/drug therapy , Phthalic Acids/therapeutic use , Polymers/chemistry , Anilides/chemistry , Animals , Cells, Cultured , Chondrogenesis/drug effects , Drug Delivery Systems , Humans , Injections, Intra-Articular , Mice , Nanotechnology/methods , Phthalic Acids/chemistry
8.
Toxicon ; 141: 1-8, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29097245

ABSTRACT

Microcystin-LR (MC-LR) can cause serious injuries upon short- and long-term exposures that can be prevented by LASSBio-596 (LB-596), an anti-inflammatory compound. We aimed to test LB-596 following subchronic exposure to MC-LR. Swiss mice received 10 intraperitoneal injections of distilled water (DW) or MC-LR (20 µg/kg bw) every 2 days. On the 10th injection animals receiving DW were gavaged with DW or 50 mg/kg bw of LB-596 for 1 or 7 days (C1D, C7D, CL1D and CL7D groups), whereas those exposed to MC-LR received either DW or 50 mg/kg of LB-596 for 1 or 7 days (T1D, T7D, TL1D and TL7D groups). Twelve hours after the last gavage we assessed respiratory mechanics, and extracted lung and liver for histology, apoptosis, inflammatory biomarkers and MC-LR content. C1D, C7D, CL1D and CL7D were all similar. Mechanical parameters were significantly higher in T1D and T7D compared to the other groups. LB-596 reversed these changes on day 1 of administration. LB-596 reduced inflammatory mediators in lung and liver on day 1 of treatment. On day 7 apoptosis in liver and lung fell even more. Briefly, 7-day administration completely reversed lung and liver changes.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Liver/pathology , Lung/pathology , Microcystins/antagonists & inhibitors , Phthalic Acids/administration & dosage , Sulfonamides/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents/therapeutic use , Apoptosis/drug effects , Inflammation , Liver/drug effects , Lung/drug effects , Male , Marine Toxins , Mice , Microcystins/analysis , Microcystins/toxicity , Phthalic Acids/therapeutic use , Respiratory Mechanics/drug effects , Sulfonamides/therapeutic use , Time Factors
9.
Expert Opin Investig Drugs ; 26(12): 1403-1408, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29068253

ABSTRACT

INTRODUCTION: Local adverse effects of steroid use and the burning sensation of calcineurin inhibitors impair patients' adherence to treatment and decrease the treatment response in atopic dermatitis (AD). Steroid phobia appears to be a psychological problem in patients with AD. Topical non-steroidal remedies are in demand. Areas covered: This manuscript reviews the current literature on preclinical and clinical studies regarding topical E6005/RVT-501, a novel phosphodiesterase 4 inhibitor. We also discuss the mechanistic background of E6005/RVT-501 in the treatment of AD. Expert opinion: Topical E6005/RVT-501 improves skin eruption and pruritus of pediatric and adult AD patients without any serious side effects. It is useful for mild to moderate lesions of AD in pediatric and adult patients. Topical E6005/RVT-501 is non-steroidal agent but its potency is equal to that of mild rank topical steroid, therefore, it may fit the demand of patients with steroid phobia. Its steroid-sparing effects may also be investigated in future clinical trials and may minimize the dose and frequency of topical steroids.


Subject(s)
Dermatitis, Atopic/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Phthalic Acids/therapeutic use , Quinazolines/therapeutic use , Administration, Topical , Adult , Animals , Antipruritics/administration & dosage , Antipruritics/pharmacology , Antipruritics/therapeutic use , Child , Dermatitis, Atopic/pathology , Humans , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 4 Inhibitors/pharmacology , Phthalic Acids/adverse effects , Phthalic Acids/pharmacology , Pruritus/drug therapy , Quinazolines/adverse effects , Quinazolines/pharmacology
10.
Semin Cutan Med Surg ; 36(3): 124-130, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28895959

ABSTRACT

Crisaborole and dupilumab represent the first 2 Food and Drug Administration (FDA)-approved therapies for atopic dermatitis (AD) in more than 15 years, and there are many promising drugs currently in development. This new wave of therapeutics capitalizes on the large body of work clarifying the pathogenesis of AD over the last several decades. In particular, type 2 cytokine-driven inflammation and skin barrier dysfunction are key processes underlying AD pathogenesis.


