Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 290
Filter
1.
Cardiovasc J Afr ; 34(2): 74-80, 2023.
Article in English | MEDLINE | ID: mdl-35687060

ABSTRACT

Ischaemic heart disease (IHD) is a leading cause of death worldwide. Understanding prosurvival signalling pathways that protect against ischaemia-reperfusion injury (IRI) may assist in the development of novel cardioprotective strategies against IHD. In this regard, the transcription factor, nuclear factor kappa-B (NFκB) is activated by tumour necrosis factor (TNF), but its role in TNF-induced cytoprotection is unknown. Therefore, to investigate the role of NFκB in TNF-induced cytoprotection, C2C12 cells were pretreated with TNF (0.5 ng/ml) in the presence and absence of an NFκB inhibitor, pyrrolidine derivative of dithiocarbamate (PDTC; 100 µM). Cells were subjected to simulated IRI and treated with PDTC, either during TNF exposure or at reperfusion. Phosphorylation of IkB was measured after the TNF stimulus. Cytoprotection by TNF in cells subjected to IRI (cell viability: 43.7 ± 8.1% in control vs 70.6 ± 6.1% with TNF, p < 0.001) was abrogated by co-administration of PDTC (40.6 ± 1.9%, p < 0.001 vs TNF) but not by exposure to PDTC at reperfusion (70.7 ± 1.7%). Cytosolic IkB phosphorylation [1.5 ± 0.2 arbitrary units (AU) for TNF vs 1.0 ± 0.0 for untreated, p < 0.01]) was increased after TNF exposure and this increase was abolished by co-administration with PDTC (0.8 ± 0.3 AU, p < 0 01 vs TNF). Our data suggest that NFκB acts as a key component in TNF-induced cytoprotection. These findings may pave the way for the development of novel therapeutic drugs that target TNF/NFκB signalling to protect against IHD.


Subject(s)
Cytoprotection , NF-kappa B , Humans , NF-kappa B/metabolism , Thiocarbamates/pharmacology , Thiocarbamates/therapeutic use , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism
2.
Acta Biomater ; 155: 333-346, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36243373

ABSTRACT

Peritendinous adhesion is a major cause of limb dysfunction and disability in clinical practice. Numerous studies suggest that activation of nuclear factor-κB (NF-κB) pathway in macrophages could be the pivotal figure in excessive collagen synthesis and thus peritendinous adhesion formation. In this study, we assumed this pathological process could be suppressed by inhibiting NF-κB phosphorylation and nuclear translocation using pyrrolidine dithiocarbamate (PDTC), a specific NF-κB inhibitor with the ability to penetrate cell membranes, in macrophages. Then, we conducted electrospinning process to incorporate PDTC into poly(L-lactic) acid (PLA) electrospinning membranes, that is, the PDTC-PLA membranes. Further, with integral film quality and stable drug release property, the PDTC-PLA membranes were subsequently analyzed in the capability and mechanism of preventing adhesion formation both in vitro and in vivo. Our results showed inhibition of macrophage proliferation as well as NF-κB pathway activation from in vitro assays and outstanding promotion in inhibiting NF-κB p65 phosphorylation and reducing adhesion formation from in vivo assays of PDTC-PLA compared to PLA membranes. In conclusion, our findings suggested that PDTC-PLA as an alternative therapeutic approach alleviated inflammation and peritendinous adhesion formation through NF-κB signaling pathway. STATEMENT OF SIGNIFICANCE: Pyrrolidine dithiocarbamate (PDTC) can be blended into poly-L-lactic acid (PLA) fibrous membranes by electrospinning process. This incorporation of PDTC into PLA is an effective way to inhibit proinflammatory activation of macrophages and to achieve advanced anti-adhesion outcome after tendon repair.


Subject(s)
NF-kappa B , Thiocarbamates , NF-kappa B/metabolism , Thiocarbamates/pharmacology , Thiocarbamates/therapeutic use , Antioxidants/pharmacology , Polyesters/pharmacology
3.
Oxid Med Cell Longev ; 2021: 5594059, 2021.
Article in English | MEDLINE | ID: mdl-34691357

