Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Cancer ; 15(6): 1551-1567, 2024.
Article in English | MEDLINE | ID: mdl-38370386

ABSTRACT

Background: Recent researches have demonstrated that cuproptosis, a copper-dependent cell death mechanism, is related to tumorigenesis, progression, clinical prognosis, tumor microenvironment, and drug sensitivity. Nevertheless, the function and impact of cuproptosis in cholangiocarcinoma (CCA), remain elusive. Methods: Utilizing data obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA-CHOL) datasets, we conducted subgroup typing of CCA according to cuproptosis-related genes (CRGs) and explored functional differences and prognostic value between groups. A CRG score was established considering clinical prognosis and gene expression. Furthermore, differences in the immune microenvironment, response to immunotherapy, metabolic patterns, and cancer progression characteristics between high- and low-risk groups were examined on the basis of these scores. In vitro experiments validated the function of the key gene glycine cleavage system protein H (GCSH) in cellular and tissues, respectively. Results: Prognostic models established on the basis of subgroup genetic differences achieved satisfactory results in validation. Metabolic-related gene expression levels and tumor microenvironment distribution were significantly different between the high and low CRG groups. GCSH was revealed as the singular prognostic CRG in CCA (HR =6.04; 95% CI: 1.15-31.80). Moreover, inhibition of the cupcoptosis key gene GCSH attenuated the malignant ability of CCA cell lines in vitro, including cell proliferation, migration and invasion, and this function of GCSH may be achieved via JAK-STAT signaling in CCA. Conclusion: The CRG scoring system accurately predicts prognosis and opens up new possibilities for cuproptosis-related therapy for CCA. The cuproptosis key gene GCSH has been preliminarily confirmed as a reliable therapeutic target or prognostic marker for CCA patients.

2.
Front Med (Lausanne) ; 6: 276, 2019.
Article in English | MEDLINE | ID: mdl-31921864

ABSTRACT

Background: Ischemia-reperfusion injury (IRI) has been considered an inevitable event in organ transplantation since the first successful kidney transplant was performed in 1954. To avoid IRI, we have established a novel procedure called ischemia-free organ transplantation. Here, we describe the first case of ischemia-free kidney transplantation (IFKT). Materials and Methods: The kidney graft was donated by a 19-year-old brain-dead donor. The recipient was a 47-year-old man with end-stage diabetic nephropathy. The graft was procured, preserved, and implanted without cessation of blood supply using normothermic machine perfusion. Results: The graft appearance, perfusion flow, and urine production suggested that the kidney was functioning well-during the whole procedure. The creatinine dropped rapidly to normal range within 3 days post-transplantation. The levels of serum renal injury markers were low post-transplantation. No rejection or vascular or infectious complications occurred. The patient had an uneventful recovery. Conclusion: This paper marks the first case of IFKT in humans. This innovation may offer a unique solution to optimizing transplant outcomes in kidney transplantation.

3.
Int J Mol Med ; 42(3): 1765, 2018 09.
Article in English | MEDLINE | ID: mdl-29845220

ABSTRACT

Subsequently to the publication of this article, the authors have realized that the address affiliation for the corresponding author, Chengheng Hu, and the authors Longyun Peng and Xinxue Liao appeared incorrectly. These authors' affiliation information should have appeared as follows (the corrected address affiliation is featured in bold): XIAO KE1,2*, JINGFU CHEN3*, LONGYUN PENG4, WEI ZHANG5, YIYING YANG5, XINXUE LIAO4, LIQIU MO6, RUIXIAN GUO7, JIANQIANG FENG6, CHENGHENG HU4 and RUQIONG NIE2 1Department of Cardiology, Shenzhen Sun Yat­sen Cardiovascular Hospital, Shenzhen; 2Department of Cardiology, Sun Yat­sen Memorial Hospital, Sun Yat­sen University, Guangzhou, Guangdong; 3Department of Cardiovascular Medicine and Dongguan Cardiovascular Institute, The Third People's Hospital of Dongguan City, Dongguan; 4Department of Cardiology and Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat­sen University; 5Department of Cardiovasology and Cardiac Care Unit (CCU), Huangpu Division of The First Affiliated Hospital, Sun Yat­sen University; 6Department of Anesthesiology, Huangpu Division of The First Affiliated Hospital, Sun Yat­sen University; 7Department of Physiology, Zhongshan School of Medicine, Sun Yat­sen University, Guangzhou, Guangdong, P.R. China *Contributed equally In addition, the address for correspondence in the correspondence box should have appeared as follows: Correspondence to: Professor Chengheng Hu, Department of Cardiology and Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat­sen University, Guangdong, 58 Zhongshan 2rd Road, Guangzhou 510080, P.R. China E­mail: huchengheng138@163.com The authors regret this error in the affiliations, and apologize for any inconvenience caused. [the original article was published in the International Journal of Molecular Medicine 39: 1001­1010, 2017; DOI: 10.3892/ijmm.2017.2891].