Subject(s)
Dermatitis, Atopic/drug therapy , Dermatologic Agents/therapeutic use , Anisoles/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antipruritics/therapeutic use , Boron Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Dermatitis, Atopic/prevention & control , Emollients/therapeutic use , Humans , Infant , Infant Formula , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Nitriles/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use , Phthalic Acids/therapeutic use , Probiotics/therapeutic use , Quinazolines/therapeutic use
11.
Nature ; 546(7659): 528-532, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28614296

ABSTRACT

Urinary tract infections (UTIs) caused by uropathogenic Escherichia coli (UPEC) affect 150 million people annually. Despite effective antibiotic therapy, 30-50% of patients experience recurrent UTIs. In addition, the growing prevalence of UPEC that are resistant to last-line antibiotic treatments, and more recently to carbapenems and colistin, make UTI a prime example of the antibiotic-resistance crisis and emphasize the need for new approaches to treat and prevent bacterial infections. UPEC strains establish reservoirs in the gut from which they are shed in the faeces, and can colonize the periurethral area or vagina and subsequently ascend through the urethra to the urinary tract, where they cause UTIs. UPEC isolates encode up to 16 distinct chaperone-usher pathway pili, and each pilus type may enable colonization of a habitat in the host or environment. For example, the type 1 pilus adhesin FimH binds mannose on the bladder surface, and mediates colonization of the bladder. However, little is known about the mechanisms underlying UPEC persistence in the gut. Here, using a mouse model, we show that F17-like and type 1 pili promote intestinal colonization and show distinct binding to epithelial cells distributed along colonic crypts. Phylogenomic and structural analyses reveal that F17-like pili are closely related to pilus types carried by intestinal pathogens, but are restricted to extra-intestinal pathogenic E. coli. Moreover, we show that targeting FimH with M4284, a high-affinity inhibitory mannoside, reduces intestinal colonization of genetically diverse UPEC isolates, while simultaneously treating UTI, without notably disrupting the structural configuration of the gut microbiota. By selectively depleting intestinal UPEC reservoirs, mannosides could markedly reduce the rate of UTIs and recurrent UTIs.


Subject(s)
Fimbriae Proteins/antagonists & inhibitors , Intestines/drug effects , Intestines/microbiology , Mannosides/pharmacology , Phthalic Acids/pharmacology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/isolation & purification , Adhesins, Escherichia coli/metabolism , Amino Acid Sequence , Animals , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Feces/microbiology , Female , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/classification , Fimbriae, Bacterial/drug effects , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Humans , Intestines/cytology , Mannosides/therapeutic use , Mice , Models, Molecular , Phthalic Acids/therapeutic use , Urinary Bladder/drug effects , Urinary Bladder/microbiology , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/classification , Uropathogenic Escherichia coli/genetics
12.
Molecules ; 22(4)2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28362351

ABSTRACT

This study evaluated the biological activity of an ether extract and barbatic acid (BAR) from Cladia aggregata on embryos and adult mollusks of Biomphalaria glabrata, cercariae of Schistosoma mansoni and the microcrustacean Artemia salina. The ether extract and BAR were obtained by successive extractions with diethyl ether. The obtained extracts were analyzed using thin-layer chromatography (TLC), high-performance liquid chromatography (HPLC), proton nuclear magnetic resonance (¹H-NMR) and infrared (IR) spectroscopy. The results demonstrated that the ether extract exerted embryotoxic effects at 50 and 100 µg/mL and molluscicidal effects at 20 and 25 µg/mL. BAR exhibited no embryotoxicity, and its molluscicidal concentration was equal to that of the ether extract. However, after 60 min of exposure, 1 µg/mL BAR presented cercaricidal activity against the parasite S. mansoni at the second larval stage. Neither substance induced toxicity against A. salina. These results indicate the potential molluscicidal activities of the ether extract and BAR against B. glabrata and S. mansoni cercariae. In addition to these effects, there was a lack of toxicity against the aquatic environment and no damage to the biota, indicating the potential of these products for large-scale control and/or eradication of schistosomiasis.