ABSTRACT

Reactive oxygen species (ROS) production is involved in the mechanism of action of a number of drugs, but the biological effects of ROS remain to be clarified. Furthermore, ferroptosis involves iron-dependent ROS production that may be derived from ferritinophagy; however, the association between ferroptosis and ferritinophagy has not been fully established. The present study demonstrated that dithiocarbamate derivatives (iron chelators) exhibited antineoplastic properties involving ferritinophagy induction, but whether the underlying mechanisms involved ferroptosis was unknown. To gain insight into the underlying mechanism, a dithiocarbamate derivative, 2-pyridylhydrazone dithiocarbamate s-acetic acid (PdtaA), was prepared. An MTT assay demonstrated that PdtaA inhibited proliferation involving ROS production (IC50 = 23.0 ± 1.5 µM for HepG2 cells). A preliminary mechanistic study revealed that PdtaA induced both apoptosis and cell cycle arrest. Notably, PdtaA also induced ferroptosis via downregulation of GPx4 and xCT, which was first reported for a dithiocarbamate derivative. Moreover, these cellular events were associated with ROS production. To explore the origin of ROS, expression of the ferritinophagy-related genes, ferritin, and nuclear receptor coactivator (NCOA4) were measured. Immunofluorescence and western blotting analysis indicated that PdtaA-induced ferritinophagy may contribute to ROS production. To investigate the role of ferritinophagy, autophagy inhibitor 3-methyladenin or genetic knockdown of NCOA4 was employed to inhibit ferritinophagy, which significantly neutralized the action of PdtaA in both apoptosis and ferroptosis. Taken together, PdtaA-induced cell cycle arrest, apoptosis, and ferroptosis were associated with ferritinophagy.


Subject(s)
Ferritins/metabolism , Ferroptosis/drug effects , Reactive Oxygen Species/metabolism , Thiocarbamates/therapeutic use , Apoptosis , Cell Proliferation , Humans , Thiocarbamates/pharmacology
4.
Biol Pharm Bull ; 44(10): 1419-1426, 2021.
Article in English | MEDLINE | ID: mdl-34602551

ABSTRACT

Sphingolipids (SLs), such as ceramide, glucosylceramide (GlcCer), and sphingomyelin, play important roles in the normal development/functions of the brain and peripheral tissues. Disruption of SL homeostasis in cells/organelles, specifically up-regulation of ceramide, is involved in multiple diseases including Alzheimer's disease (AD). One of the pathological features of AD is aggregates of amyloid beta (Aß) peptides, and SLs regulate both the formation/aggregation of Aß and Aß-induced cellular responses. Up-regulation of ceramide levels via de novo and salvage synthesis pathways is reported in Aß-treated cells and brains with AD; however, the effects of Aß on ceramide decomposition pathways have not been elucidated. Thus, we investigated the effects of the 25-35-amino acid Aß peptide (Aß25-35), the fundamental cytotoxic domain of Aß, on SL metabolism in cells treated with the fluorescent nitrobenzo-2-oxa-1,3-diazole-labeled C6-ceramide (NBD-ceramide). Aß25-35 treatment reduced the formation of NBD-GlcCer mediated by GlcCer synthase (GCS) without affecting the formation of NBD-sphingomyelin or NBD-ceramide-1-phosphate, and reduced cell viability. Aß25-35-induced responses decreased in cells treated with D609, a putative inhibitor of sphingomyelin synthases. Aß25-35-induced cytotoxicity significantly increased in GCS-knockout cells and pharmacological inhibition of GCS alone demonstrated cytotoxicity. Our study revealed that Aß25-35-induced cytotoxicity is at least partially mediated by the inhibition of GCS activity.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Glucosyltransferases/antagonists & inhibitors , Norbornanes/pharmacology , Peptide Fragments/metabolism , Thiocarbamates/pharmacology , Alzheimer Disease/pathology , Cell Line , Glucosyltransferases/metabolism , Humans , Norbornanes/therapeutic use , Thiocarbamates/therapeutic use , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Transferases (Other Substituted Phosphate Groups)/metabolism
5.
Scand J Gastroenterol ; 56(10): 1131-1139, 2021 10.
Article in English | MEDLINE | ID: mdl-34310252

ABSTRACT

OBJECTIVE: To understand the protective effect of NF-κB signaling pathway inhibitor pyrrolidinedithiocarbamate (PDTC) on mice with chronic atrophic gastritis (CAG). METHODS: Helicobacter pylori (H. pylori) infection combined with high-salt diet was used to construct the CAG mouse model, and 100 or 200 mg/kg/day PDTC was intragastrically treated for 8 weeks. Then, hematoxylin and eosin (HE) and Alcian blue-periodic acid-Schiff (AB-PAS) staining were used to observe the pathology of gastric mucosa, while immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), enzyme-linked immuno sorbent assay (ELISA) and western blotting were determined to detect the expression of related molecules. RESULTS: The nuclear content of NF-κB p65 in the gastric mucosa of the CAG mice was increased accompanying by the structural disorder of the gastric mucosal epithelium, inflammatory cell infiltration, intestinal metaplasia, and increased MUC2 expression, but the symptoms were alleviated after PDTC treatment. In addition, the expressions of TNF-α, IL-1ß, IL-6 and COX2 in the gastric mucosa and serum of CAG mice were higher than those control mice, which were reduced in CAG mice treated with either 100 or 200 mg/kg PDTC. Furthermore, 100 mg/kg and 200 mg/kg PDTC treatments reduced the serum PGE2 in CAG mice with the decreased PCNA and Ki-67 expression in gastric mucosa. The therapeutic effect of 200 mg/kg PDTC was significantly better than that of 100 mg/kg PDTC. CONCLUSION: PDTC inhibited inflammation and the excessive proliferation of gastric mucosal epithelial cells, thereby exerting a potential therapeutic effect on CAG.


