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










Publication year range
1.
Chem Biol Interact ; 382: 110643, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37481222

ABSTRACT

To investigate the role of the liver kinase (LK) B1 protein, an activator of AMP-activated protein kinase (AMPK), in AMPK signaling suppression when exposed to vesicant, a kind of chemical warfare agent. Cultured human bronchial epithelial cells were inflicted with sulfur mustard (SM) analog, 2-chloroethyl ethyl sulfide (CEES) of 0.2-1.0 mM concentration, and cell proliferation, apoptosis, autophagy, and cellular ATP level were analyzed up to 24 h after the exposure. Focusing on LKB1, heat shock protein (HSP) 90, and cell division cycle (CDC) 37 proteins, the protein expression, phosphorylation, and interaction were examined with western blot, immunofluorescence staining, and/or immunoprecipitation. AMPK signaling was found to be inhibited 24 h after being exposed to either sub-cytotoxic (0.5 mM) or cytotoxic (1.0 mM) concentration of CEES based on MTS assay. Consistently, the degradation of the LKB1 protein and its less interaction with the HSP90/CDC37 complex was confirmed. It was found that 1.0, not 0.5 mM CEES also decreased the CDC37 protein, proteasome activity, and cellular ATP content that modulates HSP90 protein conformation. Inhibiting proteasome activity could alternatively activate autophagy. Finally, either 0.5 or 1.0 mM CEES activated HSP70 and autophagy, and the application of an HSP70 inhibitor blocked autophagy and autophagic degradation of the LKB1 protein. In conclusion, we reported here that AMPK signaling inactivation by CEES was a result of LKB1 protein loss via less protein complex formation and enhanced degradation.


Subject(s)
Mustard Gas , Humans , Mustard Gas/toxicity , AMP-Activated Protein Kinases , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases , Molecular Chaperones , HSP90 Heat-Shock Proteins , Epithelial Cells/metabolism , Adenosine Triphosphate , Cell Cycle Proteins/metabolism , Chaperonins/metabolism
2.
Mol Neurobiol ; 60(10): 5915-5930, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37380822

ABSTRACT

MOTS-c is a 16-amino acid mitochondrial-derived peptide reported to be involved in regulating energy metabolism. However, few studies have reported the role of MOTS-c on neuron degeneration. In this study, it was aimed to explore the action of MOTS-c in rotenone-induced dopaminergic neurotoxicity. In an in vitro study, it was observed that rotenone could influence the expression and localization of MOTS-c significantly in PC12 cells, with more MOTS-c translocating into the nucleus from mitochondria. Further study showed that the translocation of MOTS-c from the mitochondria into the nucleus could directly interact with Nrf2 to regulate HO-1 and NQO1 expression in PC12 cells exposed to rotenone, which had been suggested to be involved in the antioxidant defense system. In vivo and in vitro experiments demonstrated that exogenous MOTS-c pretreatment could protect PC12 cells and rats from mitochondrial dysfunction and oxidative stress induced by rotenone. Moreover, MOTS-c pretreatment significantly decreased the loss of TH, PSD95, and SYP protein expression in the striatum of rats exposed to rotenone. In addition, MOTS-c pretreatment could clearly alleviate the downregulated expression of Nrf2, HO-1, and NQO1, as well as the upregulated Keap1 protein expression in the striatum of rotenone-treated rats. Taken together, these findings suggested that MOTS-c could directly interact with Nrf2 to activate the Nrf2/HO-1/NQO1 signal pathway to defend the antioxidant system to prevent dopaminergic neurons from rotenone-induced oxidative stress and neurotoxicity in vitro and in vivo.


Subject(s)
Antioxidants , Rotenone , Rats , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Rotenone/toxicity , Rotenone/metabolism , Dopaminergic Neurons/metabolism , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Oxidative Stress , Mitochondria/metabolism
3.
Toxicol Mech Methods ; 33(4): 271-278, 2023 May.
Article in English | MEDLINE | ID: mdl-36106344

ABSTRACT

Using sulfur mustard analog 2-chloroethyl ethyl sulfide (CEES), we established an in vitro model by poisoning cultured immortalized human bronchial epithelial cells. Nile Red staining revealed lipids accumulated 24 h after a toxic dose of CEES (0.9 mM). Lipidomics analysis showed most of the increased lipids were triglycerides (TGs), and the increase in TGs was further confirmed using a Triglyceride-Glo™ Assay kit. Protein and mRNA levels of DGAT1, an important TG biogenesis enzyme, were increased following 0.4 mM CEES exposure. Under higher dose CEES (0.9 mM) exposure, protein and mRNA levels of PPARγ coactivator-1ɑ (PGC-1ɑ), a well-known transcription factor that regulates fatty acid oxidation, were decreased. Finally, application with DGAT1 inhibitor A 922500 or PGC1ɑ agonist ZLN005 was able to block the CEES-induced TGs increase. Overall, our dissection of CEES-induced TGs accumulation provides new insight into energy metabolism dysfunction upon vesicant exposure.HIGHLIGHTSIn CEES (0.9 mM)-injured cells:Triglycerides (TGs) were abundant in the accumulated lipids.Expression of DGAT1, not DGAT2, was increased.Expression of PGC1ɑ, not PGC1ß, was reduced.DGAT1 inhibitor or PGC1ɑ agonist blocked the CEES-mediated increase in TGs.