4.
Int J Mol Med ; 40(1): 201-208, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28560421

ABSTRACT

Recently, a novel mechanism known as 'programmed necrosis' or necroptosis has been shown to be another important mechanism of cell death in the heart. In this study, we investigated the role of necroptosis in high glucose (HG)-induced injury and inflammation, as well as the underlying mechanisms. In particular, we focused on the interaction between necroptosis and reactive oxygen species (ROS) in H9c2 cardiac cells. Our results demonstrated that the exposure of H9c2 cardiac cells to 35 mM glucose (HG) markedly enhanced the expression level of receptor-interacting protein 3 (RIP3), a kinase which promotes necroptosis. Importantly, co-treatment of the cells with 100 µM necrostatin-1 (a specific inhibitor of necroptosis) and HG for 24 h attenuated not only the increased expression level of RIP3, but also the HG-induced injury and inflammation, as evidenced by an increase in cell viability, a decrease in ROS generation, the attenuation of the dissipation of mitochondrial membrane potential and a decrese in the secretion levels of inflammatory cytokines, i.e., interleukin (IL)-1ß and tumor necrosis factor (TNF)-α. Furthermore, treatment of the cells with 1 mM N-acetyl­L­cysteine (a scavenger of ROS) for 60 min prior to exposure to HG significantly reduced the HG-induced increase in the RIP3 expression level, as well as the injury and inflammatory response described above. Taken together, the findings of this study clearly demonstrate a novel damage mechanism involving the positive interaction between necroptosis and ROS attributing to HG-induced injury and inflammation in H9c2 cardiac cells.


Subject(s)
Glucose/pharmacology , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Cell Line , Gene Expression Regulation/drug effects , Inflammation/metabolism , Inflammation/pathology , Myocytes, Cardiac/pathology , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
5.
Int J Mol Med ; 39(4): 1001-1010, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28204829

ABSTRACT

It has been reported that exogenous hydrogen sulfide (H2S) protects against high glucose (HG)-induced cardiac injury and has a modulatory effect on heat shock protein (HSP) and Akt, which play a cardioprotective role. In this study, we examined whether the HSP90/Akt pathway contributes to the protective effects of exogenous H2S against HG-induced injury to H9c2 cardiac cells. Our results revealed that the exposure of H9c2 cardiac cells to 35 mM glucose (HG) for 1 to 24 h decreased the expression of HSP90 and markedly reduced the expression level of phosphorylated (p)-Akt in a time-dependent manner. Co-exposure of the cells to HG and geldanamycin (GA; an inhibitor of HSP90) aggravated the inhibition of the p-Akt expression level by HG. Of note, treatment of the cells with 400 µM NaHS (a donor of H2S) for 30 min prior to exposure to HG significantly attenuated the HG-induced decrease in the expression levels of both HSP90 and p-Akt, along with inhibition of HG-induced cell injury, as indicated by the increase in cell viability and superoxide dismutase (SOD) activity, and by a decrease in the number of apoptotic cells, reactive oxygen species (ROS) generation, as well as by the decreased dissipation of mitochondrial membrance potential (MMP). Importantly, treatment of the cells with GA or LY294002 (an inhibitor of Akt) prior to exposure to NaHS and HG considerably blocked the cardioprotective effects of NaHS against the HG-induced injury mentioned above. On the whole, the findings of this study demonstrate that the inhibition of the HSP90/Akt pathway may be an important mechanism responsible for HG-induced cardiomyocyte injury. We also provide novel evidence that exogenous H2S protects H9c2 cells against HG-induced injury by activating the HSP90/Akt pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Glucose/adverse effects , HSP90 Heat-Shock Proteins/metabolism , Hydrogen Sulfide/pharmacology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Cell Line , Cell Survival/drug effects , Chromones/pharmacology , Glucose/pharmacology , Morpholines/pharmacology , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Reactive Oxygen Species/metabolism
6.
Mol Med Rep ; 14(4): 3237-42, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27498747

ABSTRACT

Tolerance to hypoxia can be induced by reducing oxygen consumption. Dexmedetomidine (DEX) decreases locomotor activity and induces bradycardia and hypothermia in mice. The present study examined the hypothesis that DEX improves hypoxia tolerance in mice. Adult mice received an intraperitoneal injection of 1, 5, 10, 20, 40, 80, 160 or 320 µg/kg DEX, 20 mg/kg propranolol or saline. Acute hypoxic conditions were induced by placing the mice in a limited enclosed container with soda lime. Core body temperature (CBT) and heart rate (HR) were measured prior to and 30 min after drug administration. Survival time was monitored in the sealed container. Survival times (mean ± standard deviation) of mice in the saline, 1, 5, 10, 20, 40, 80, 160 and 320 µg/kg DEX, and the 20 mg/kg propranolol groups were 22.4±1.1, 23.4±1.1, 26.0±0.9, 36.9±5.2, 42.4±2.9, 43.2±2.3, 58.2±4.2, 80.5±4.0, 79.2±6.0, and 38.2±2.8 min, respectively. Pretreatment with propranolol and 10, 20, 40, 80, 160 or 320 µg/kg DEX, but not 1 or 5 µg/kg, significantly prolonged survival time compared with saline­injected mice (P<0.05 or P<0.01). CBT and HR decreased in a similar manner. The correlation coefficients between survival time and CBT, and survival time and HR were ­0.802 and ­0.726, respectively. Thus, DEX dose­dependently enhances hypoxia tolerance in mice. In conclusion, it is suggested that DEX may be used in clinical practice as a novel protective agent for organs and tissues during hypoxic injury.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/therapeutic use , Dexmedetomidine/therapeutic use , Heart Rate/drug effects , Hypnotics and Sedatives/therapeutic use , Hypoxia/drug therapy , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Animals , Dexmedetomidine/administration & dosage , Heart/drug effects , Heart/physiopathology , Hypnotics and Sedatives/administration & dosage , Hypoxia/physiopathology , Male , Mice
7.
Int J Mol Med ; 37(3): 763-72, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26820501