Subject(s)
Gastritis, Atrophic , Animals , Gastritis, Atrophic/drug therapy , Mice , NF-kappa B/metabolism , Pyrrolidines , Signal Transduction , Thiocarbamates/pharmacology , Thiocarbamates/therapeutic use
6.
Eur J Histochem ; 65(1)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33728865

ABSTRACT

Previous studies have suggested that pyrrolidine dithiocarbamate (PDTC), a nuclear factor κB (NF-κB) inhibitor, play a role in deterring nerve injury-induced neuropathic pain (NP) The activation of NF-κB pathway may contribute to spinal microglial activation, CX3CR1 and tumor necrosis factor-alpha (TNF-a) up-regulation. The aim of this study was to clarify whether PDTC could inhibit the development of neuropathic pain via decreasing TNF-a-induced CX3CR1 up-regulation. Sprague-Dawley rats were randomly divided into sham group and NP group. Rats in each group were treated with intrathecal infusion of PDTC (100 or 1000 pmol/d) or saline. The sciatic nerve chronic constriction injury (CCI) model was used to induce NP in rats. Mechanical stimuli and radiant heat were used to evaluate mechanical allodynia and thermal hyperalgesia. Spinal microglial marker OX42 and TNF-a were detected by immunohistochemistry. In vitro BV-2 microglia activation was induced by TNF-a incubation, and the levels of CX3CR1 were assessed by Western blot and reverse transcription-polymerase chain reaction. Pain behavior and immunohistochemistry results showed that intrathecal infusion of PDTC at 100 or 1000 pmol/d prevented the development of mechanical and thermal hyperalgesia, spinal microglial activation and TNF-a expression induced by sciatic nerve CCI in rats. In vitro experiment results showed that PDTC inhibited the TNF-a-induced CX3CR1 up-regulation in BV-2 microglial cells. In conclusion, intrathecal infusion of PDTC could attenuate the pain-related behaviors induced by sciatic nerve CCI through suppressing the spinal microglia activation and TNF-a up-regulation in rats. The NF-κB activation might be responsible for TNF-a-induced CX3CR1 up-regulation in microglia.


Subject(s)
Microglia/drug effects , Neuralgia/drug therapy , Pyrrolidines/therapeutic use , Signal Transduction/drug effects , Thiocarbamates/therapeutic use , Transcription Factor RelA/antagonists & inhibitors , Animals , CX3C Chemokine Receptor 1/metabolism , Male , Mice , Phosphorylation/drug effects , Rats, Sprague-Dawley , Sciatic Nerve/injuries , Spinal Cord/pathology , Transcription Factor RelA/chemistry , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
7.
Cells ; 10(2)2021 02 05.
Article in English | MEDLINE | ID: mdl-33562611

ABSTRACT

Brassinin is a phytochemical derived from Chinese cabbage, a cruciferous vegetable. Brassinin has shown anticancer effects on prostate and colon cancer cells, among others. However, its mechanisms and effects on hepatocellular carcinoma (HCC) have not been elucidated yet. Our results confirmed that brassinin exerted antiproliferative effects by reducing proliferating cell nuclear antigen (PCNA) activity, a proliferation indicator and inducing cell cycle arrest in human HCC (Huh7 and Hep3B) cells. Brassinin also increased mitochondrial Ca2+ levels and depolarized the mitochondrial membrane in both Huh7 and Hep3B cells. Moreover, brassinin generated high amounts of reactive oxygen species (ROS) in both cell lines. The ROS scavenger N-acetyl-L-cysteine (NAC) inhibited this brassinin-induced ROS production. Brassinin also regulated the AKT and mitogen-activated protein kinases (MAPK) signaling pathways in Huh7 and Hep3B cells. Furthermore, co-administering brassinin and pharmacological inhibitors for JNK, ERK1/2 and P38 decreased cell proliferation in both HCC cell lines more than the pharmacological inhibitors alone. Collectively, our results demonstrated that brassinin exerts antiproliferative effects via mitochondrial dysfunction and MAPK pathway regulation on HCC cells.