Subject(s)
Biomphalaria/drug effects , Phthalic Acids/pharmacology , Phthalic Acids/therapeutic use , Schistosomiasis/drug therapy , Animals , Artemia/drug effects , Embryo, Nonmammalian/drug effects , Ether , Molluscacides/chemistry , Molluscacides/pharmacology , Molluscacides/therapeutic use , Phthalic Acids/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Extracts/toxicity , Schistosoma mansoni/drug effects , Schistosomiasis/parasitology , Toxicity Tests
13.
Article in English | MEDLINE | ID: mdl-28410666

ABSTRACT

Although the effectiveness of CysLT1 receptor antagonists on asthma has been clinically established, the effects of CysLT2 receptor antagonists are still unclear. The purpose of this study was to develop a new CysLT1 and CysLT2 receptors-mediated anaphylaxis guinea pig model using S-hexyl GSH, a γ-glutamyl transpeptidase (GTP) inhibitor, to suppress conversion of LTC4 to LTD4. Actively sensitized guinea pigs were challenged with OVA in the absence or presence of S-hexyl GSH, and survival rate following anaphylactic response was monitored. OVA-induced fatal anaphylaxis in the absence of S-hexyl GSH was almost completely inhibited by montelukast, a CysLT1 receptor antagonist, but not by the CysLT2 receptor antagonist BayCysLT2RA. However, under treatment with S-hexyl-GSH, the inhibitory effect of motelukast was dramatically diminished, whereas that of BayCysLT2RA was markedly increased. The dual CysLT1/2 receptor antagonist ONO-6950 effectively inhibited anaphylactic response in both S-hexyl GSH-treated and non-treated animals. LC/MS/MS analysis revealed that S-hexyl GSH treatment actually inhibited LTC4 metabolism in the blood and lung tissues. Using S-hexyl GSH, we developed a novel CysLT1 and CysLT2 receptors-mediated anaphylaxis guinea pig model that can be useful for not only screening both CysLT2 and CysLT1/2 receptors antagonists, but also for functional analysis of CysLT2 receptors.


Subject(s)
Anaphylaxis/drug therapy , Butyrates/administration & dosage , Cyclohexanecarboxylic Acids/administration & dosage , Indoles/administration & dosage , Leukotriene Antagonists/administration & dosage , Phthalic Acids/administration & dosage , Receptors, Leukotriene/metabolism , Anaphylaxis/chemically induced , Anaphylaxis/metabolism , Animals , Butyrates/therapeutic use , Cyclohexanecarboxylic Acids/therapeutic use , Disease Models, Animal , Glutathione/adverse effects , Glutathione/analogs & derivatives , Guinea Pigs , Indoles/therapeutic use , Leukotriene Antagonists/therapeutic use , Leukotriene C4/blood , Leukotriene C4/metabolism , Leukotriene D4/blood , Leukotriene D4/metabolism , Male , Ovalbumin/adverse effects , Phthalic Acids/therapeutic use , Survival Analysis
14.
Genome Med ; 9(1): 15, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28187790

ABSTRACT

BACKGROUND: Retinoid therapy is widely employed in clinical oncology to differentiate malignant cells into their more benign counterparts. However, certain high-risk cohorts, such as patients with MYCN-amplified neuroblastoma, are innately resistant to retinoid therapy. Therefore, we employed a precision medicine approach to globally profile the retinoid signalling response and to determine how an excess of cellular MYCN antagonises these signalling events to prevent differentiation and confer resistance. METHODS: We applied RNA sequencing (RNA-seq) and interaction proteomics coupled with network-based systems level analysis to identify targetable vulnerabilities of MYCN-mediated retinoid resistance. We altered MYCN expression levels in a MYCN-inducible neuroblastoma cell line to facilitate or block retinoic acid (RA)-mediated neuronal differentiation. The relevance of differentially expressed genes and transcriptional regulators for neuroblastoma outcome were then confirmed using existing patient microarray datasets. RESULTS: We determined the signalling networks through which RA mediates neuroblastoma differentiation and the inhibitory perturbations to these networks upon MYCN overexpression. We revealed opposing regulation of RA and MYCN on a number of differentiation-relevant genes, including LMO4, CYP26A1, ASCL1, RET, FZD7 and DKK1. Furthermore, we revealed a broad network of transcriptional regulators involved in regulating retinoid responsiveness, such as Neurotrophin, PI3K, Wnt and MAPK, and epigenetic signalling. Of these regulators, we functionally confirmed that MYCN-driven inhibition of transforming growth factor beta (TGF-ß) signalling is a vulnerable node of the MYCN network and that multiple levels of cross-talk exist between MYCN and TGF-ß. Co-targeting of the retinoic acid and TGF-ß pathways, through RA and kartogenin (KGN; a TGF-ß signalling activating small molecule) combination treatment, induced the loss of viability of MYCN-amplified retinoid-resistant neuroblastoma cells. CONCLUSIONS: Our approach provides a powerful precision oncology tool for identifying the driving signalling networks for malignancies not primarily driven by somatic mutations, such as paediatric cancers. By applying global omics approaches to the signalling networks regulating neuroblastoma differentiation and stemness, we have determined the pathways involved in the MYCN-mediated retinoid resistance, with TGF-ß signalling being a key regulator. These findings revealed a number of combination treatments likely to improve clinical response to retinoid therapy, including co-treatment with retinoids and KGN, which may prove valuable in the treatment of high-risk MYCN-amplified neuroblastoma.


