Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Virus Res ; 342: 199325, 2024 04.
Article in English | MEDLINE | ID: mdl-38309472

ABSTRACT

The COVID-19 pandemic caused by SARS-CoV-2 becomes a serious threat to global health and requires the development of effective antiviral therapies. Current therapies that target viral proteins have limited efficacy with side effects. In this study, we investigated the antiviral activity of MIT-001, a small molecule reactive oxygen species (ROS) scavenger targeting mitochondria, against SARS-CoV-2 and other zoonotic viruses in vitro. The antiviral activity of MIT-001 was quantified by RT-qPCR and plaque assay. We also evaluated the functional analysis of MIT-001 by JC-1 staining to measure mitochondrial depolarization, total RNA sequencing to investigate gene expression changes, and immunoblot to quantify protein expression levels. The results showed that MIT-001 effectively inhibited the replication of B.1.617.2 and BA.1 strains, Zika virus, Seoul virus, and Vaccinia virus. Treatment with MIT-001 restored the expression of heme oxygenase-1 (HMOX1) and NAD(P)H: quinone oxidoreductase 1 (NqO1) genes, anti-oxidant enzymes reduced by SARS-CoV-2, to normal levels. The presence of MIT-001 also alleviated mitochondrial depolarization caused by SARS-CoV-2 infection. These findings highlight the potential of MIT-001 as a broad-spectrum antiviral compound that targets for zoonotic RNA and DNA viruses, providing a promising therapeutic approach to combat viral infection.


Subject(s)
COVID-19 , Zika Virus Infection , Zika Virus , Humans , Animals , SARS-CoV-2 , Reactive Oxygen Species , Pandemics , Fishes , Antiviral Agents/pharmacology
2.
Int J Mol Sci ; 24(20)2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37894838

ABSTRACT

Ovarian aging is a major obstacle in assisted reproductive medicine because it leads to ovarian dysfunction in women of advanced age. Currently, there are no effective treatments to cure age-related ovarian dysfunction. In this study, we investigated the effect of MIT-001 on the function of aged ovaries. Young and old mice were utilized in this study. MIT-001 was intraperitoneally administered, and the number of follicles and oocytes was analyzed. Each group was then retrieved for RNA and protein isolation. Total RNA was subjected to mRNA next-generation sequencing. Protein extracts from ovarian lysates were used to evaluate various cytokine levels in the ovaries. MIT-001 enhanced follicles and the number of oocytes were compared with non-treated old mice. MIT-001 downregulated immune response-related transcripts and cytokines in the ovaries of old mice. MIT-001 modulates the immune complex responsible for generating inflammatory signals and has the potential to restore the function of old ovaries and improve female fertility.


Subject(s)
Oocytes , Ovarian Diseases , Female , Mice , Animals , Humans , Aged , Aging , Cytokines/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , RNA/pharmacology
3.
Nat Commun ; 14(1): 5728, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37714840

ABSTRACT

Arachidonic and adrenic acids in the membrane play key roles in ferroptosis. Here, we reveal that lipoprotein-associated phospholipase A2 (Lp-PLA2) controls intracellular phospholipid metabolism and contributes to ferroptosis resistance. A metabolic drug screen reveals that darapladib, an inhibitor of Lp-PLA2, synergistically induces ferroptosis in the presence of GPX4 inhibitors. We show that darapladib is able to enhance ferroptosis under lipoprotein-deficient or serum-free conditions. Furthermore, we find that Lp-PLA2 is located in the membrane and cytoplasm and suppresses ferroptosis, suggesting a critical role for intracellular Lp-PLA2. Lipidomic analyses show that darapladib treatment or deletion of PLA2G7, which encodes Lp-PLA2, generally enriches phosphatidylethanolamine species and reduces lysophosphatidylethanolamine species. Moreover, combination treatment of darapladib with the GPX4 inhibitor PACMA31 efficiently inhibits tumour growth in a xenograft model. Our study suggests that inhibition of Lp-PLA2 is a potential therapeutic strategy to enhance ferroptosis in cancer treatment.