Subject(s)
Cell Cycle Checkpoints/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/therapeutic use , Indoles/therapeutic use , Liver Neoplasms/drug therapy , Thiocarbamates/therapeutic use , Cell Line, Tumor , Cell Proliferation , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Indoles/pharmacology , Mitochondria , Thiocarbamates/pharmacology
8.
Naunyn Schmiedebergs Arch Pharmacol ; 393(10): 1899-1910, 2020 10.
Article in English | MEDLINE | ID: mdl-32440769

ABSTRACT

We examined the effect of the NFκB inhibitor pyrrolidine-1-carbodithioic acid (PDTC) on inducible nitric oxide synthase (iNOS), matrix metalloproteinase-2 (MMP-2) activity, and oxidative and inflammatory kidney damage in alloxan-induced diabetes. Two weeks after diabetes induction (alloxan-130 mg/kg), control and diabetic rats received PDTC (100 mg/kg) or vehicle for 8 weeks. Body weight, glycemia, urea, and creatinine were measured. Kidney changes were measured in hematoxylin/eosin sections and ED1 by immunohistochemistry. Kidney thiobarbituric acid reactive substances (TBARS), superoxide anion (O2-), and nitrate/nitrite (NOx) levels, and catalase and superoxide dismutase (SOD) activities were analyzed. Also, kidney nox4 and iNOS expression, and NFkB nuclear translocation were measured by western blot, and MMP-2 by zymography. Glycemia and urea increased in alloxan rats, which were not modified by PDTC treatment. However, PDTC attenuated kidney structural alterations and macrophage infiltration in diabetic rats. While diabetes increased both TBARS and O2- levels, PDTC treatment reduced TBARS in diabetic and O2- in control kidneys. A decrease in NOx levels was found in diabetic kidneys, which was prevented by PDTC. Diabetes reduced catalase activity, and PDTC increased catalase and SOD activities in both control and diabetic kidneys. PDTC treatment reduced MMP-2 activity and iNOS and p65 NFκB nuclear expression found increased in diabetic kidneys. Our results show that the NFκB inhibitor PDTC reduces renal damage through reduction of Nox4, iNOS, macrophages, and MMP-2 in the alloxan-induced diabetic model. These findings suggest that PDTC inhibits alloxan kidney damage via antioxidative and anti-inflammatory mechanisms.


Subject(s)
Alloxan/toxicity , Kidney Diseases/drug therapy , Matrix Metalloproteinase 2 , NADPH Oxidase 4/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Pyrrolidines/therapeutic use , Thiocarbamates/therapeutic use , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Kidney Diseases/chemically induced , Kidney Diseases/enzymology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors/therapeutic use , NADPH Oxidase 4/metabolism , Nitric Oxide Synthase Type II/metabolism , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Thiocarbamates/pharmacology
9.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32400857

ABSTRACT

AIM: To investigate the relationship between PI3K/Akt/NF-κB cellular signal pathway and the expression of P-gp and LRP in multidrug resistance (MDR) cell of nasopharyngeal carcinoma. METHOD: The PI3K, p-Akt and NF-κB/p65 as the activity of PI3K/Akt/NF-κB were detected by Western blot. The expressions of LRP and P-gp were detected by Western blot and real-time PCR. RESULT: The RIs of CNE/DDP group to DDP, 5-Fu, VCR, ADR and PTX were 35.04, 18.14, 24.13, 12.00 and 10.18, respectively. The RIs of LY-294002 group were 11.77, 5.83, 3.07, 3.86 and 3.34, and PDTC group were 11.08, 6.55, 7.66, 2.18 and 4.05. The expressions of PI3K, p-Akt and NF-κBp65, LRP and P-gp were increased and mRNA of LRP and P-gp were up-regulated in CNE/DDP. The expression of p-Akt in LY-294002 group was down-regulated. The expression of NF-κB p65 in PDTC group was decreased. The mRNA of LRP and P-gp in LY-294002 group and PDTC group were decreased. CONCLUSION: MDR of nasopharyngeal carcinoma cell can be regulated by activating PI3K/Akt/NF-κB signal pathway and then increase the expression of P-gp and LRP. The MDR of nasopharyngeal carcinoma cell can be reversed by inhibiting PI3K/Akt/NF-κB signal pathway.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Signal Transduction/drug effects , Thiocarbamates/pharmacology , Thiocarbamates/therapeutic use , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism , Vault Ribonucleoprotein Particles/metabolism
10.
Invest New Drugs ; 38(2): 525-532, 2020 04.
Article in English | MEDLINE | ID: mdl-31183632

ABSTRACT

Novel quinoline-dithiocarbamate hybrids were synthesized and designed by the molecular hybridization strategy. All these derivatives were evaluated for their antiproliferative activity against three selected cancer cell lines (MGC-803, HepG-2 and PC-3). Among them, compound 10c displayed the best antiproliferative activity against PC-3 cells with an IC50 value of 0.43 µM. Celluar mechanisms investigated that compound 10c could inhibit the migration against PC-3 cells by regulation the expression levels of E-cadherin and N-cadherin. Compound 10c induced morphological changes of PC-3 cells and regulated apoptosis-related proteins (Bcl-2, Bax and Cleaved-Parp). In addition, compound 10c inhibited tubulin polymerization in vitro with an IC50 value of 4.02 µM. Importantly, compound 10c inhibited the growth of PC-3 cells in vivo with the low toxicity toward mice. These results suggested that compound 10c might be an antitumor agent with potential for treating prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Quinolines/therapeutic use , Thiocarbamates/therapeutic use , Tubulin Modulators/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasms/metabolism , Polymerization , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinolines/chemical synthesis , Quinolines/pharmacology , Thiocarbamates/chemical synthesis , Thiocarbamates/pharmacology , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology
12.
Molecules ; 24(15)2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31374887