Subject(s)
Mustard Gas , Humans , Diacylglycerol O-Acyltransferase/genetics , Epithelial Cells/drug effects , Lipids , Mustard Gas/analogs & derivatives , Mustard Gas/toxicity , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , RNA, Messenger , Sulfides
4.
Acta Pharmacol Sin ; 43(6): 1419-1429, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34593973

ABSTRACT

The multi-generation heredity trait of hypertension in human has been reported, but the molecular mechanisms underlying multi-generational inheritance of hypertension remain obscure. Recent evidence shows that prenatal inflammatory exposure (PIE) results in increased incidence of cardiovascular diseases, including hypertension. In this study we investigated whether and how PIE contributed to multi-generational inheritance of hypertension in rats. PIE was induced in pregnant rats by intraperitoneal injection of LPS or Poly (I:C) either once on gestational day 10.5 (transient stimulation, T) or three times on gestational day 8.5, 10.5, and 12.5 (persistent stimulation, P). Male offspring was chosen to study the paternal inheritance. We showed that PIE, irrespectively induced by LPS or Poly (I:C) stimulation during pregnancy, resulted in multi-generational inheritance of significantly increased blood pressure in rat descendants, and that prenatal LPS exposure led to vascular remodeling and vasoconstrictor dysfunction in both thoracic aorta and superior mesenteric artery of adult F2 offspring. Furthermore, we revealed that PIE resulted in global alteration of DNA methylome in thoracic aorta of F2 offspring. Specifically, PIE led to the DNA hypomethylation of G beta gamma (Gßγ) signaling genes in both the F1 sperm and the F2 thoracic aorta, and activation of PI3K/Akt signaling was implicated in the pathologic changes and dysregulated vascular tone of aortic tissue in F2 LPS-P offspring. Our data demonstrate that PIE reprogrammed DNA methylome of cells from the germline/mature gametes contributes to the development of hypertension in F2 PIE offspring. This study broadens the current knowledge regarding the multi-generation effect of the cumulative early life environmental factors on the development of hypertension.


Subject(s)
Heredity , Hypertension , Prenatal Exposure Delayed Effects , Animals , Epigenome , Female , Humans , Hypertension/chemically induced , Hypertension/genetics , Inflammation/chemically induced , Inflammation/genetics , Lipopolysaccharides/toxicity , Male , Phosphatidylinositol 3-Kinases/genetics , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/genetics , Rats
5.
Toxicol Lett ; 354: 14-23, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34757179

ABSTRACT

Respiratory system injury is the main cause of mortality for nitrogen mustard (NM)-induced damage. Previous studies indicate that reactive oxygen species (ROS) participates in NM-mediated respiratory injuries, but the detailed mechanism is not quite clear. Human bronchial epithelial cell lines 16HBE and BEAS-2B were treated with HN2, a type of NM. In detail, it was shown that HN2 treatment induced impaired cell viability, excessive mitochondrial ROS production and enhanced cellular apoptosis in bronchial epithelial cells. Moreover, impaired Sirt3/SOD2 axis was observed upon HN2 treatment, with decreased Sirt3 and increased acetylated SOD2 expression levels. Sirt3 overexpression partially ameliorated HN2-induced cell injury. Meanwhile, vitamin D3 treatment partially attenuated HN2-induced apoptosis and improved the mitochondrial functions upon HN2 intervention. In addition, HN2 exposure decreased VDR expression, thus inhibiting the Nrf2 phosphorylation and Sirt3 activation. Inhibition of Nrf2 or Sirt3 could decrease the protective effects of vitamin D3 and enhance mitochondrial ROS production via modulating mitochondrial redox balance. In conclusion, impaired VDR/Nrf2/Sirt3 axis contributed to NM-induced apoptosis, while vitamin D3 supplementation provides protective effects via the activation of VDR and the improvement of mitochondrial functions. This study provides novel mechanism and strategy for NM exposure-induced pulmonary injuries.