Subject(s)
Ferroptosis , Neoplasms , Humans , 1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , Lipid Metabolism/drug effects , Neoplasms/drug therapy
4.
Am J Respir Cell Mol Biol ; 69(1): 57-72, 2023 07.
Article in English | MEDLINE | ID: mdl-36930952

ABSTRACT

Various environmental compounds are inducers of lung injury. Mitochondria are crucial organelles that can be affected by many lung diseases. NecroX is an indole-derived antioxidant that specifically targets mitochondria. We aimed to evaluate the therapeutic potential and related molecular mechanisms of NecroX in preclinical models of fatal lung injury. We investigated the therapeutic effects of NecroX on two different experimental models of lung injury induced by polyhexamethylene guanidine (PHMG) and bleomycin, respectively. We also performed transcriptome analysis of lung tissues from PHMG-exposed mice and compared the expression profiles with those from dozens of bleomycin-induced fibrosis public data sets. Respiratory exposure to PHMG and bleomycin led to fatal lung injury manifesting extensive inflammation followed by fibrosis. These specifically affected mitochondria regarding biogenesis, mitochondrial DNA integrity, and the generation of mitochondrial reactive oxygen species in various cell types. NecroX significantly improved the pathobiologic features of the PHMG- and bleomycin-induced lung injuries through regulation of mitochondrial oxidative stress. Endoplasmic reticulum stress was also implicated in PHMG-associated lung injuries of mice and humans, and NecroX alleviated PHMG-induced lung injury and the subsequent fibrosis, in part, via regulation of endoplasmic reticulum stress in mice. Gene expression profiles of PHMG-exposed mice were highly consistent with public data sets of bleomycin-induced lung injury models. Pathways related to mitochondrial activities, including oxidative stress, oxidative phosphorylation, and mitochondrial translation, were upregulated, and these patterns were significantly reversed by NecroX. These findings demonstrate that NecroX possesses therapeutic potential for fatal lung injury in humans.


Subject(s)
Lung Injury , Humans , Lung Injury/chemically induced , Lung Injury/drug therapy , Lung Injury/pathology , Guanidine/pharmacology , Lung/pathology , Guanidines/pharmacology , Oxidative Stress , Fibrosis , Bleomycin/pharmacology , Endoplasmic Reticulum Stress
5.
Pharmaceutics ; 15(1)2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36678851

ABSTRACT

Uncontrolled chronic inflammation and necrosis is characteristic of inflammatory bowel disease (IBD). This study aimed to investigate the effect of necrosis inhibitor (NI, NecroX-7) on a dextran sulfate sodium (DSS) induced chronic colitis model of mice. DSS was administered on days 1-5, and the NI was administered intraperitoneally (3 mg/kg, 30 mg/kg) on days 1, 3, and 5 as well as every other day during the first five days of a three-week cycle. Three cycles of administration were performed. Colitis was evaluated based on the disease activity index (DAI) score, colon length, and histological score. Reverse transcription polymerase chain reaction testing, the Western blot assay, and immunohistochemical staining were performed to determine inflammatory cytokine levels. The NI reduced body weight change and the DAI score. Colon length and the histological score were longer and lower in the NI-treated groups, respectively. The NI decreased the expression of pro-inflammatory cytokines, particularly in tumor necrosis factor alpha (TNF-α) and phosphorylated nuclear factor kappa B (p-NF-κB). Immunohistochemical staining revealed decreased inducible nitric oxide synthase (iNOS) and high mobility group box 1 (HMGB1) levels. Overall, the NI improved DSS induced chronic colitis by attenuating the mRNA expression of pro-inflammatory cytokines such as TNF-α. Therefore, NI use is a potential, novel treatment approach for IBD.

6.
Mol Cells ; 45(11): 855-867, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36172977

ABSTRACT

For proper function of proteins, their subcellular localization needs to be monitored and regulated in response to the changes in cellular demands. In this regard, dysregulation in the nucleocytoplasmic transport (NCT) of proteins is closely associated with the pathogenesis of various neurodegenerative diseases. However, it remains unclear whether there exists an intrinsic regulatory pathway(s) that controls NCT of proteins either in a commonly shared manner or in a target-selectively different manner. To dissect between these possibilities, in the current study, we investigated the molecular mechanism regulating NCT of truncated ataxin-3 (ATXN3) proteins of which genetic mutation leads to a type of polyglutamine (polyQ) diseases, in comparison with that of TDP-43. In Drosophila dendritic arborization (da) neurons, we observed dynamic changes in the subcellular localization of truncated ATXN3 proteins between the nucleus and the cytosol during development. Moreover, ectopic neuronal toxicity was induced by truncated ATXN3 proteins upon their nuclear accumulation. Consistent with a previous study showing intracellular calcium-dependent NCT of TDP-43, NCT of ATXN3 was also regulated by intracellular calcium level and involves Importin α3 (Imp α3). Interestingly, NCT of ATXN3, but not TDP-43, was primarily mediated by CBP. We further showed that acetyltransferase activity of CBP is important for NCT of ATXN3, which may acetylate Imp α3 to regulate NCT of ATXN3. These findings demonstrate that CBP-dependent acetylation of Imp α3 is crucial for intracellular calcium-dependent NCT of ATXN3 proteins, different from that of TDP-43, in Drosophila neurons.