ABSTRACT

Dithiocarbamates represent a class of compounds that were evaluated in different biomedical applications because of their chemical versatility. For this reason, several pharmacological activities have already been attributed to these compounds, such as antiparasitic, antiviral, antifungal activities, among others. Therefore, compounds that are based on dithiocarbamates have been evaluated in different in vivo and in vitro models as potential new antimicrobials. Thus, the purpose of this review is to present the possibilities of using dithiocarbamate compounds as potential new antitrypanosomatids-drugs, which could be used for the pharmacological control of Chagas disease, leishmaniasis, and African trypanosomiasis.


Subject(s)
Antiparasitic Agents/therapeutic use , Leishmaniasis/drug therapy , Thiocarbamates/therapeutic use , Trypanosoma/drug effects , Animals , Antiparasitic Agents/chemistry , Chagas Disease/drug therapy , Chagas Disease/parasitology , Humans , Leishmaniasis/parasitology , Thiocarbamates/chemistry , Trypanosoma/pathogenicity , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology
13.
Eur Rev Med Pharmacol Sci ; 23(14): 6264-6271, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31364129

ABSTRACT

OBJECTIVE: To explore the influences of toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB) pathway on the memory function and inflammatory factors in rats with cerebral small vessel disease (CSVD). MATERIALS AND METHODS: CSVD model in rats was established. Expressions of TLR4/NF-κB-related proteins and inflammatory factors were detected. CSVD rats were treated with the TLR4/NF-κB pathway agonist and inhibitor to evaluate the regulatory effect of TLR4/NF-κB pathway on the expressions of TLR4, NF-κB p50 and NF-κB p65. Moreover, their influences on the cerebral edema, memory function and expressions of inflammatory factors [interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α)] in CSVD rats were also analyzed. RESULTS: In model group, the mRNA and protein expressions of TLR4 and NF-κB-related proteins in rat hippocampus were significantly higher than those in sham group (p<0.01), and the expressions of IL-1ß and TNF-αsignificantly increased (p<0.05). The agonist lipopolysaccharide (LPS) significantly increased the proportion of TLR4-positive cells (p<0.01) and protein expression of TLR4 (p<0.01). The inhibitor CLI-095 obviously reduced the proportion of TLR4-positive cells and TLR4 expression (p<0.05). Pyrrolidine dithiocarbamate (PDTC) remarkably reduced the expressions of NF-κB p50 and NF-κB p65 in model group (p<0.05). LPS promoted cerebral edema, leading to memory dysfunction and enhanced inflammatory response in rats of model group. The inhibitor CLI-095+PDTC significantly reduced cerebral edema, lowered memory impairment and relieved inflammatory response in CSVD rats (p<0.05). CONCLUSIONS: The inhibitor of the TLR4/NF-κB signaling pathway can restore memory function and reduce inflammatory response in CSVD rats.


Subject(s)
Cerebral Small Vessel Diseases/immunology , Cerebral Small Vessel Diseases/psychology , Lipopolysaccharides/adverse effects , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cerebral Small Vessel Diseases/chemically induced , Cerebral Small Vessel Diseases/drug therapy , Disease Models, Animal , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , NF-kappa B/genetics , Proline/analogs & derivatives , Proline/pharmacology , Proline/therapeutic use , Rats , Signal Transduction/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thiocarbamates/pharmacology , Thiocarbamates/therapeutic use , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
14.
Balkan Med J ; 36(4): 245-250, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31140237