Subject(s)
Apoptosis/drug effects , Bronchi/drug effects , Cholecalciferol/pharmacology , Epithelial Cells/drug effects , Nitrogen Mustard Compounds/toxicity , Protective Agents/pharmacology , Respiratory System/drug effects , Cells, Cultured/drug effects , Humans , Respiratory System/physiopathology
6.
Toxicol Res (Camb) ; 10(5): 1034-1044, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34733488

ABSTRACT

Sulfur mustard (a type of vesicant) can directly damage lung bronchial epithelium via aerosol inhalation, and prevalent cell death is an early event that obstructs the respiratory tract. JNK/c-Jun is a stress response pathway, but its role in cell death of the injured cells is not clear. Here, we report that JNK/c-Jun was activated in immortalized human bronchial epithelial (HBE) cells exposed to a lethal dose (20 µM) of nitrogen mustard (NM, a sulfur mustard analog). c-Jun silencing using small-interfering RNA (siRNA) rendered the cells resistant to NM-mediated cell death by blocking poly(ADP-ribose) polymerase 1 (PARP1) cleavage and DNA fragmentation. In addition, the transduction of upstream extrinsic (Fasl-Fas-caspase-8) and intrinsic (loss of Bcl-2 and mitochondrial membrane potential, ΔΨm) apoptosis pathways, as well as phosphorylated (p)-H2AX (Ser139), an epigenetic marker contributing to DNA fragmentation and PARP1 activity, was partially suppressed. To mimic the detachment of cells by NM, HBE cells were trypsinized and seeded on culture plates that were pre-coated with poly-HEMA to prevent cell adhesion. The JNK/c-Jun pathway was found to be activated in the detached cells. In conclusion, our results indicate that JNK/c-Jun pathway activation is necessary for NM-caused HBE cell death and further suggest that c-Jun silencing may be a potential approach to protect HBE cells from vesicant damage.

7.
Clin Transl Med ; 11(2): e312, 2021 02.
Article in English | MEDLINE | ID: mdl-33634989

ABSTRACT

Nitrogen mustard (NM) causes severe skin injury with an obvious inflammatory response, which is lack of effective and targeted therapies. Vitamin D3 (VD3) has excellent anti-inflammatory properties and is considered as a potential candidate for the treatment of NM-induced dermal toxicity; however, the underlying mechanisms are currently unclear. Cyclooxygenase-2 (COX2; a widely used marker of skin inflammation) plays a key role in NM-induced cutaneous inflammation. Herein, we initially confirmed that NM markedly promoted COX2 expression in vitro and in vivo. NM also increased NOD-like receptor family pyrin domain containing 3 (NLRP3) expression, caspase-1 activity, and interleukin-1ß (IL-1ß) release. Notably, treatment with a caspase-1 inhibitor (zYVAD-fmk), NLRP3 inhibitor (MCC950), and NLRP3 or caspase-1 siRNA attenuated NM-induced NLRP3 inflammasome activation, with subsequent suppression of COX2 expression and IL-1ß release in keratinocytes. Meanwhile, NM increased mitochondrial reactive oxygen species (mtROS) and decreased manganese superoxide dismutase 2 (SOD2) and sirtuin 3 (SIRT3) activities. Mito-TEMPO (a mtROS scavenger) ameliorated NM-caused NLRP3 inflammasome activation in keratinocytes. Moreover, VD3 improved SIRT3 and SOD2 activities, decreased mtROS contents, inactivated the NLRP3 inflammasome, and attenuated cutaneous inflammation induced by NM in vitro and in vivo. The beneficial activity of VD3 against NM-triggered cutaneous inflammation was enhanced by the inhibitors of IL-1, mtROS, NLRP3, caspase-1, and NLRP3 or caspase-1 siRNAs, which was abolished in SIRT3 inhibitor or SIRT3 siRNA-treated keratinocytes and skins from SIRT3-/- mice. In conclusion, VD3 ameliorated NM-induced cutaneous inflammation by inactivating the NLRP3 inflammasome, which was partially mediated through the SIRT3-SOD2-mtROS signaling pathway.


Subject(s)
Dermatitis, Contact/etiology , Inflammasomes/drug effects , Mechlorethamine/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sirtuin 3/metabolism , Superoxide Dismutase/metabolism , Vitamin D/therapeutic use , Animals , Dermatitis, Contact/drug therapy , Female , HaCaT Cells/drug effects , HaCaT Cells/metabolism , Humans , Inflammasomes/physiology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism
8.
Signal Transduct Target Ther ; 6(1): 29, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33487631