Subject(s)
Drosophila , alpha Karyopherins , Animals , Acetylation , Active Transport, Cell Nucleus , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , Ataxin-3/genetics , Ataxin-3/metabolism , Calcium/metabolism , Drosophila/metabolism , Neurons/metabolism
7.
Int J Mol Sci ; 22(10)2021 May 11.
Article in English | MEDLINE | ID: mdl-34064719

ABSTRACT

Inflammation is a major cause of several chronic diseases and is reported to be recovered by the immuno-modulation of mesenchymal stem cells (MSCs). While most studies have focussed on the anti-inflammatory roles of MSCs in stem cell therapy, the impaired features of MSCs, such as the loss of homeostasis by systemic aging or pathologic conditions, remain incompletely understood. In this study, we investigated whether the altered phenotypes of human placenta-derived MSCs (hPD-MSCs) exposed to inflammatory cytokines, including TNF-α and IFN-γ, could be protected by MIT-001, a small anti-inflammatory and anti-necrotic molecule. MIT-001 promoted the spindle-like shape and cytoskeletal organization extending across the long cell axis, whereas hPD-MSCs exposed to TNF-α/IFN-γ exhibited increased morphological heterogeneity with an abnormal cell shape and cytoskeletal disorganization. Importantly, MIT-001 improved mitochondrial distribution across the cytoplasm. MIT-001 significantly reduced basal respiration, ATP production, and cellular ROS levels and augmented the spare respiratory capacity compared to TNF-α/IFN-γ-exposed hPD-MSCs, indicating enhanced mitochondrial quiescence and homeostasis. In conclusion, while TNF-α/IFN-γ-exposed MSCs lost homeostasis and mitochondrial quiescence by becoming over-activated in response to inflammatory cytokines, MIT-001 was able to rescue mitochondrial features and cellular phenotypes. Therefore, MIT-001 has therapeutic potential for clinical applications to treat mitochondrion-related inflammatory diseases.


Subject(s)
Cytoskeleton/physiology , Mesenchymal Stem Cells/physiology , Mitochondria/physiology , Organic Chemicals/pharmacology , Placenta/cytology , Cytoskeleton/drug effects , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mitochondria/drug effects , Oxygen Consumption , Placenta/drug effects , Placenta/metabolism , Pregnancy , Reactive Oxygen Species/metabolism
8.
Clin Ther ; 42(10): 1946-1954.e2, 2020 10.
Article in English | MEDLINE | ID: mdl-32980184

ABSTRACT

PURPOSE: LC28-0126 is a reactive oxygen species scavenger being developed for the treatment of various conditions caused by oxidative stress, such as oral mucositis, graft-versus-host disease, and lethal reperfusion injury in acute myocardial infarction. The aim of this study was to assess the tolerability and pharmacokinetic properties of LC28-0126 with multiple IV administrations in healthy male subjects. METHODS: A dose-block-randomized, double-blind, placebo-controlled, multiple ascending-dose study was conducted. Subjects received 3-, 10-, 20-, or 30-mg doses of LC28-0126 or inactive control vehicle, infused over 30 min, once daily for 7 days. Blood and urine samples were collected for pharmacokinetics assessment. Tolerability was assessed by the documentation of adverse events, including abnormal findings on physical examination, vital sign measurements, blood oxygen saturation monitoring, 12-lead ECG, continuous ECG monitoring, and clinical laboratory testing. FINDINGS: A total of 32 subjects completed the study. After multiple dosing, the plasma concentration of LC28-0126 showed a steep decrease after infusion, followed by slow elimination. Systemic exposure of LC28-0126 was increased proportionally to doses ranging from 3 to 30 mg. The accumulation ratios were 2.58-2.79 on multiple dosing. The fractions excreted unchanged in urine were found to be <5%. All reported drug-related adverse events were injection-site reactions, and no serious adverse events were reported. IMPLICATIONS: Multiple administrations of LC28-0126 exhibited a dose-proportional pharmacokinetic profile and were well tolerated at a dose range of 3-30 mg. ClinicalTrials.gov identifier: NCT03196804.