ABSTRACT

Background: Nuclear factor-kB is highly activated in cardiovascular disorders. However, few articles have targeted at the role of nuclear factor-kB inhibitor in heart failure. Aims: To evaluate the effects of nuclear factor-kB inhibitor pyrrolidine dithiocarbamate on cardiocyte apoptosis and cardiac function in a rat heart failure model. Study Design: Animal experiment. Methods: A stable and reproducible rat heart failure model (n=64) was prepared by injecting homologous microthrombotic particles into the left ventricle of Sprague­Dawley rats while obstructing the ascending aorta to produce coronary microembolization. Rats with heart failure were randomized into untreated (HFu) and pyrrolidine dithiocarbamate-treated (HFp) groups; the latter received an intraperitoneal injection of pyrrolidine dithiocarbamate (100 mg/kg/day) 1 h prior to surgery as well as on postoperative days 1-7. The sham group comprised 32 Sprague­Dawley rats. Eight rats from each group were sacrificed on days 1, 3, 7, and 14 postoperatively. Masson's trichrome staining was used to determine the micro-fibrotic area to indicate the severity of myocardial loss. Terminal transferase uridine triphosphate nick end labeling staining was used to detect apoptotic cardiomyocytes. Echocardiography and hemodynamics were performed to evaluate left ventricular function. Results: Rats with heart failure exhibited pathological changes evidenced by patchy myocardial fibrosis, remarkably elevated severity of myocardial loss, and persistently reduced left ventricular function. At the end of the study, compared with the HFu group, myocardial infarct size was reduced by 28% (p=0.001), cardiocyte apoptosis was suppressed (7.17%±1.47% vs 2.83%±0.75%, p<0.001), cardiac function parameters such as left ventricular ejection fraction (80%±4% vs 61%±6%), left ventricular + dP/dt max (4828±289 vs 2918±76 mmHg.s−1), left ventricular - dP/dt max (4398±269 vs 2481±365 mmHg.s−1), and left ventricular systolic pressure (126±13 vs 100±10 mmHg) were significantly increased, and left ventricular end-diastolic pressure was reduced (18±2 vs 13±1 mmHg) (p<0.001, for all) in the HFu group. Conclusion: Our rat model can adequately mimic heart failure via coronary vessel embolization. Moreover, pyrrolidine dithiocarbamate treatment can reduce cardiocyte apoptosis and improve cardiac function, which may be beneficial for patients with heart failure secondary to myocardial infarction.


Subject(s)
Apoptosis , Heart Failure , NF-kappa B , Pyrrolidines , Thiocarbamates , Animals , Rats/genetics , Rats/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Disease Models, Animal , Embolism , Heart Failure/drug therapy , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics/drug effects , Hemodynamics/physiology , NF-kappa B/analysis , NF-kappa B/drug effects , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Rats, Sprague-Dawley , Thiocarbamates/metabolism , Thiocarbamates/pharmacology , Thiocarbamates/therapeutic use
15.
Oxid Med Cell Longev ; 2018: 4950705, 2018.
Article in English | MEDLINE | ID: mdl-29765497

ABSTRACT

Diversified biological activities of dithiocarbamates have attracted widespread attention; improving their feature or exploring their potent action of mechanism is a hot topic in medicinal research. Herein, we presented a study on synthesis and investigation of a novel dithiocarbamate, DpdtbA (di-2-pyridylhydrazone dithiocarbamate butyric acid ester), on antitumor activity. The growth inhibition assay revealed that DpdtbA had important antitumor activity for gastric cancer (GC) cell lines (IC50 = 4.2 ± 0.52 µM for SGC-7901, 3.80 ± 0.40 µM for MGC-803). The next study indicated that growth inhibition is involved in ROS generation in mechanism; accordingly, the changes in mitochondrial membrane permeability, apoptotic genes, cytochrome c, bax, and bcl-2 were observed, implying that the growth inhibition of DpdtbA is involved in ROS-mediated apoptosis. On the other hand, the upregulated p53 upon DpdtbA treatment implied that p53 could also mediate the apoptosis. Yet the excess generation of ROS induced by DpdtbA led to cathepsin D translocation and increase of autophagic vacuoles and LC3-II, demonstrating that autophagy was also a contributor to growth inhibition. Further investigation showed that DpdtbA could induce cell cycle arrest at the G1 phase. This clearly indicated the growth inhibition of DpdtbA was via triggering ROS formation and evoking p53 response, consequently leading to alteration in gene expressions that are related to cell survival.


Subject(s)
Butyrates/metabolism , Hydrazones/therapeutic use , Reactive Oxygen Species/metabolism , Thiocarbamates/therapeutic use , Apoptosis , Autophagy , Cell Proliferation , Humans , Hydrazones/pharmacology , Signal Transduction , Thiocarbamates/pharmacology
16.
Oxid Med Cell Longev ; 2018: 1324173, 2018.
Article in English | MEDLINE | ID: mdl-30595795

ABSTRACT

Oxidative stress is closely linked to the toxic responses of various cell types in normal and pathophysiological conditions. Deoxynivalenol (DON), an inducer of stress responses in the ribosome and the endoplasmic reticulum (ER), causes mitochondrial dysfunction and mitochondria-dependent apoptosis through oxidative stress in humans and animals. The NF-κB pathway, which is closely linked to oxidative stress, is hypothesized to be a critical signaling pathway for DON-induced toxicity and is a potential target for intervention. The present study was conducted to explore the protective effects of pyrrolidine dithiocarbamate (PDTC) from the toxic effects of DON in rat anterior pituitary GH3 cells. Our results showed that DON activated the NF-κB transcription factors and induced cellular oxidative stress, mitochondrial dysfunction, and apoptosis. Morphological studies using transmission electron microscopy (TEM) and cell apoptosis analyses suggested that PDTC prevented DON-induced mitochondrial dysfunction and apoptosis, probably by preventing the DON-induced translocation of NF-κB p65 into the nucleus, and by inhibiting DON-induced iNOS expression. This led to the blocking of the NF-κB pathway and inhibition of iNOS activity.