ABSTRACT

Nitrogen mustard (NM) causes severe vesicating skin injury, which lacks effective targeted therapies. The major limitation is that the specific mechanism of NM-induced skin injury is not well understood. Recently, autophagy has been found to play important roles in physical and chemical exposure-caused cutaneous injuries. However, whether autophagy contributes to NM-induced dermal toxicity is unclear. Herein, we initially confirmed that NM dose-dependently caused cell death and induced autophagy in keratinocytes. Suppression of autophagy by 3-methyladenine, chloroquine, and bafilomycin A1 or ATG5 siRNA attenuated NM-induced keratinocyte cell death. Furthermore, NM increased transient receptor potential vanilloid 1 (TRPV1) expression, intracellular Ca2+ content, and the activities of Ca2+/calmodulin-dependent kinase kinase ß (CaMKKß), AMP-activated protein kinase (AMPK), unc-51-like kinase 1 (ULK1), and mammalian target of rapamycin (mTOR). NM-induced autophagy in keratinocytes was abolished by treatment with inhibitors of TRPV1 (capsazepine), CaMKKß (STO-609), AMPK (compound C), and ULK1 (SBI-0206965) as well as TRPV1, CaMKKß, and AMPK siRNA transfection. In addition, an mTOR inhibitor (rapamycin) had no significant effect on NM-stimulated autophagy or cell death of keratinocytes. Finally, the results of the in vivo experiment in NM-treated skin tissues were consistent with the findings of the in vitro experiment. In conclusion, NM-caused dermal toxicity by overactivating autophagy partially through the activation of TRPV1-Ca2+-CaMKKß-AMPK-ULK1 signaling pathway. These results suggest that blocking TRPV1-dependent autophagy could be a potential treatment strategy for NM-caused cutaneous injury.


Subject(s)
Autophagy-Related Protein 5/genetics , Autophagy/genetics , Skin Diseases/genetics , TRPV Cation Channels/genetics , AMP-Activated Protein Kinase Kinases/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy-Related Protein-1 Homolog/genetics , Blister/genetics , Blister/pathology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cell Death/drug effects , Chloroquine/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Macrolides/pharmacology , Mechlorethamine/toxicity , Mice , RNA, Small Interfering/genetics , Sirolimus/pharmacology , Skin/drug effects , Skin/injuries , Skin/pathology , Skin Diseases/chemically induced , Skin Diseases/drug therapy , Skin Diseases/pathology , TOR Serine-Threonine Kinases/genetics
9.
J Mol Neurosci ; 71(11): 2336-2352, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33515431

ABSTRACT

Mitochondria harbor small circular genomes (mtDNA) that encode 13 oxidative phosphorylation (OXPHOS) proteins, and types of damage to mtDNA may contribute to neuronal damage. Recent studies suggested that regulation of mtDNA repair proteins may be a potential strategy for treating neuronal damage. The mtDNA repair system contains its own repair enzymes and is independent from the nuclear DNA repair system. Endo/exonuclease G-like(EXOG) is a mitochondria-specific 5-exo/endonuclease required for repairing endogenous single-strand breaks (SSBs) in mtDNA. However, whether EXOG plays a key role in neuronal damage induced by rotenone remains unknown. Thus, in this study, we aimed to investigate the effect of EXOG on mtDNA repair and mitochondrial functional maintenance in rotenone-induced neurotoxicity. Our results indicated that rotenone influenced the expression and location of EXOG in PC12 cells. Meanwhile, after rotenone exposure, the expression was reduced for proteins responsible for mtDNA repair, including DNA polymerase γ (POLG), high-temperature requirement protease A2 (HtrA2), and the heat-shock factor 1-single-stranded DNA-binding protein 1 (HSF1-SSBP1) complex. Further analysis demonstrated that EXOG knockdown led to reduced mtDNA copy number and mtDNA transcript level and increased mtDNA deletion, which further aggravated the mtDNA damage and mitochondrial dysfunction under rotenone stress. In turn, EXOG overexpression protected PC12 cells from mtDNA damage and mitochondrial dysfunction induced by rotenone. As a result, EXOG knockdown reduced cell viability and tyrosine hydroxylase expression, while EXOG overexpression alleviated rotenone's effect on cell viability and tyrosine hydroxylase expression in PC12 cells. Further, we observed that EXOG influenced mtDNA repair by regulating protein expression of the HSF1-SSBP1 complex and POLG. Furthermore, our study showed that PGC-1α upregulation with ZLN005 led to increased protein levels of EXOG, POLG, HSF1, and SSBP1, all of which contribute to mtDNA homeostasis. Therefore, PGC-1α may be involved in mtDNA repair through interacting with multiple mtDNA repair proteins, especially with the help of EXOG. In summary, EXOG regulation by PGC-1α plays an essential role in rotenone-induced neurotoxicity in PC12 cells. EXOG represents a protective effect strategy in PC12 cells exposed to rotenone.