Subject(s)
Anilides/therapeutic use , N-Myc Proto-Oncogene Protein/drug effects , Neuroblastoma/drug therapy , Phthalic Acids/therapeutic use , Signal Transduction , Transforming Growth Factor beta/drug effects , Tretinoin/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/genetics , Neuroblastoma/metabolism , Precision Medicine , Retinoids/therapeutic use
15.
ACS Appl Mater Interfaces ; 8(8): 5148-59, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26844837

ABSTRACT

Recently, cartilage tissue engineering (CTE) attracts increasing attention in cartilage defect repair. In this work, kartogenin (KGN), an emerging chondroinductive nonprotein small molecule, was incorporated into a thermogel of poly(L-lactide-co-glycolide)-poly(ethylene glycol)-poly(L-lactide-co-glycolide) (PLGA-PEG-PLGA) to fabricate an appropriate microenvironment of bone marrow mesenchymal stem cells (BMSCs) for effective cartilage regeneration. More integrative and smoother repaired articular surface, more abundant characteristic glycosaminoglycans (GAGs) and collagen II (COL II), and less degeneration of normal cartilage were obtained in the KGN and BMSCs coloaded thermogel group in vivo. In conclusion, the KGN-loaded PLGA-PEG-PLGA thermogel can be utilized as an alternative support for BMSCs to regenerate damaged cartilage in vivo.


Subject(s)
Anilides/therapeutic use , Cartilage/growth & development , Mesenchymal Stem Cell Transplantation , Phthalic Acids/therapeutic use , Regeneration/drug effects , Tissue Engineering , Anilides/chemistry , Animals , Bone Marrow Cells/drug effects , Cartilage/drug effects , Cartilage/pathology , Cellular Microenvironment/drug effects , Collagen/metabolism , Fibrin/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Phthalic Acids/chemistry , Polyesters/chemistry , Polyesters/therapeutic use , Polyethylene Glycols/chemistry , Polyethylene Glycols/therapeutic use , Rabbits , Rats
16.
Cell Physiol Biochem ; 38(2): 821-35, 2016.
Article in English | MEDLINE | ID: mdl-26905925

ABSTRACT

BACKGROUND/AIMS: Exogenous surfactant has been proposed as adjunctive therapy for acute respiratory distress syndrome (ARDS), but it is inactivated by different factors present in the alveolar space. We hypothesized that co-administration of LASSBio596, a molecule with significant anti-inflammatory properties, and exogenous surfactant could reduce lung inflammation, thus enabling the surfactant to reduce edema and improve lung function, in experimental ARDS. METHODS: ARDS was induced by cecal ligation and puncture surgery in BALB/c mice. A sham-operated group was used as control (CTRL). After surgery (6 hours), CTRL and ARDS animals were assigned to receive: (1) sterile saline solution; (2) LASSBio596; (3) exogenous surfactant or (4) LASSBio596 plus exogenous surfactant (n = 22/group). RESULTS: Regardless of exogenous surfactant administration, LASSBio596 improved survival rate and reduced collagen fiber content, total number of cells and neutrophils in PLF and blood, cell apoptosis, protein content in BALF, and urea and creatinine levels. LASSBio596 plus surfactant yielded all of the aforementioned beneficial effects, as well as increased BALF lipid content and reduced surface tension. CONCLUSION: LASSBio596 exhibited major anti-inflammatory and anti-fibrogenic effects in experimental sepsis-induced ARDS. Its association with surfactant may provide further advantages, potentially by reducing surface tension.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Biological Products/therapeutic use , Lung/drug effects , Phthalic Acids/therapeutic use , Pulmonary Surfactants/therapeutic use , Respiratory Distress Syndrome/drug therapy , Sulfonamides/therapeutic use , Animals , Apoptosis/drug effects , Lung/immunology , Lung/pathology , Male , Mice, Inbred BALB C , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Surface Tension/drug effects
17.
J Orthop Res ; 34(10): 1780-1789, 2016 10.
Article in English | MEDLINE | ID: mdl-26895619