ABSTRACT

Hyperglycemia, as well as diabetes mellitus, has been shown to impair ATP-sensitive K+ (KATP) channels in human vascular smooth muscle cells. Hydrogen sulfide (H2S) is also known to be an opener of KATP channels. We previously demonstrated the cardioprotective effects exerted by H2S against high-glucose (HG, 35 mM glucose)-induced injury in H9c2 cardiac cells. As such, we hypothesized that KATP channels play a role in the cardioprotective effects of H2S against HG-induced injury. In this study, to examine this hypothesis, H9c2 cardiac cells were treated with HG for 24 h to establish a model of HG-induced insults. Our findings revealed that treatment of the cells with HG markedly decreased the expression level of KATP channels. However, the decreased expression of KATP channels was reversed by the treatment of the cells with 400 µM sodium hydrogen sulfide (NaHS, a donor of H2S) for 30 min prior to exposure to HG. Additionally, the HG-induced cardiomyocyte injuries, including cytotoxicity, apoptosis, oxidative stress and mitochondrial damage, were ameliorated by treatment with NaHS or 100 µM diazoxide (a mitochondrial KATP channel opener) or 50 µM pinacidil (a non-selective KATP channel opener) for 30 min prior to exposure to HG, as indicated by an increase in cell viability, as well as a decrease in the number of apoptotic cells, the expression of cleaved caspase-3, the generation of reactive oxygen species (ROS) and the dissipation of mitochondrial membrane potential (MMP). Notably, treatment of the H9c2 cardiac cells with 100 µM 5-hydroxydecanoic acid (5-HD, a mitochondrial KATP channel blocker) or 1 mM glibenclamide (Gli, a non-selective KATP channel blocker) for 30 min prior to treatment with NaHS and exposure to HG significantly attenuated the above-mentioned cardioprotective effects exerted by NaHS. Notably, treatment of the cells with 500 µM N-acetyl­L­cysteine (NAC, a scavenger of ROS) for 60 min prior to exposure to HG markedly reduced the HG-induced inhibitory effect on the expression of KATP channels. Taken together, our results suggest that KATP channels play an important role in the cardioprotective effects of exogenous H2S against HG-induced injury. This study also provides novel data demonstraring that there is an antagonistic interaction between ROS and KATP channels in HG-exposed H9c2 cardiac cells.


Subject(s)
Apoptosis/physiology , Cell Survival/physiology , Decanoic Acids/pharmacology , Glucose/pharmacology , Hydrogen Sulfide/pharmacology , Hydroxy Acids/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Cell Line , Membrane Potential, Mitochondrial/drug effects , Potassium Channels , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
8.
Paediatr Anaesth ; 26(3): 239-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26612740

ABSTRACT

BACKGROUND: Dexmedetomidine decreases cardiac complications in adults undergoing cardiovascular surgery. This systematic review assessed whether perioperative dexmedetomidine improves congenital heart disease (CHD) surgery outcomes in children. METHODS: The PubMed, Embase, and Cochrane Library databases were searched for randomized controlled trials (RCTs) or observational studies that were published until 16 April 2015 and compared dexmedetomidine with placebo or an alternative anesthetic agent during pediatric CHD surgery. The assessed outcomes included hemodynamics, ventilation length, intensive care unit (ICU) and hospital stays, blood glucose and serum cortisol levels, postoperative analgesia requirements, and postoperative delirium. RESULTS: Five RCTs and nine observational studies involving 2229 patients were included. In pooled analyses, dexmedetomidine was associated with shorter length of mechanical ventilation (mean difference: -93.36, 95% CI: -137.45, -49.27), lower postoperative fentanyl (mean difference: -24.11, 95% CI: -36.98, -11.24) and morphine (mean difference: -0.07, 95% CI: -0.14, 0.00) requirements, reduced stress response (i.e., lower blood glucose and serum cortisol levels), and lower risk of delirium (OR: 0.39, 95% CI: 0.21, 0.74). The hemodynamics of dexmedetomidine-treated patients appeared more stable, but there were no significant differences in the ICU or hospital stay durations. Dexmedetomidine may increase the bradycardia and hypotension risk (OR: 3.14, 95% CI: 1.47, 6.69). CONCLUSIONS: Current evidence indicates that dexmedetomidine improves outcomes in children undergoing CHD surgery. However, this finding largely relies on data from observational studies; high-quality RCTs are warranted because of the potential for subject selection bias.