Subject(s)
Hydrocarbons, Halogenated/administration & dosage , Ketones/administration & dosage , Adult , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Electrocardiography , Humans , Hydrocarbons, Halogenated/adverse effects , Hydrocarbons, Halogenated/pharmacokinetics , Ketones/adverse effects , Ketones/pharmacokinetics , Male , Young Adult
9.
Virulence ; 11(1): 1225-1239, 2020 12.
Article in English | MEDLINE | ID: mdl-32835604

ABSTRACT

The global incidence of Mycobacterium abscessus (Mabc), a rapidly growing nontuberculous mycobacterial strain that causes treatment-refractory pulmonary diseases, is increasing. Despite this, the host factors that allow for protection against infection are largely unknown. In this study, we found that sirtuin 3 (SIRT3), a mitochondrial protein deacetylase, plays a critical role in host defense against Mabc infection. Mabc decreased SIRT3 and upregulated mitochondrial oxidative stress in macrophages. SIRT3 deficiency led to increased bacterial loads, histopathological, and mitochondrial damage, and pathological inflammation during Mabc infection. Administration of scavengers of mitochondrial reactive oxygen species significantly decreased the in vivo Mabc burden and excessive inflammation, and induced SIRT3 expression in infected lungs. Notably, SIRT3 agonist (resveratrol) significantly decreased Mabc growth and attenuated inflammation in mice and zebrafishes, indicating the key role for SIRT3 in metazoan host defense. Collectively, these data strongly suggest that SIRT3 is a host-directed therapeutic target against Mabc infection by controlling mitochondrial homeostasis.


Subject(s)
Homeostasis , Host-Pathogen Interactions , Mitochondria/physiology , Mycobacterium Infections, Nontuberculous/prevention & control , Sirtuin 3/genetics , Animals , Gene Expression Regulation , Macrophages/microbiology , Macrophages/physiology , Male , Mice , Mycobacterium abscessus/growth & development , Mycobacterium abscessus/pathogenicity , Oxidative Stress , Reactive Oxygen Species , Sirtuin 3/metabolism , Zebrafish/microbiology
10.
Hypertension ; 71(6): 1143-1155, 2018 06.
Article in English | MEDLINE | ID: mdl-29661840

ABSTRACT

Opening of mitochondrial permeability transition pore and Ca2+ overload are main contributors to myocardial ischemia-reperfusion injury, which paradoxically causes a wide variety of myocardial damage. We investigated the protective role of a novel necrosis inhibitor (NecroX-7; NecX) against myocardial ischemia-reperfusion injury using in vitro and in vivo models. H9C2 rat cardiomyoblasts and neonatal cardiomyocytes were exposed to hypoxia-reoxygenation stress after pre-treatment with NecX, vitamin C, a combination of vitamin C and E, N-acetylcysteine, an apoptosis inhibitor (Z-VAD-fmk), or cyclosporine A. The main mechanism of cell death after hypoxia-reoxygenation stress was not apoptosis but necrosis, which was prevented by NecX. Protective effect of NecX was based on its potent reactive oxygen species scavenging activity, especially on mitochondrial reactive oxygen species. NecX preserved mitochondrial membrane potential through prevention of Ca2+ influx and inhibition of mitochondrial permeability transition pore opening, which was more potent than that by cyclosporine A. Using Sprague-Dawley rats exposed to myocardial ischemia for 45 minutes followed by reperfusion, we compared therapeutic efficacies of NecX with cyclosporine A, vitamin C, a combination of vitamin C and E, and 5% dextrose, each administered 5 minutes before reperfusion. NecX markedly inhibited myocardial necrosis and reduced fibrotic area to a greater extent than did cyclosporine A and other treated groups. In addition, NecX preserved systolic function and prevented pathological dilatory remodeling of left ventricle. The novel necrosis inhibitor has a significant protective effect against myocardial ischemia-reperfusion injury through inhibition of mitochondrial permeability transition pore opening, indicating that it is a promising candidate for cardioprotective adjunctive measure on top of reperfusion therapy.


Subject(s)
Indoles/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/pathology , Animals , Cell Death , Cells, Cultured , Disease Models, Animal , Male , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
11.
Thorax ; 73(8): 758-768, 2018 08.
Article in English | MEDLINE | ID: mdl-29622694