ABSTRACT

Osteoarthritis (OA) is a major degenerative joint disease characterized by progressive loss of articular cartilage, synovitis, subchondral bone changes, and osteophyte formation. Currently there is no treatment for OA except temporary pain relief and end-stage joint replacement surgery. We performed a pilot study to determine the effect of kartogenin (KGN, a small molecule) on both cartilage and subchondral bone in a rat model of OA using multimodal imaging techniques. OA was induced in rats (OA and KGN treatment group) by anterior cruciate ligament transection (ACLT) surgery in the right knee joint. Sham surgery was performed on the right knee joint of control group rats. KGN group rats received weekly intra-articular injection of 125 µM KGN 1 week after surgery until week 12. All rats underwent in vivo magnetic resonance imaging (MRI) at 3, 6, and 12 weeks after surgery. Quantitative MR relaxation measures (T1ρ and T2 ) were determined to evaluate changes in articular cartilage. Cartilage and bone turnover markers (COMP and CTX-I) were determined at baseline, 3, 6, and 12 weeks. Animals were sacrificed at week 12 and the knee joints were removed for micro-computed tomography (micro-CT) and histology. KGN treatment significantly lowered the T1ρ and T2 relaxation times indicating decreased cartilage degradation. KGN treatment significantly decreased COMP and CTX-I levels indicating decreased cartilage and bone turnover rate. KGN treatment also prevented subchondral bone changes in the ACLT rat model of OA. Thus, kartogenin is a potential drug to prevent joint deterioration in post-traumatic OA. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1780-1789, 2016.


Subject(s)
Anilides/therapeutic use , Cartilage, Articular/drug effects , Osteoarthritis/drug therapy , Phthalic Acids/therapeutic use , Anilides/pharmacology , Animals , Biomarkers/blood , Disease Models, Animal , Drug Evaluation, Preclinical , Magnetic Resonance Imaging , Male , Osteoarthritis/blood , Osteoarthritis/diagnostic imaging , Phthalic Acids/pharmacology , Pilot Projects , Rats, Sprague-Dawley , X-Ray Microtomography
18.
J Dermatolog Treat ; 27(3): 241-6, 2016.
Article in English | MEDLINE | ID: mdl-26581111

ABSTRACT

OBJECTIVES: The purpose of the present studies was to assess the safety, tolerability and pharmacokinetics of topical application of a novel phosphodiesterase inhibitor, E6005, in healthy volunteers and in patients with atopic dermatitis (AD). METHODS: In two randomized, investigator-blind, vehicle-controlled studies, we evaluated the topical application of E6005 ointment at concentrations ranging from 0.01% to 0.2% in healthy volunteers (Study 001) and in patients with AD (Study 101). RESULTS: Thirty-six subjects were enrolled in Study 001 and 40 in Study 101. Neither skin irritation nor photosensitization was observed with application of E6005 in Study 001. Four subjects receiving E6005 in Study 001 experienced a treatment-emergent adverse event (application site edema, increased alanine aminotransferase or erythema); three of these subjects discontinued the study. Two subjects receiving E6005 in Study 101 experienced an adverse event (gout or enterocolitis); one discontinued the study. Plasma concentrations of E6005 were below the limit of quantification (1 ng/ml) in both studies. CONCLUSION: E6005 ointment exhibited acceptable safety and tolerability. Topical application of E6005 ointment resulted in very low systemic exposure to E6005 in healthy volunteers and in patients with AD.