Subject(s)
Dexmedetomidine , Heart Defects, Congenital/surgery , Hypnotics and Sedatives , Adolescent , Adult , Blood Glucose/drug effects , Child , Child, Preschool , Delirium , Hemodynamics/drug effects , Humans , Hydrocortisone/blood , Infant , Length of Stay/statistics & numerical data , Postoperative Complications , Randomized Controlled Trials as Topic , Respiration, Artificial , Treatment Outcome , Young Adult
9.
Cell Mol Neurobiol ; 36(7): 1179-88, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26683659

ABSTRACT

Dexmedetomidine has been reported to provide neuroprotection against hypoxia-induced damage. However, the underlying mechanisms remain unclear. We examined whether dexmedetomidine's neuroprotective effects were mediated by the NF-κB/COX-2 pathways. Adult male C57BL/6 mice were subjected to a 30-min hypoxic treatment followed by recovery to normal conditions. They received dexmedetomidine (16 or 160 µg/kg) or 25 mg/kg atipamezole, an α2-adrenoreceptor antagonist, intraperitoneally before exposure to hypoxia. The whole brain was harvested 6, 18, or 36 h after the hypoxia to determine the histopathological outcome and cleaved caspase-3, Bax/Bcl, NF-κB, and COX-2 levels. Hypoxia treatment induced significant neurotoxicity, including destruction of the tissue structure and upregulation of the protein levels of caspase-3, the ratio of Bax/Bcl-2, NF-κB, and COX-2. Dexmedetomidine pretreatment effectively improved histological outcome and restored levels of caspase-3, the Bax/Bcl-2 ratio, NF-κB, and COX-2. Atipamezole reversed the neuroprotection induced by dexmedetomidine. Neuroprotection was achieved by PDTC and NS-398, inhibitors of NF-κB and COX-2, respectively. Dexmedetomidine use before hypoxia provides neuroprotection. Inhibition of NF-κB/COX-2 pathways activation may contribute to the neuroprotection of dexmedetomidine.


Subject(s)
Cyclooxygenase 2/metabolism , Dexmedetomidine/pharmacokinetics , Hypoxia/drug therapy , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Trauma, Nervous System/drug therapy , Animals , Caspase 3/metabolism , Male , Mice, Inbred C57BL , Up-Regulation/drug effects
10.
Int J Mol Med ; 35(1): 177-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25412187

ABSTRACT

Hyperglycemia has been reported to activate the nuclear factor-κB (NF-κB) pathway. We have previously demonstrated that exogenous hydrogen sulfide (H2S) protects cardiomyocytes against high glucose (HG)-induced injury by inhibiting the activity of p38 mitogen-activated protein kinase (MAPK), which can activate the NF-κB pathway and induce interleukin (IL)-1ß production. In the present study, we aimed to investigate the hypothesis that exogenous H2S protects cardiomyocytes against HG-induced injury and inflammation through the inhibition of the NF-κB/IL-1ß pathway. H9c2 cardiac cells were treated with 35 mM glucose (HG) for 24 h to establish a model of HG-induced damage. Our results demonstrated that treatment of the cells with 400 µM sodium hydrogen sulfide (NaHS, a donor of H2S) or 100 µM pyrrolidine dithiocarbamate (PDTC, an inhibitor of NF-κB) for 30 min prior to exposure to HG markedly attenuated the HG-induced increase in the expression levels of the phosphorylated (p)-NF-κB p65 subunit. Notably, pre-treatment of the H9c2 cardiac cells with NaHS or PDTC significantly suppressed the HG-induced injury, including cytotoxicity, apoptosis, oxidative stress and mitochondrial insults, as evidenced by an increase in cell viability, as well as a decrease in the number of apoptotic cells, the expression of cleaved caspase-3, the generation of reactive oxygen species (ROS) and the dissipation of mitochondrial membrane potential (MMP). In addition, pre-treatment of the cells with NaHS or PDTC ameliorated the HG-induced inflammatory response, leading to a decrease in the levels of IL-1ß, IL-6 and tumor necrosis factor-α (TNF-α). Importantly, co-treatment of the H9c2 cells with 20 ng/ml IL-1 receptor antagonist (IL-1Ra) and HG markedly reduced the HG-induced increase in p-NF-κB p65 expression, cytotoxicity, the number of apoptotic cells, as well as the production of TNF-α. In conclusion, the present study presents novel mechanistic evidence that exogenous H2S protects H9c2 cardiac cells against HG-induced inflammation and injury, including cytotoxicity, apoptosis, overproduction of ROS and the dissipation of MMP, by inhibiting the NF-κB/IL-1ß pathway. We also provide new data indicating that the positive interaction between the NF-κB pathway and IL-1ß is critical in HG-induced injury and inflammation in H9c2 cardiac cells.