ABSTRACT

BACKGROUND: Respiratory fungal exposure is known to be associated with severe allergic lung inflammation. Airway epithelium is an essential controller of allergic inflammation. An innate immune recognition receptor, nucleotide-binding domain, leucine-rich-containing family, pyrin-domain-containing-3 (NLRP3) inflammasome, and phosphoinositide 3 kinase (PI3K)-δ in airway epithelium are involved in various inflammatory processes. OBJECTIVES: We investigated the role of NLRP3 inflammasome in fungi-induced allergic lung inflammation and examined the regulatory mechanism of NLRP3 inflammasome, focusing on PI3K-δ in airway epithelium. METHODS: We used two in vivo models induced by exposure to Aspergillus fumigatus (Af) and Alternaria alternata (Aa), as well as an Af-exposed in vitro system. We also checked NLRP3 expression in lung tissues from patients with allergic bronchopulmonary aspergillosis (ABPA). RESULTS: Assembly/activation of NLRP3 inflammasome was increased in the lung of Af-exposed mice. Elevation of NLRP3 inflammasome assembly/activation was observed in Af-stimulated murine and human epithelial cells. Similarly, pulmonary expression of NLRP3 in patients with ABPA was increased. Importantly, neutralisation of NLRP3 inflammasome derived IL-1ß alleviated pathophysiological features of Af-induced allergic inflammation. Furthermore, PI3K-δ blockade improved Af-induced allergic inflammation through modulation of NLRP3 inflammasome, especially in epithelial cells. This modulatory role of PI3K-δ was mediated through the regulation of mitochondrial reactive oxygen species (mtROS) generation. NLRP3 inflammasome was also implicated in Aa-induced eosinophilic allergic inflammation, which was improved by PI3K-δ blockade. CONCLUSION: These findings demonstrate that fungi-induced assembly/activation of NLRP3 inflammasome in airway epithelium may be modulated by PI3K-δ, which is mediated partly through the regulation of mtROS generation. Inhibition of PI3K-δ may have potential for treating fungi-induced severe allergic lung inflammation.


Subject(s)
Alternariosis/enzymology , Alternariosis/immunology , Aspergillosis, Allergic Bronchopulmonary/enzymology , Aspergillosis, Allergic Bronchopulmonary/immunology , Endoplasmic Reticulum Stress/immunology , Immunity, Innate/immunology , Phosphatidylinositol 3-Kinases/immunology , Animals , Aspergillus fumigatus , Biomarkers/analysis , Bronchi/cytology , Cells, Cultured , Epithelial Cells/immunology , Female , Humans , Inflammasomes/immunology , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/immunology
12.
Br J Clin Pharmacol ; 83(6): 1205-1215, 2017 06.
Article in English | MEDLINE | ID: mdl-28002882

ABSTRACT

AIMS: A novel necrosis inhibitor, LC28-0126, is expected to have a cellular protective effect from ischaemic reperfusion injury in acute myocardial infarction. The objective of this study was to investigate the safety, tolerability and pharmacokinetics of LC28-0126 after a single intravenous administration in healthy male subjects. METHODS: The study was a dose-block-randomized, double-blind, placebo-controlled, single ascending dose, first-in-human trial. Subjects were randomly assigned to receive 0.3, 1, 3, 10, 25, 50, 100 or 200 mg of LC28-0126. LC28-0126 was infused for 30 min and 5 min in cohorts 1 and 2, respectively. An interim analysis to assess the tolerability and pharmacokinetics was conducted in each dose group. Blood samples were taken to determine plasma LC28-0126 concentrations from predose to 48 or 144 h postdose, and urine samples were taken from predose to 48 or 72 h postdose. RESULTS: Overall, 89 subjects were randomly assigned to the dose groups of the two cohorts. LC28-0126 was well tolerated, and no serious adverse events were reported. LC28-0126 showed rapid disposition in the distribution phase. Overall, the fraction of unchanged LC28-0126 excreted during the 48 or 72 h after administration was below 5%. The systemic exposure of LC28-0126 tends to be increased in a dose-proportional manner in the dose range of 0.3-200 mg. CONCLUSIONS: A single intravenous dose of LC28-0126 was safe and well tolerated up to 200 mg. Furthermore, LC28-0126 demonstrated a predictable pharmacokinetic profile after a single intravenous infusion of doses ranging from 0.3 to 200 mg.


Subject(s)
Necrosis/prevention & control , Adult , Area Under Curve , Asian People , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Middle Aged , Myocardial Reperfusion Injury/prevention & control , No-Observed-Adverse-Effect Level , Young Adult
13.
Int J Oncol ; 50(1): 185-192, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27922686