Subject(s)
DNA Repair , Endonucleases/metabolism , Mitochondria/metabolism , Rotenone/toxicity , Uncoupling Agents/toxicity , Animals , DNA Damage , DNA Polymerase gamma/metabolism , DNA, Mitochondrial/genetics , DNA-Binding Proteins/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , PC12 Cells , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats , Serine-Arginine Splicing Factors/metabolism
10.
Toxicol Lett ; 319: 256-263, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31639410

ABSTRACT

Transcription factor activator protein (AP)-1 can be activated in nitrogen-mustard-injured mouse skin, and is thought to participate in the inflammatory response. AP-1 consists of homo- or heterodimers of Fos [c-Fos, Fos-B, fos-related antigen (Fra)-1 and Fra-2] and Jun (c-Jun, JunB and JunD) family members, and information about their expression, location and function are still unclear. In nitrogen-mustard-exposed mouse skin, we found p-ERK activation increased Fra-1 and FosB. Unlike the nucleus location of c-Fos and FosB, Fra-1 and Fra-2 were mainly expressed in the cytoplasm. In nitrogen-mustard-exposed cultured immortalized human keratinocytes (HaCaT cells), Fra-1 in the nucleus functioned as an inhibitor of inflammatory cytokine interleukin (IL)-8. Co-immunoprecipitation showed that Fra-1 formed dimers with IL-8 transcription factors c-Jun, JunB and JunD. Fra-1 depletion increased c-Fos and FosB in the nucleus, accompanied by increased heterodimers of c-Fos/c-Jun, c-Fos/JunB, c-Fos/JunD, and FosB/JunB. In conclusion, Fra-1 trapped in the cytoplasm after nitrogen mustard exposure might be a driving force for IL-8 over-expression in injured skin.


Subject(s)
Chemical Warfare Agents/toxicity , Epidermis/injuries , Epidermis/metabolism , Interleukin-8/biosynthesis , Mechlorethamine/toxicity , Proto-Oncogene Proteins c-fos/metabolism , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Humans , Keratinocytes/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Hairless , RNA, Small Interfering/pharmacology
11.
J Cell Biochem ; 120(10): 18219-18235, 2019 10.
Article in English | MEDLINE | ID: mdl-31245869

ABSTRACT

This study aims to explore the predictive noninvasive biomarker for obstructive coronary artery disease (CAD). By using the data set GSE90074, weighted gene co-expression network analysis (WGCNA), and protein-protein interactive network, construction of differentially expressed genes in peripheral blood mononuclear cells was conducted to identify the most significant gene clusters associated with obstructive CAD. Univariate and multivariate stepwise logistic regression analyses and receiver operating characteristic analysis were used to predicate the diagnostic accuracy of biomarker candidates in the detection of obstructive CAD. Furthermore, functional prediction of candidate gene biomarkers was further confirmed in ST-segment elevation myocardial infarction (STEMI) patients or stable CAD patients by using the datasets of GSE62646 and GSE59867. We found that the blue module discriminated by WGCNA contained 13 hub-genes that could be independent risk factors for obstructive CAD (P < .05). Among these 13 hub-genes, a four-gene signature including neutrophil cytosol factor 2 (NCF2, P = .025), myosin-If (MYO1F, P = .001), sphingosine-1-phosphate receptor 4 (S1PR4, P = .015), and ficolin-1 (FCN1, P = .012) alone or combined with two risk factors (male sex and hyperlipidemia) may represent potential diagnostic biomarkers in obstructive CAD. Furthermore, the messenger RNA levels of NCF2, MYO1F, S1PR4, and FCN1 were higher in STEMI patients than that in stable CAD patients, although S1PR4 showed no statistical difference (P > .05). This four-gene signature could also act as a prognostic biomarker to discriminate STEMI patients from stable CAD patients. These findings suggest a four-gene signature (NCF2, MYO1F, S1PR4, and FCN1) alone or combined with two risk factors (male sex and hyperlipidemia) as a promising prognostic biomarker in the diagnosis of STEMI. Well-designed cohort studies should be implemented to warrant the diagnostic value of these genes in clinical purpose.


Subject(s)
Biomarkers/metabolism , Coronary Artery Disease/genetics , Gene Expression Profiling , Lectins/genetics , Myosin Type I/genetics , NADPH Oxidases/genetics , Sphingosine-1-Phosphate Receptors/genetics , Arterial Occlusive Diseases/diagnosis , Arterial Occlusive Diseases/genetics , Cells, Cultured , Coronary Artery Disease/diagnosis , Female , Gene Ontology , Gene Regulatory Networks , Humans , Leukocytes, Mononuclear/metabolism , Male , ROC Curve , Ficolins
12.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1086-1103, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29353068

ABSTRACT

Parkinson's disease (PD) is the second most common age-related neurodegenerative disease. Mitochondrial dysfunction has been the focus of the pathogenesis of PD. The mitochondrial ATP-sensitive potassium channel (mitoKATP) plays a significant role in mitochondrial physiology and has been extensively shown to protect against ischemic and brain reperfusion injury. However, there have long been controversies regarding its role in Parkinson's disease. We investigated the role of mitoKATP channels in rotenone-induced PD model in vivo and vitro and the interactions of mitoKATP channels, mitochondrial dynamics and PD. The results indicated that the use of diazoxide to activate mitoKATP channels resulted in the aggravation of rotenone-induced dopamine neurodegeneration in PC12 cells and SD rats. In contrast, the use of 5-hydroxydecanoate (5-HD) to inhibit mitoKATP channels improved rotenone-induced dopamine neurodegeneration, which was not consistent with mitoKATP channels in ischemic and brain reperfusion injury. Further analysis determined that the mitoKATP channel was involved in PD mainly via the regulation of mitochondrial biogenesis and fission/fusion. And the pore subunits of Kir6.1, the major component of mitoKATP channels, was the key contributor in its interaction with mitochondrial dynamics in rotenone-induced dopamine neurodegeneration. Therefore, it can be concluded that mitoKATP channels regulate mitochondrial dynamics to participate in rotenone-induced PD mainly attributes to the pore subunits of Kir6.1. And additionally, though mitoKATP channels may represent a direction of one potential target for neuroprotection, it should be noted that the effects are different in the activation or inhibition of mitoKATP channels in different models.