Subject(s)
Hydrogen Sulfide/pharmacology , Hyperglycemia/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cytokines/metabolism , Enzyme Activation/drug effects , Inflammation Mediators/metabolism , Matrix Metalloproteinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Rats , Reactive Oxygen Species/metabolism
11.
Am J Dermatopathol ; 37(2): 167-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24335519

ABSTRACT

Eccrine nevus shows increase in number or size of eccrine glands, whereas hair follicle nevus is composed of densely packed normal vellus hairs, and eccrine-pilar angiomatous nevus reveals increase of eccrine, pilar, and angiomatous structures. No case with increased number of both eccrine glands and hair follicles only in the dermis has been previously reported. A 10-month-old girl presented with cutaneous hamartoma with overlying skin hyperpigmentation on her left hypochondrium since 3 months of age, in whom the lesion was completely excised. Histopathology demonstrated evidently increased number of both eccrine glands and hair follicles in the dermis with reactive hyperplasia of collagen fibers. No recurrence occurred after the tumor was completely excised. A term "hybrid eccrine gland and hair follicle hamartoma" is proposed for this unique lesion.


Subject(s)
Eccrine Glands/pathology , Hair Follicle/pathology , Hamartoma/pathology , Neoplasms, Adnexal and Skin Appendage/pathology , Nevus/pathology , Skin Neoplasms/pathology , Biopsy , Eccrine Glands/surgery , Female , Hair Follicle/surgery , Hamartoma/classification , Hamartoma/surgery , Humans , Infant , Neoplasms, Adnexal and Skin Appendage/classification , Neoplasms, Adnexal and Skin Appendage/surgery , Nevus/classification , Nevus/surgery , Predictive Value of Tests , Skin Neoplasms/classification , Skin Neoplasms/surgery , Terminology as Topic
12.
Mol Med Rep ; 10(2): 923-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24841769

ABSTRACT

Leptin, an adipokine synthesized mainly by non­neuronal tissues, has been reported to contribute to the pathogenesis of neuropathic pain. It has been hypothesized that morphine tolerance and neuropathic pain share some common pathological mechanisms. The present study was designed to examine whether spinal leptin is implicated in the development of morphine antinociceptive tolerance, and whether spinal leptin induces the activation of signal transducer and activator of transcription 3 (STAT3) signaling pathway and the NR1 subunit of N­methyl­D­aspartate (NMDA) receptor, in morphine antinociceptive tolerance in rats. The results demonstrated that intrathecal (i.t.) administration of a leptin antagonist (LA) prevented the development of morphine antinociceptive tolerance in rats. Further studies revealed that the levels of the spinal leptin and the leptin receptor (Ob­R) were time­dependently increased following chronic morphine treatment. Mechanistic examination indicated that chronic morphine triggered activation of the STAT3 pathway and an increase in the expression of the NR1 subunit of the NMDA receptor, which was ameliorated by i.t. administration of AG490 [a Janus kinase (JAK)­STAT inhibitor]. The increased activation of STAT3 and the NR1 subunit was markedly attenuated by i.t. treatment with LA. In addition, the spinal administration of AG490 or MK­801 (a non­competitive NMDA receptor inhibitor) blocked the development of morphine antinociceptive tolerance. Taken together, these results have demonstrated, for the first time, to the best of our knowledge, that spinal leptin contributes to the development of morphine antinociceptive tolerance by activating the spinal STAT3­NMDA receptor pathway.


Subject(s)
Analgesics, Opioid/pharmacology , Leptin/metabolism , Morphine/pharmacology , Signal Transduction/drug effects , Spinal Cord/metabolism , Animals , Dizocilpine Maleate/pharmacology , Drug Tolerance , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Leptin/antagonists & inhibitors , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Leptin/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Tyrphostins/pharmacology
13.
CNS Neurosci Ther ; 20(8): 728-36, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24629168

ABSTRACT

AIMS: Connexin 43 (Cx43) has been reported to be involved in neuropathic pain, but whether it contributes to morphine antinociceptive tolerance remains unknown. The present study investigated the role of spinal Cx43 in the development of morphine tolerance and its mechanisms in rats. METHODS: Morphine tolerance was induced by intrathecal (i.t.) administration of morphine (15 µg) daily for seven consecutive days. The analgesia effect was assessed by hot-water tail-flick test. Expression of proteins was detected by Western blot and immunohistochemistry assay. RESULTS: Chronic morphine markedly increased the expression of spinal Cx43. Gap26, a specific Cx43 mimic peptide, attenuated not only morphine antinociceptive tolerance, but also the up-regulation of spinal Cx43 expression, the activation of astrocytes, and N-methyl-D-aspartic acid (NMDA) receptors (NR1 and NR2B subunits), as well as the decreased GLT-1 expression induced by chronic morphine. MK-801, a noncompetitive NMDA receptors antagonist, suppressed the chronic morphine-induced spinal Cx43 up-regulation, astrocytes activation and decline of GLT-1 expression. CONCLUSIONS: The spinal astrocytic Cx43 contributes to the development of morphine antinociceptive tolerance by activating astrocytes and NMDA receptors, and inhibiting GLT-1 expression. We also demonstrate that the role of interaction between the spinal astrocytic Cx43 and neuronal NMDA receptors is important in morphine tolerant rats.