ABSTRACT

A major goal of breast cancer research is to prevent the molecular events that lead to tumour metastasis. It is well-established that both cytoplasmic and mitochondrial reactive oxygen species (ROS) play important roles in cell migration and metastasis. Accordingly, this study examined the molecular mechanisms of the anti-metastatic effects of NecroX-5, a mitochondrial ROS scavenger. NecroX-5 inhibited lung cancer metastasis by ameliorating migration in a mouse model. In human cancer cells, the inhibition of migration by NecroX-5 is cell type-dependent. We observed that the effect of NecroX-5 correlated with a reduction in mitochondrial ROS, but mitochondrial ROS reduction by MitoQ did not inhibit cell migration. NecroX-5 decreased intracellular calcium concentration by blocking Ca2+ influx, which mediated the inhibition of cell migration, AKT downregulation and the reduction of mitochondrial ROS levels. However, the reduction of mitochondrial ROS was not associated with supressed migration and AKT downregulation. Our study demonstrates the potential of NecroX-5 as an inhibitor of breast cancer metastasis.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement/drug effects , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Oncogene Protein v-akt/biosynthesis , Sulfones/administration & dosage , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Metastasis , Oncogene Protein v-akt/genetics , Organophosphorus Compounds/administration & dosage , Reactive Oxygen Species/metabolism , Ubiquinone/administration & dosage , Ubiquinone/analogs & derivatives , Xenograft Model Antitumor Assays
14.
Neuroreport ; 27(15): 1128-33, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27541272

ABSTRACT

NecroX-5 is a derivative of cyclopentylamino carboxymethylthiazolylindole (NecroX), an inhibitor of necrosis/necroptosis. NecroX-5 has been shown to scavenge mitochondrial reactive oxygen and nitrogen species, and thus preventing necrotic cell death against various kinds of oxidative stress in several tissues, including the brain. To examine the effect of NecroX-5 on retinal degeneration (RD), RD was induced in Sprague-Dawley rats by an intraperitoneal injection of N-methyl-N-nitrosourea and in BALB/c mice by blue light-emitting diode exposure. Scotopic electroretinography recording was used to evaluate retinal function. For histological evaluation, hematoxylin and eosin staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunohistochemistry were performed. Electroretinography recordings showed that a-waves and b-waves were significantly reduced in both RD rats and mice, whereas the amplitudes of both waves were significantly increased in both NecroX-5-treated RD rats and mice compared with untreated RD animals. In hematoxylin and eosin staining and terminal deoxynucleotidyl transferase dUTP nick end labeling assay, the outer nuclear layer where photoreceptors reside appeared to be more preserved, and there were fewer apoptotic cells in NecroX-5-treated RD retinas than in untreated RD retinas. In addition, immunohistochemistry with antiglial fibrillary acidic protein and anti-8-hydroxy-2'-deoxyguanosine showed lower levels of retinal injury and oxidative stress in NecroX-5-treated RD retinas than in untreated RD retinas. These results indicated that NecroX-5 protects retinal neurons from experimentally induced RD, suggesting that NecroX-5 may have a potential for the treatment of RD as a medication.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/therapeutic use , Neuroprotective Agents/therapeutic use , Retinal Degeneration/drug therapy , Sulfones/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Electroretinography , Glial Fibrillary Acidic Protein/metabolism , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley
15.
Atherosclerosis ; 252: 166-174, 2016 09.
Article in English | MEDLINE | ID: mdl-27425215

ABSTRACT

BACKGROUND AND AIMS: A large necrotic core is a key feature of atherosclerotic plaque instability. Necrotic cellular debris accumulates in the lipid-rich core and promotes inflammation, destabilization and ultimately rupture of the plaque. Although the role of necrosis in atherosclerosis is rather clear-cut, not many strategies have been performed up till now to specifically target plaque necrosis. In the present study, we tested the plaque stabilizing potential of NecroX-7, a novel compound with antioxidative and anti-necrotic properties. METHODS: Male apolipoprotein E (Apoe) knockout mice were treated with NecroX-7 (30 mg/kg) or vehicle, 3 times per week, via intraperitoneal injections for 16 weeks. Meanwhile, mice were fed a western-type diet to induce plaque formation. RESULTS: NecroX-7 reduced total plaque burden in the thoracic aorta as compared to vehicle-treated mice, without affecting total plasma cholesterol. Plaques in the aortic root of NecroX-7-treated mice showed a significant decrease in necrotic core area, 8-oxodG, iNOS and MMP13 expression, while collagen content and minimum fibrous cap thickness were increased. Moreover, NecroX-7 treatment reduced the expression of multiple inflammation markers such as TNFα, IL1ß, iNOS, HMGB1 and RAGE in a NF-κB-dependent manner. In vitro, NecroX-7 prevented tert-butyl hydroperoxide (tBHP)-induced mitochondrial ROS formation, necrosis, iNOS expression and HMGB1 release in primary macrophages. CONCLUSIONS: NecroX-7 improves features of plaque stability in Apoe knockout mice by reducing necrotic core formation, oxidative stress and inflammation, and by increasing collagen deposition and fibrous cap thickness. Therefore, NecroX-7 could be a promising pleiotropic drug for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , HMGB1 Protein/metabolism , Macrophages/metabolism , Organic Chemicals/pharmacology , Oxidative Stress , Plaque, Atherosclerotic/drug therapy , Animals , Atherosclerosis/metabolism , Bone Marrow Cells/cytology , Cholesterol, LDL/metabolism , Collagen/metabolism , Inflammation , Lipid Peroxidation , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Plaque, Atherosclerotic/metabolism
16.
Korean J Physiol Pharmacol ; 20(3): 305-14, 2016 May.
Article in English | MEDLINE | ID: mdl-27162485