Subject(s)
Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Phosphodiesterase 4 Inhibitors , Phthalic Acids , Quinazolines , Administration, Topical , Adult , Aged , Double-Blind Method , Healthy Volunteers , Humans , Male , Middle Aged , Ointments , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 4 Inhibitors/pharmacokinetics , Phosphodiesterase 4 Inhibitors/therapeutic use , Phthalic Acids/adverse effects , Phthalic Acids/pharmacokinetics , Phthalic Acids/therapeutic use , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Young Adult
19.
Eur J Pharmacol ; 768: 41-8, 2015 Dec 05.
Article in English | MEDLINE | ID: mdl-26455478

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a progressive lung disease associated with significant morbidity and mortality. Although several oral phosphodiesterase 4 (PDE4) inhibitors have been developed for the treatment of COPD, their use has been restricted because of side effects including nausea and emesis. We hypothesized that delivery of a dry powdered PDE4 inhibitor by inhalation would minimize systemic absorption and enable local PDE4 inhibition to suppress inflammation within the lung. Neutrophilic pulmonary inflammation was induced in mice by intratracheal administration of lipopolysaccharide. Mice were treated intratracheally with a new dry powder PDE4 inhibitor, E6005 (methyl 4-[({3-[6,7-dimethoxy-2-(methylamino)quinazolin-4-yl]phenyl}amino) carbonyl] benzoate). The pharmacokinetics, cell profiles and levels of cytokines, chemokines, and lipid mediators in bronchoalveolar lavage fluid (BALF), and lung histology were assessed. Intratracheal administration of E6005 to mice resulted in high concentrations of the compound in the lungs. Histological analysis of E6005-treated mice demonstrated reduced inflammation of lung tissue that correlated with a decrease in BALF levels of neutrophils, proinflammatory cytokines, chemokines, and cysteinyl leukotrienes. Thus, intratracheal administration of E6005 effectively suppresses neutrophilic pulmonary inflammation, suggesting that the new inhaled dry powder PDE4 inhibitor represents an alternative to the conventional oral formulation for treating COPD.


Subject(s)
Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/pharmacology , Phthalic Acids/administration & dosage , Phthalic Acids/pharmacology , Pneumonia/drug therapy , Quinazolines/administration & dosage , Quinazolines/pharmacology , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid , Chemokines/metabolism , Female , Lipid Metabolism/drug effects , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Phosphodiesterase 4 Inhibitors/pharmacokinetics , Phosphodiesterase 4 Inhibitors/therapeutic use , Phthalic Acids/pharmacokinetics , Phthalic Acids/therapeutic use , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use
20.
Arthritis Res Ther ; 17: 20, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25641548

ABSTRACT

INTRODUCTION: Microfracture does not properly repair full-thickness cartilage defects. The purpose of this study was to evaluate the effect of intraarticular injection of the small-molecule compound kartogenin (KGN) on the restoration of a full-thickness cartilage defect treated with microfracture in a rabbit model. METHODS: Full-thickness cartilage defects (3.5 mm in diameter and 3 mm in depth) were created in the patellar groove of the right femurs of 24 female New Zealand White rabbits. The rabbits were divided into two groups (12 in each group) based on postsurgery treatment differences, as follows: microfracture plus weekly intraarticular injection of KGN (group 1) and microfracture plus dimethyl sulfoxide (group 2). Six rabbits from each group were illed at 4 and 12 weeks after surgery, and their knees were harvested. The outcome was assessed both macroscopically, by using the International Cartilage Repair Society (ICRS) macroscopic evaluation system, and histologically, by using the modified O'Driscoll histologic scoring system. Immunohistochemistry for type II and I collagen was also conducted. RESULTS: At 4 weeks, group 1 showed better defect filling and a greater number of chondrocyte-like cells compared with group 2. At 12 weeks, group 1 showed statistically significantly higher ICRS scores and modified O'Driscoll scores compared with group 2. More hyaline cartilage-like tissue was found in the defects of group 1 at 12 weeks. CONCLUSIONS: Intraarticular injection of KGN enhances the quality of full-thickness cartilage defects repair after microfracture, with better defect filling and increased hyaline-like cartilage formation.


Subject(s)
Anilides/therapeutic use , Cartilage, Articular/injuries , Fractures, Cartilage/drug therapy , Knee Injuries/drug therapy , Phthalic Acids/therapeutic use , Animals , Female , Fractures, Cartilage/pathology , Injections, Intra-Articular/methods , Knee Injuries/pathology , Models, Animal , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...