Subject(s)
Mitochondria/metabolism , NF-kappa B/metabolism , Pyrrolidines/therapeutic use , Thiocarbamates/therapeutic use , Antioxidants , Apoptosis , Humans , Oxidative Stress , Pyrrolidines/pharmacology , Signal Transduction , Thiocarbamates/pharmacology
17.
Ann Clin Lab Sci ; 48(6): 743-750, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30610044

ABSTRACT

Bone marrow mesenchymal stromal cells (BMSCs) have positive therapeutic effects on inflammation associated diseases. However, the underlying mechanism is largely unknown. This study was conducted to investigate whether BMSCs could alleviate the inflammation reaction in lipopolysaccaride (LPS)-induced acute kidney injury (septic-AKI) of rats via inhibition of toll-like receptors (TLR4)-nuclear factor-kappa B (NF-κB) signaling pathway. The septic-AKI rat model was established by injecting the 1ml/mg LPS through the femoral vein. Based on this model, rats were subjected to BMSC transplantation, PDTC (a kind of NF-κB inhibitor) administration alone, and combined treatment of the first two together. Results showed that LPS treatment caused the increases of the concentration of blood urea nitrogen (BUN) and serum creatinine (SCr), accompanied by tissue injury and the up-regulation of TLR4 and NF-κB, that was its key downstream signaling molecule, in both mRNA and protein level. Notably, it has been found that BMSCs transplantation significantly reversed the already upregulated concentration of BUN and SCr, dramatically attenuated the event of the tissue injury, and prominently reduced mortality after AKI. These were paralleled by down-regulation of the level of TLR4 and NF-κB. These effects of BMSCs transplantation were similar to those of PDTC treatment. Importantly, the effects in the combination therapy of BMSCs transplantation and PDTC group were much stronger than those of either BMSCs or PDTC used alone. These findings suggest that BMSCs transplantation contributes to therapeutic effects in LPS-induced AKI rat model, and that the most obvious effects occurred in the combined treatment group, with BMSCs and PDTC together, which was tightly associated with inhibition of the TLR4-NF-κB signaling pathway.


Subject(s)
Acute Kidney Injury/complications , Inflammation/etiology , Inflammation/surgery , Mesenchymal Stem Cell Transplantation/methods , Signal Transduction/physiology , Acute Kidney Injury/chemically induced , Analysis of Variance , Animals , Antioxidants/therapeutic use , Blood Urea Nitrogen , Disease Models, Animal , Female , Gene Expression Regulation/physiology , Lipopolysaccharides/toxicity , Mesenchymal Stem Cells/physiology , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Proline/analogs & derivatives , Proline/therapeutic use , RNA, Messenger/metabolism , Rats , Thiocarbamates/therapeutic use , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Neurosci Bull ; 34(1): 64-73, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28612319

ABSTRACT

Tetanic stimulation of the sciatic nerve (TSS) triggers long-term potentiation in the dorsal horn of the spinal cord and long-lasting pain hypersensitivity. CX3CL1-CX3CR1 signaling is an important pathway in neuronal-microglial activation. Nuclear factor κB (NF-κB) is a key signal transduction molecule that regulates neuroinflammation and neuropathic pain. Here, we set out to determine whether and how NF-κB and CX3CR1 are involved in the mechanism underlying the pathological changes induced by TSS. After unilateral TSS, significant bilateral mechanical allodynia was induced, as assessed by the von Frey test. The expression of phosphorylated NF-κB (pNF-κB) and CX3CR1 was significantly up-regulated in the bilateral dorsal horn. Immunofluorescence staining demonstrated that pNF-κB and NeuN co-existed, implying that the NF-κB pathway is predominantly activated in neurons following TSS. Administration of either the NF-κB inhibitor ammonium pyrrolidine dithiocarbamate or a CX3CR1-neutralizing antibody blocked the development and maintenance of neuropathic pain. In addition, blockade of NF-κB down-regulated the expression of CX3CL1-CX3CR1 signaling, and conversely the CX3CR1-neutralizing antibody also down-regulated pNF-κB. These findings suggest an involvement of NF-κB and the CX3CR1 signaling network in the development and maintenance of TSS-induced mechanical allodynia. Our work suggests the potential clinical application of NF-κB inhibitors or CX3CR1-neutralizing antibodies in treating pathological pain.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Hyperalgesia/metabolism , Sciatic Nerve/physiology , Signal Transduction/physiology , Animals , Antibodies/therapeutic use , Antioxidants/therapeutic use , CX3C Chemokine Receptor 1/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/etiology , Nerve Tissue Proteins/metabolism , Pain Threshold/physiology , Physical Stimulation/adverse effects , Proline/analogs & derivatives , Proline/therapeutic use , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/metabolism , Thiocarbamates/therapeutic use , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Chin Med J (Engl) ; 130(18): 2163-2169, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28836571