ABSTRACT

Inflammatory and fibrotic responses are accelerated during the reperfusion period, and excessive fibrosis and inflammation contribute to cardiac malfunction. NecroX compounds have been shown to protect the liver and heart from ischemia-reperfusion injury. The aim of this study was to further define the role and mechanism of action of NecroX-5 in regulating infl ammation and fi brosis responses in a model of hypoxia/reoxygenation (HR). We utilized HR-treated rat hearts and lipopolysaccharide (LPS)-treated H9C2 culture cells in the presence or absence of NecroX-5 (10 µmol/L) treatment as experimental models. Addition of NecroX-5 signifi cantly increased decorin (Dcn) expression levels in HR-treated hearts. In contrast, expression of transforming growth factor beta 1 (TGFß1) and Smad2 phosphorylation (pSmad2) was strongly attenuated in NecroX-5-treated hearts. In addition, signifi cantly increased production of tumor necrosis factor alpha (TNFα), TGFß1, and pSmad2, and markedly decreased Dcn expression levels, were observed in LPS-stimulated H9C2 cells. Interestingly, NecroX-5 supplementation effectively attenuated the increased expression levels of TNFα, TGFß1, and pSmad2, as well as the decreased expression of Dcn. Thus, our data demonstrate potential antiinflammatory and anti-fibrotic effects of NecroX-5 against cardiac HR injuries via modulation of the TNFα/Dcn/TGFß1/Smad2 pathway.

17.
Korean J Physiol Pharmacol ; 20(2): 201-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26937217

ABSTRACT

Although the antioxidant and cardioprotective effects of NecroX-5 on various in vitro and in vivo models have been demonstrated, the action of this compound on the mitochondrial oxidative phosphorylation system remains unclear. Here we verify the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity during hypoxia-reoxygenation (HR). Necrox-5 treatment (10 µM) and non-treatment were employed on isolated rat hearts during hypoxia/reoxygenation treatment using an ex vivo Langendorff system. Proteomic analysis was performed using liquid chromatography-mass spectrometry (LC-MS) and non-labeling peptide count protein quantification. Real-time PCR, western blot, citrate synthases and mitochondrial complex activity assays were then performed to assess heart function. Treatment with NecroX-5 during hypoxia significantly preserved electron transport chain proteins involved in oxidative phosphorylation and metabolic functions. NecroX-5 also improved mitochondrial complex I, II, and V function. Additionally, markedly higher peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC1α) expression levels were observed in NecroX-5-treated rat hearts. These novel results provide convincing evidence for the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity and in preserving PGC1α during cardiac HR injuries.

18.
Sci Rep ; 5: 17784, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626943

ABSTRACT

In recent decades, global pharmaceutical companies have suffered from an R&D innovation gap between the increased cost of a new drug's development and the decreased number of approvals. Drug repositioning offers another opportunity to fill the gap because the approved drugs have a known safety profile for human use, allowing for a reduction of the overall cost of drug development by eliminating rigorous safety assessment. In this study, we compared the transcriptional profile of LC28-0126, an investigational drug for acute myocardial infarction (MI) at clinical trial, obtained from healthy male subjects with molecular activity profiles in the Connectivity Map. We identified dyphilline, an FDA-approved drug for bronchial asthma, as a top ranked connection with LC28-0126. Subsequently, we demonstrated that LC28-0126 effectively ameliorates the pathophysiology of neutrophilic bronchial asthma in OVALPS-OVA mice accompanied with a reduction of inflammatory cell counts in the bronchoalveolar lavage fluid (BALF), inhibition of the release of proinflammatory cytokines, relief of airway hyperactivity, and improvement of histopathological changes in the lung. Taken together, we suggest that LC28-0126 could be a potential therapeutic for bronchial asthma. In addition, this study demonstrated the potential general utility of computational drug repositioning using clinical profiles of the investigational drug.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/pharmacokinetics , Asthma/drug therapy , Asthma/metabolism , Cytokines/biosynthesis , Gene Expression Regulation/drug effects , Adult , Animals , Anti-Asthmatic Agents/adverse effects , Asthma/pathology , Disease Models, Animal , Humans , Male , Mice , Middle Aged
19.
J Immunol ; 194(11): 5223-32, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25911749