Subject(s)
Analgesics, Opioid/pharmacology , Astrocytes/drug effects , Connexin 43/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Morphine/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/cytology , Animals , Astrocytes/metabolism , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Male , Peptides/pharmacology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects
14.
Cell Physiol Biochem ; 32(6): 1668-80, 2013.
Article in English | MEDLINE | ID: mdl-24356372

ABSTRACT

BACKGROUND/AIM: We have demonstrated that exogenous hydrogen sulfide (H2S) protects H9c2 cardiac cells against the doxorubicin (DOX)-induced injuries by inhibiting p38 mitogen-activated protein kinase (MAPK) pathway and that the p38 MAPK/nuclear factor-κB (NF-κB) pathway is involved in the DOX-induced inflammatory response and cytotoxicity. The present study attempts to test the hypothesis that exogenous H2S might protect cardiomyocytes against the DOX-induced inflammation and cytotoxicity through inhibiting p38 MAPK/NF-κB pathway. METHODS: H9c2 cardiac cells were exposed to 5µM DOX for 24 h to establish a model of DOX cardiotoxicity. The cells were pretreated with NaHS( a donor of H2S) or other drugs before exposure to DOX. Cell viability was analyzed by cell counter kit 8 ( CCK-8), The expression of NF-κB p65 and inducible nitric oxide synthase (iNOS) was detected by Western blot assay. The levels of interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α) were tested by enzyme-linked immunosorbent assay (ELISA). RESULTS: Our findings demonstrated that pretreatment of H9c2 cardiac cells with NaHS for 30 min before exposure to DOX markedly ameliorated the DOX-induced phosphorylation and nuclear translocation of NF-κB p65 subunit. Importantly, the pretreatment with NaHS significantly attenuated the p38 MAPK/NF-κB pathway-mediated inflammatory responses induced by DOX, as evidenced by decreases in the levels of IL-1ß, IL-6 and TNF-α. In addition, application of NaHS or IL-1ß receptor antagonist (IL-1Ra) or PDTC (an inhibitor of NF-κB) attenuated the DOX-induced expression of iNOS and production of nitric oxide (NO), respectively. Furthermore, IL-1Ra also dramatically reduced the DOX-induced cytotoxicity and phosphorylation of NF-κB p65. The pretreatment of H9c2 cells with N-acetyl-L-cysteine (NAC), a scavenger of reactive oxygen species (ROS) prior to exposure to DOX depressed the phosphorylation of NF-κB p65 induced by DOX. CONCLUSION: The present study has demonstrated the new mechanistic evidence that exogenous H2S attenuates the DOX-induced inflammation and cytotoxicity by inhibiting p38 MAPK/NF-κB pathway in H9c2 cardiac cells. We also provide novel data that the interaction between NF-κB pathway and IL-1ß is important in the induction of DOX-induced inflammation and cytotoxicity in H9c2 cardiac cells.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Inflammation/chemically induced , NF-kappa B/metabolism , Signal Transduction/drug effects , Sulfites/pharmacology , Acetylcysteine/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Inflammation/pathology , Interleukin-1beta/analysis , Interleukin-6/analysis , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Proline/analogs & derivatives , Proline/pharmacology , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species/metabolism , Thiocarbamates/pharmacology , Tumor Necrosis Factor-alpha/analysis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Mol Med Rep ; 8(2): 603-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23807148

ABSTRACT

A number of studies have demonstrated that inflammation plays a role in doxorubicin (DOX)-induced cardiotoxicity. However, the molecular mechanism by which DOX induces cardiac inflammation has yet to be fully elucidated. The present study aimed to investigate the role of the p38 mitogen-activated protein kinase (MAPK)/nuclear factor-κB (NF-κB) pathway in DOX-induced inflammation and cytotoxicity. The results of our study demonstrated that the exposure of H9c2 cardiac cells to DOX reduced cell viability and stimulated an inflammatory response, as demonstrated by an increase in the levels of interleukin-1ß (IL-1ß) and IL-6, as well as tumor necrosis factor-α (TNF-α) production. Notably, DOX exposure induced the overexpression of phosphorylated p38 MAPK and phosphorylation of the NF-κB p65 subunit, which was markedly inhibited by SB203580, a specific inhibitor of p38 MAPK. The inhibition of NF-κB by pyrrolidine dithiocarbamate (PDTC), a selective inhibitor of NF-κB, significantly ameliorated DOX-induced inflammation, leading to a decrease in the levels of IL-1ß and IL-6, as well as TNF-α production in H9c2 cells. The pretreatment of H9c2 cells with either SB203580 or PDTC before exposure to DOX significantly attenuated DOX-induced cytotoxicity. In conclusion, our study provides novel data demonstrating that the p38 MAPK/NF-κB pathway is important in the induction of DOX-induced inflammation and cytotoxicity in H9c2 cardiac myocytes.