Subject(s)
KATP Channels/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Parkinson Disease, Secondary/metabolism , Animals , Male , Mitochondria/pathology , PC12 Cells , Parkinson Disease, Secondary/pathology , Rats , Rats, Sprague-Dawley , Rotenone/adverse effects , Rotenone/pharmacology
13.
Toxicology ; 389: 67-73, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28720507

ABSTRACT

N-methyl-2,2-di(chloroethyl)amine (HN2) is a kind of bifunctional alkyltating agent, which can react with nucleophilic groups in DNA and/or protein to form HN2-bridged crosslinking of target molecules, such as DNA-protein crosslinkings (DPC). O6-methylguanine-DNA methyltransferase (MGMT) is a DNA damage repair enzyme which solely repairs alkyl adduct on DNA directly. However, MGMT was detected to act as a protein cross-linked with DNA via alkylation in presence of HN2, and unexpectedly turned into a DNA damage enhancer in the form of MGMT-DNA cross-link (mDPC). Present study aimed to explore the possible ways to lessen the incorporation of MGMT into DPC as well as to save it for DNA repair. To find out the influencing factors of mDPC formation and cleavage, human bronchial epithelial cell line 16HBE was exposed to HN2 and the factors related with MGMT expression and degradation were investigated. When c-Myc, a negative transcriptional factor of MGMT was inhibited by 10058-F4, MGMT expression and mDPC formation were increased, and more γ-H2AX was also detected. Sustained treatment with O6BG, a specific exogenous substrate and depleter of MGMT, could reduce the level of MGMT and mDPC formation. In contrast, a transient 1h pre-treatment of O6GB before HN2 exposure would cause a high MGMT and mDPC level. MGMT was increasingly ubiquitinated after HN2 exposure in a time-dependent manner. At the same time, MGMT was also SUMOylated with a downward time-dependent manner compared to its ubiquitination. Inhibitors of E1, E2 or E3 ligases of ubiqutination all led to the accumulation of mDPC and total-DPC (tDPC) with the difference as that mDPC was sensitive to E1 inhibitor while tDPC more sensitive to E2 and E3 inhibitor. Our results demonstrated the control of mDPC level could be realized through transcription inhibitory effect of c-Myc, O6GB application, and the acceleration of mDPC ubiquitination and subsequent degradation.


Subject(s)
Alkylating Agents/toxicity , Bronchi/drug effects , Cross-Linking Reagents/toxicity , DNA Adducts/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Epithelial Cells/drug effects , Mechlorethamine/toxicity , Tumor Suppressor Proteins/metabolism , Bronchi/metabolism , Bronchi/pathology , Cell Line , DNA Adducts/chemistry , DNA Modification Methylases/chemistry , DNA Modification Methylases/genetics , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Histones/metabolism , Humans , NF-kappa B p50 Subunit/metabolism , Protein Binding , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Sumoylation , Time Factors , Transcription, Genetic/drug effects , Transfection , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
14.
Mol Neurobiol ; 54(5): 3783-3797, 2017 07.
Article in English | MEDLINE | ID: mdl-27271125

ABSTRACT

Parkinson's disease is a common neurodegenerative disease in the elderly, and mitochondrial defects underlie the pathogenesis of PD. Impairment of mitochondrial homeostasis results in reactive oxygen species formation, which in turn can potentiate the accumulation of dysfunctional mitochondria, forming a vicious cycle in the neuron. Mitochondrial fission/fusion and biogenesis play important roles in maintaining mitochondrial homeostasis. It has been reported that PGC-1α is a powerful transcription factor that is widely involved in the regulation of mitochondrial biogenesis, oxidative stress, and other processes. Therefore, we explored mitochondrial biogenesis, mitochondrial fission/fusion, and especially PGC-1α as the key point in the signaling mechanism of their interaction in rotenone-induced dopamine neurotoxicity. The results showed that mitochondrial number and mass were reduced significantly, accompanied by alterations in proteins known to regulate mitochondrial fission/fusion (MFN2, OPA1, Drp1, and Fis1) and mitochondrial biogenesis (PGC-1α and mtTFA). Further experiments proved that inhibition of mitochondrial fission or promotion of mitochondrial fusion has protective effects in rotenone-induced neurotoxicity and also promotes mitochondrial biogenesis. By establishing cell models of PGC-1α overexpression and reduced expression, we found that PGC-1α can regulate MFN2 and Drp1 protein expression and phosphorylation to influence mitochondrial fission/fusion. In summary, it can be concluded that PGC-1α-mediated cross talk between mitochondrial biogenesis and fission/fusion contributes to rotenone-induced dopaminergic neurodegeneration.