ABSTRACT

Graft-versus-host disease (GVHD) is a major complication associated with allogeneic hematopoietic stem cell transplantation. Despite the prominent role of the adaptive immune system, the importance of controlling the innate immune system in the pathogenesis of GVHD has recently been rediscovered. High-mobility group box 1 (HMGB1) is a crucial damage-associated molecular pattern signal that functions as a potent innate immune mediator in GVHD. In the present study, we investigated treatment of experimental GVHD through HMGB1 blockade using the compound cyclopentylamino carboxymethylthiazolylindole (NecroX)-7. Treated animals significantly attenuated GVHD-related mortality and inhibited severe tissue damage. These protective effects correlated with the decrease in HMGB1 expression and lower levels of reactive oxidative stress. Additionally, NecroX-7 inhibited the HMGB1-induced release of TNF and IL-6, as well as the expression of TLR-4 and receptor for advanced glycation end products. We also observed increased regulatory T cell numbers, which may be associated with regulation of differentiation signals independent of HMGB1. Taken together, these data indicate that NecroX-7 protects mice against lethal GVHD by reciprocal regulation of regulatory T/Th1 cells, attenuating systemic HMGB1 accumulation and inhibiting HMGB1-mediated inflammatory response. Our results indicate the possibility of a new use for a clinical drug that is effective for the treatment of GVHD.


Subject(s)
Free Radical Scavengers/therapeutic use , Graft vs Host Disease/drug therapy , HMGB1 Protein/metabolism , Organic Chemicals/therapeutic use , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Cell Differentiation/drug effects , Cell Line , Cell Proliferation , Female , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Stress/drug effects , Protein Kinase C/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Receptor for Advanced Glycation End Products , Receptors, Immunologic/biosynthesis , T-Lymphocytes, Regulatory/cytology , Toll-Like Receptor 4/biosynthesis , Transplantation, Homologous , Tumor Necrosis Factor-alpha/metabolism
20.
Liver Int ; 35(4): 1341-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25443620

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic steatohepatitis (NASH) is associated with cirrhosis and hepatocellular carcinoma. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play key roles in the development of the disease. However, the therapeutic target of NASH has not been fully defined and new treatments are needed. We investigated the protective effects of the antioxidant indole-derived NecroX-7 in a NASH mouse model using leptin-deficient ob/ob and methionine- and choline-deficient (MCD) diet-fed ob/ob mice. METHODS: Six-week-old male mice were divided into three groups: ob/+ mice, ob/ob mice treated with vehicle and ob/ob mice treated daily with NecroX-7 (20 mg/kg) for 4 weeks. To study the effects of NecroX-7 in a fibrosis model, NASH was induced by feeding ob/ob mice an MCD diet. The effects of NecroX-7 on NASH progression were evaluated using biochemical, histological and molecular markers. RESULTS: NecroX-7-treated ob/ob mice had a marked decrease in serum aspartate aminotransferase and alanine transaminase compared with vehicle-treated controls. Interestingly, hepatic steatosis and lipid peroxidation were significantly improved by NecroX-7 treatment. NecroX-7 inhibited tert-butylhydroperoxide- and H2 O2 -induced mitochondrial ROS/RNS in primary hepatocytes and attenuated mitochondrial dysfunction in vitro and in vivo. Furthermore, NecroX-7-treated mice exhibited fewer infiltrating macrophages and reduced hepatic tumour necrosis factor-alpha expression. Hepatic fibrosis in MCD-fed ob/ob mice was significantly decreased by NecroX-7 treatment. CONCLUSIONS: NecroX-7 treatment improved hepatic steatosis and fibrosis in murine NASH models. These effects occurred through the suppression of whole-cell ROS/RNS and inflammatory responses and suggest that NecroX-7 has a potential therapeutic benefit in steatohepatitis.


Subject(s)
Antioxidants/pharmacology , Inflammation/drug therapy , Liver/drug effects , Mitochondria, Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Organic Chemicals/pharmacology , Oxidative Stress/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Animals , Cytoprotection , Energy Metabolism/drug effects , Hep G2 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Obese , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...