Subject(s)
Doxorubicin/toxicity , Inflammation/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Enzyme Activation/drug effects , Inflammation/chemically induced , Phosphorylation/drug effects , Rats , Transcription Factor RelA/metabolism
16.
Int J Mol Med ; 31(3): 644-50, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23338126

ABSTRACT

We previously demonstrated the protective effect of hydrogen sulfide (H2S) against doxorubicin (DOX)-induced cardiotoxicity through inhibition of endoplasmic reticulum stress. The aim of the present study was to explore the role of p38 mitogen-activated protein kinase (MAPK) in DOX-induced cardiotoxicity and ascertain whether exogenous H2S protects DOX-induced injury by inhibiting p38 MAPK in cardiomyoblasts (H9c2). We observed that exposure of H9c2 cells to 5 µM DOX not only markedly induced injuries, including cytotoxicity, apoptosis, overproduction of reactive oxygen species (ROS) and dissipation of mitochondrial membrane potential (MMP), but also enhanced the expression level of phosphorylated (p)-p38 MAPK. The DOX-induced increase in expression of p-p38 MAPK was significantly attenuated by pretreatment of H9c2 cells with either 400 µM sodium hydrogen sulfide (NaHS) (a donor of H2S) or 1,000 µM N-acetyl-L-cysteine (NAC, an ROS scavenger) prior to exposure to DOX. Pretreatment with either 400 µM NaHS or 3 µM SB203580, a selective inhibitor of p38 MAPK, ameliorated DOX-induced cardiomyocyte injuries, as evidenced by an increase in cell viability, and decreases in the number of apoptotic cells, ROS generation as well as dissipation of MMP. In conclusion, the findings of the present study demonstrated that the activation of p38 MAPK contributes to DOX-induced injuries, including cytotoxicity, apoptosis, mitochondrial damage and oxidative stress in H9c2 cells. We also provide novel evidence that exogenous H2S protects H9c2 cells against DOX-induced cardiotoxicity by inhibition of the p38 MAPK pathway.


Subject(s)
Doxorubicin/adverse effects , Hydrogen Sulfide/pharmacology , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/drug effects , p38 Mitogen-Activated Protein Kinases , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Cardiotoxins , Cell Line , Cell Survival/drug effects , Imidazoles/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Phosphorylation , Pyridines/pharmacology , Rats , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Am J Med Sci ; 344(6): 473-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23187120

ABSTRACT

BACKGROUND: The chemokine monocyte chemoattractant protein-1 (MCP-1) has been shown to contribute to neuropathic pain. However, whether MCP-1 is involved in the development of morphine antinociceptive tolerance is incompletely understood. METHODS: Morphine antinociceptive tolerance was induced by intrathecal administration of 15 µg of morphine daily for 7 days. Immunohistochemistry was used to test the changes in the morphology of spinal MCP-1 immunoreactivity and OX-42-IR. The role of MCP-1 in morphine antinociceptive tolerance is explored by hot-water tail-flick test. RESULTS: Our findings showed that intrathecal chronic morphine exposure obviously increased MCP-1 immunoreactivity in the spinal cord. Moreover, the increased MCP-1 immunoreactivity was observed mainly in the spinal neurons. Intrathecal injections of MCP-1-neutralizing antibody significantly reduced the development of morphine antinociceptive tolerance, suggesting that spinal neuronal MCP-1 contributes to tolerance to morphine antinociception. Treatment with MCP-1-neutralizing antibody also reduced the spinal microglial activation induced by chronic morphine treatment. CONCLUSIONS: This study revealed for the first time that spinal neuronal MCP-1 is a key mediator of the spinal microglial activation and that spinal MCP-1 is involved in morphine antinociceptive tolerance. Inhibition of MCP-1 may provide a new therapy for morphine tolerance management.


Subject(s)
Analgesics, Opioid/administration & dosage , Chemokine CCL2/physiology , Drug Tolerance/physiology , Morphine/administration & dosage , Spinal Cord/physiology , Animals , Antibodies, Neutralizing/administration & dosage , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/immunology , Immunohistochemistry , Male , Microglia/drug effects , Microglia/physiology , Nociception/drug effects , Nociception/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Up-Regulation/drug effects
18.
Int J Mol Med ; 30(5): 1126-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22895544