ABSTRACT

BACKGROUND:: Acute kidney injury (AKI) is the most common and life-threatening systemic complication of rhabdomyolysis. Inflammation plays an important role in the development of rhabdomyolysis-induced AKI. This study aimed to investigate the kidney model of AKI caused by rhabdomyolysis to verify the role of macrophage Toll-like receptor 4/nuclear factor-kappa B (TLR4/NF-κB) signaling pathway. METHODS:: C57BL/6 mice were injected with a 50% glycerin solution at bilateral back limbs to induce rhabdomyolysis, and CLI-095 or pyrrolidine dithiocarbamate (PDTC) was intraperitoneally injected at 0.5 h before molding. Serum creatinine levels, creatine kinase, the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6, and hematoxylin and eosin stainings of kidney tissues were tested. The infiltration of macrophage, mRNA levels, and protein expression of TLR4 and NF-κB were investigated by immunofluorescence double-staining techniques, reverse transcriptase-quantitative polymerase chain reaction, and Western blotting, respectively. In vitro, macrophage RAW264.7 was stimulated by ferrous myoglobin; the cytokines, TLR4 and NF-κB expressions were also detected. RESULTS:: In an in vivo study, using CLI-095 or PDTC to block TLR4/NF-κB, functional and histologic results showed that the inhibition of TLR4 or NF-κB alleviated glycerol-induced renal damages (P < 0.01). CLI-095 or PDTC administration suppressed proinflammatory cytokine (TNF-α, IL-6, and IL-1ß) production and macrophage infiltration into the kidney (P < 0.01). Moreover, in an in vitro study, CLI-095 or PDTC suppressed myoglobin-induced expression of TLR4, NF-κB, and proinflammatory cytokine levels in macrophage RAW264.7 cells (P < 0.01). CONCLUSION:: The pharmacological inhibition of TLR4/NF-κB exhibited protective effects on rhabdomyolysis-induced AKI by the regulation of proinflammatory cytokine production and macrophage infiltration.


Subject(s)
Acute Kidney Injury/drug therapy , NF-kappa B/metabolism , Rhabdomyolysis/complications , Toll-Like Receptor 4/metabolism , Acute Kidney Injury/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Kidney/drug effects , Kidney/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Proline/analogs & derivatives , Proline/pharmacology , Proline/therapeutic use , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , RAW 264.7 Cells , Rhabdomyolysis/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thiocarbamates/pharmacology , Thiocarbamates/therapeutic use , Toll-Like Receptor 4/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
20.
Undersea Hyperb Med ; 44(2): 109-119, 2017.
Article in English | MEDLINE | ID: mdl-28777901

ABSTRACT

BACKGROUND: Decompression sickness (DCS) induced by fast buoyancy ascent escape (FBAE) is a special DCS, characterized with cardiopulmonary injuries. Serum metabonomics of this type of DCS has not yet been studied. We proposed a metabonomics approach for assessing serum metabonomics changes and evaluating the preventive effect of pyrrolidine dithiocarbamic acid (PDTC) in FBAE-induced DCS rats. METHODS: Sixty-five (65) rats were divided into three groups, including the Control, DCS and PDTC groups. After receiving physiological saline or PDTC pretreatment, rats in the DCS and PDTC groups received the same protocol of simulated FBAE. Following this, a metabonomics approach - combined with pattern recognition methods including PCA and PLS-DA - was used to characterize the global serum metabolic profile on survival rats (five rats per group) associated with abnormal FBAE-induced DCS. As the VIP-value threshold cutoff of the metabolites was set to 2, metabolites above this threshold were filtered out as potential target biomarkers. RESULTS: Sixteen (16) distinct potential biomarkers in rat plasma were identified. PDTC significantly lowered DSC mortality from 60% to 10%, and alleviated ultrastructural alteration of the left ventricular apex compared to the DCS group. It was found that abnormal FBAE-induced DCS was closely related to disturbed fatty acid metabolism, glycerophospholipid metabolism, sterol lipid metabolism, and bile acid metabolism. With the presented metabonomic method, we systematically analyzed the protective effects of PDTC. CONCLUSION: The results demonstrated that PDTC administration could provide satisfactory effects on abnormal FBAE-induced DCS through partially regulating the perturbed metabolic pathways.


Subject(s)
Antioxidants/therapeutic use , Biomarkers/blood , Decompression Sickness/blood , Decompression Sickness/prevention & control , Metabolomics , Pyrrolidines/therapeutic use , Thiocarbamates/therapeutic use , Animals , Bile Acids and Salts/blood , Decompression Sickness/etiology , Decompression Sickness/mortality , Disease Models, Animal , Fatty Acids/blood , Glycerophospholipids/blood , Heart Ventricles/ultrastructure , Male , Microscopy, Electron, Transmission , Rats , Rats, Sprague-Dawley , Sterols/blood , Submarine Medicine
SELECTION OF CITATIONS
SEARCH DETAIL
...