Subject(s)
Dopaminergic Neurons/pathology , Mitochondrial Dynamics , Neurotoxins/toxicity , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rotenone/toxicity , Animals , Cell Survival/drug effects , DNA, Mitochondrial/genetics , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Dynamics/drug effects , PC12 Cells , Phosphorylation/drug effects , Rats
15.
Sci Rep ; 6: 30146, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27443826

ABSTRACT

Maternal inflammation contributes to the increased incidence of adult cardiovascular disease. The current study investigated the susceptibility of cardiac damage responding to isoproterenol (ISO) in adult offspring that underwent maternal inflammation (modeled by pregnant Sprague-Dawley rats with lipopolysaccharides (LPS) challenge). We found that 2 weeks of ISO treatment in adult offspring of LPS-treated mothers led to augmented heart damage, characterized by left-ventricular systolic dysfunction, cardiac hypertrophy and myocardial fibrosis. Mechanistically, prenatal exposure to LPS led to up-regulated expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, antioxidant enzymes, and p38 MAPK activity in left ventricular of adult offspring at resting state. ISO treatment exaggerated ROS generation, p38 MAPK activation but down-regulated reactive oxygen species (ROS) elimination capacity in the left ventricular of offspring from LPS-treated mothers, while antioxidant N-acetyl-L-cysteine (NAC) reversed these changes together with improved cardiac functions. The p38 inhibitor SB202190 alleviated the heart damage only via inhibiting the expression of NADPH oxidases. Collectively, our data demonstrated that prenatal inflammation programs pre-existed ROS activation in the heart tissue, which switches on the early process of oxidative damages on heart rapidly through a ROS-p38 MAPK-NADPH oxidase-ROS positive feedback loop in response to a myocardial hypertrophic challenge in adulthood.


Subject(s)
Heart Diseases/metabolism , Heart/physiopathology , Inflammation/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antioxidants/metabolism , Female , Heart/drug effects , Heart Diseases/chemically induced , Imidazoles/pharmacology , Inflammation/chemically induced , Isoproterenol/pharmacology , Male , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pregnancy , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Toxicol Appl Pharmacol ; 305: 267-273, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27342729

ABSTRACT

Nitrogen mustard (NM), a bifunctional alkylating agent (BAA), contains two alkyl arms and can act as a cross-linking bridge between DNA and protein to form a DNA-protein cross-link (DPC). O(6)-methylguanine-DNA methyltransferase (MGMT), a DNA repair enzyme for alkyl adducts removal, is found to enhance cell sensitivity to BAAs and to promote damage, possibly due to its stable covalent cross-linking with DNA mediated by BAAs. To investigate MGMT-DNA cross-link (mDPC) formation and its possible dual roles in NM exposure, human bronchial epithelial cell line 16HBE was subjected to different concentrations of HN2, a kind of NM, and we found mDPC was induced by HN2 in a concentration-dependent manner, but the mRNA and total protein of MGMT were suppressed. As early as 1h after HN2 treatment, high mDPC was achieved and the level maintained for up to 24h. Quick total DPC (tDPC) and γ-H2AX accumulation were observed. To evaluate the effect of newly predicted protease DVC1 on DPC cleavage, we applied siRNA of MGMT and DVC1, MG132 (proteasome inhibitor), and NMS-873 (p97 inhibitor) and found that proteolysis plays a role. DVC1 was proven to be more important in the cleavage of mDPC than tDPC in a p97-dependent manner. HN2 exposure induced DVC1 upregulation, which was at least partially contributed to MGMT cleavage by proteolysis because HN2-induced mDPC level and DNA damage was closely related with DVC1 expression. Homologous recombination (HR) was also activated. Our findings demonstrated that MGMT might turn into a DNA damage promoter by forming DPC when exposed to HN2. Proteolysis, especially DVC1, plays a crucial role in mDPC repair.