ABSTRACT

Hypoxia and/or ischemia are implicated in neurodegenerative disorders. In these diseases, hypoxia/ischemia may induce oxidative stress, including production of reactive oxygen species (ROS), which result in a decrease in glutamate transporter expression. Hydrogen sulfide (H2S), as the third gasotransmitter, has neuroprotective effects and potent antioxidant properties. In the present study, we investigated the role of glutamate transporter-1 (GLT-1) in the protection of H2S against chemical hypoxia-induced injury in PC12 cells. We found that cobalt chloride (CoCl2), a chemical hypoxia agent, reduced the expression of GLT-1 in a time-dependent manner. Pretreatment with NaHS (a donor of H2S) reversed the CoCl2-induced downregulation of GLT-1 expression. Pretreatment with DHK (a selective inhibitor of GLT-1) for 30 min prior to NaHS preconditioning significantly inhibited the cytoprotection of H2S against CoCl2-induced injuries, leading to an increase in cytotoxicity and apoptosis as well as to a loss of mitochondrial membrane potential (MMP). In addition, we found that similar to the effect of NaHS, pretreatment with NAC (a ROS scavenger) or U0126 (a MEK1/2 inhibitor) blocked the downregulation of GLT-1 expression induced by CoCl2. Collectively, we demonstrated for the first time that ROS and extracellular signal-regulated kinase 1/2 (ERK1/2)-mediated reduction of GLT-1 expression may be involved in chemical hypoxia-induced neural injury and that H2S attenuates this injury partly by upregulating GLT-1 expression in PC12 cells.


Subject(s)
Down-Regulation/drug effects , Excitatory Amino Acid Transporter 2/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Sulfide/pharmacology , Neuroprotective Agents/pharmacology , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cobalt/pharmacology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , MAP Kinase Signaling System , PC12 Cells , Rats
19.
Int J Mol Med ; 30(2): 314-20, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22614123

ABSTRACT

Both the phosphatidylinositol 3-kinase (PI3K)/Akt pathway and heme oxygenase-1 (HO-1) create a survival signal against oxidative stress-induced injuries. Although we have demonstrated that hydrogen peroxide (H2O2) preconditioning confers adaptive cytoprotection against oxidative stress-induced injury in PC12 cells, it remains unknown whether these defense systems are involved in the protective effect of H2O2 preconditioning. In the current study, PC12 cells were preconditioned with 100 µM H2O2 for 90 min, followed by 24 h recovery and subsequent exposure to 300 µM H2O2 for further 12 h. The findings showed that preconditioning with 100 µM H2O2 upregulated HO-1 expression. Zinc protoporphyrin IX (ZnPP), a selective inhibitor of HO-1, at a concentration of 15 µM, significantly attenuated H2O2 preconditioning-elicited cytotoxicity, apoptosis, oxidative stress and mitochondrial membrane potential (ΔΨm) loss in PC12 cells. In addition, H2O2 preconditioning enhanced phosphorylation of Akt. Treatment with 25 µM LY294002, a selective inhibitor of PI3K, for 20 min before H2O2 preconditioning blocked not only H2O2 preconditioning-induced HO-1 induction, but also the protective effect of H2O2 preconditioning against cytotoxicity. The present study provides novel evidence for the effect of preconditioning with H2O2 on the induction of HO-1, which contributes to the adaptive cytoprotection of H2O2 preconditioning against oxidative stress-induced cellular injury via a PI3K/Akt-dependent mechanism in PC12 cells.


Subject(s)
Heme Oxygenase-1/metabolism , Hydrogen Peroxide/pharmacology , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cytoprotection/drug effects , Cytoprotection/genetics , Gene Expression , Heme Oxygenase-1/genetics , PC12 Cells , Phosphorylation/drug effects , Rats , Up-Regulation/drug effects , Up-Regulation/genetics
20.
Mol Cell Biochem ; 362(1-2): 149-57, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22134701

ABSTRACT

Hydrogen sulfide (H(2)S) has been shown to exert cardioprotective effects. However, the roles of extracellular signal-regulated protein kinases 1/2 (ERK1/2) in H(2)S-induced cardioprotection have not been completely elucidated. In this study, cobalt chloride (CoCl(2)), a chemical hypoxia mimetic agent, was applied to treat H9c2 cells to establish a chemical hypoxia-induced cardiomyocyte injury model. The results showed that pretreatment with NaHS (a donor of H(2)S) before exposure to CoCl(2) attenuated the decreased cell viability, the increased apoptosis rate, the loss of mitochondrial membrane potential (ΔΨm), and the intracellular accumulation of reactive oxygen species (ROS) in H9c2 cells. Exposure of H9c2 cells to CoCl(2) or hydrogen peroxide (H(2)O(2)) upregulated expression of phosphorylated (p) ERK1/2, which was reduced by pretreatment with NaHS or N-acetyl-L-cysteine, a ROS scavenger. More importantly, U0126, a selective inhibitor of ERK1/2, mimicked the above cytoprotection of H(2)S against CoCl(2)-induced injury in H9c2 cells. In conclusion, these results indicate that H(2)S protects H9c2 cells against chemical hypoxia-induced injury partially by inhibiting ROS-mediated activation of ERK1/2.


Subject(s)
Cell Hypoxia , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Sulfide/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Cardiotonic Agents/pharmacology , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Cobalt/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Membrane Potential, Mitochondrial/drug effects , Nitriles/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...