Subject(s)
Alkylating Agents/toxicity , DNA Damage/physiology , Mechlorethamine/toxicity , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Cell Line , DNA/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Histones/metabolism , Humans , Proteolysis
17.
Oxid Med Cell Longev ; 2016: 6705621, 2016.
Article in English | MEDLINE | ID: mdl-26770656

ABSTRACT

It has been confirmed that mitochondrial impairment may underlie both sporadic and familial Parkinson's disease (PD). Mitochondrial fission/fusion and biogenesis are key processes in regulating mitochondrial homeostasis. Therefore, we explored whether the protective effect of resveratrol in rotenone-induced neurotoxicity was associated with mitochondrial fission/fusion and biogenesis. The results showed that resveratrol could not only promote mitochondrial mass and DNA copy number but also improve mitochondrial homeostasis and neuron function in rats and PC12 cells damaged by rotenone. We also observed effects with alterations in proteins known to regulate mitochondrial fission/fusion and biogenesis in rotenone-induced neurotoxicity. Therefore, our findings suggest that resveratrol may prevent rotenone-induced neurotoxicity through regulating mitochondrial fission/fusion and biogenesis.


Subject(s)
Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Neurotoxins/toxicity , Organelle Biogenesis , Rotenone/toxicity , Stilbenes/pharmacology , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , DNA, Mitochondrial/genetics , Gene Dosage , Male , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , PC12 Cells , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Resveratrol , Rotarod Performance Test , Survival Analysis , Transcription Factors/metabolism , Transcription, Genetic/drug effects
18.
Onco Targets Ther ; 8: 2709-19, 2015.
Article in English | MEDLINE | ID: mdl-26445554

ABSTRACT

BACKGROUND: MicroRNA-34a (miR-34a) is a potential prognostic factor for survival in patients with several types of cancer according to previous clinical researches. We conducted a systematic review and meta-analysis to summarize the significance of increased miR-34a expression in the prognosis of patients' overall survival. MATERIALS AND METHODS: The present systematic review and meta-analysis of 15 researches included 2,597 patients. Overexpression of miR-34a may predict good overall survival ([OS], HR =0.76, 95% confidence interval: 0.55-1.06, P=0.105), but the effect was not significant enough. Subgroup analysis results showed miR-34a was an ideal predictor for digestive system cancer (OS, HR =0.50, 95% confidence interval: 0.25-0.99, P=0.048). The predictive effects of elevated expression of miR-34a on the OS of untreated and treated patients were not of obvious differences. CONCLUSION: This systematic review and meta-analysis showed that miR-34a has a predictive effect on overall survival of patients with digestive system cancer.

19.
Int J Mol Sci ; 16(8): 17018-28, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26225959

ABSTRACT

BACKGROUND: MiR-198 has been considered as an inhibitor of cell proliferation, invasion, migration and a promoter of apoptosis in most cancer cells, while its effect on non-cancer cells is poorly understood. METHODS: The effect of miR-198 transfection on HaCaT cell proliferation was firstly detected using Cell Count Kit-8 and the cell cycle progression was analyzed by flow cytometry. Using bioinformatics analyses and luciferase assay, a new target of miR-198 was searched and identified. Then, the effect of the new target gene of miR-198 on cell proliferation and cell cycle was also detected. RESULTS: Here we showed that miR-198 directly bound to the 3'-UTR of CCND2 mRNA, which was a key regulator in cell cycle progression. Overexpressed miR-198 repressed CCND2 expression at mRNA and protein levels and subsequently led to cell proliferation inhibition and cell cycle arrest in the G1 phase. Transfection ofSiCCND2 in HaCaT cells showed similar inhibitory effects on cell proliferation and cell cycle progression. CONCLUSION: In conclusion, we have identified that miR-198 inhibited HaCaT cell proliferation by directly targeting CCND2.


Subject(s)
Cyclin D2/genetics , Keratinocytes/cytology , Keratinocytes/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Binding Sites , Cell Cycle Checkpoints/genetics , Cell Line , Cell Proliferation , Cyclin D2/metabolism , Enzyme Assays , G1 Phase/genetics , Humans , Luciferases/metabolism , MicroRNAs/genetics , Molecular Sequence Data , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transfection
20.
Arch Med Sci ; 9(3): 561-8, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23847683

ABSTRACT

INTRODUCTION: The aim of this study was to construct a lentivirus vector with survivin promoter (pSur)-driven apoptin and test its efficiency in suppressing the growth of tumor cells. MATERIAL AND METHODS: Expression cassettes with different fragments of survivin gene promoter (pSur, 161 bp, 272 bp, 990 bp) driving 6XHis-tagged apoptin were constructed to generate recombinant lentivirus, of which the inhibitory effect on tumor cells was compared. The activity of different pSur in 293FT, and 272 bp pSur in primary bone marrow mesenchymal stem cells (BMSCs), SW480, Hela and MCF-7 was examined by Western blot. Their ability to induce apoptosis in SW480 cells was determined by annexin-V staining. The inhibitory effect of letivirus containing different pSur-driven apoptin on nude mice-xenografted SW480 cells was assessed by tumor size and pathological observation. RESULTS: The 272 bp and 990 bp pSur displayed comparable effects in terms of promoter activity, cell apoptosis/necrosis and G1 phase arrest in vitro, and growth of xenograft tumor in vivo. When lentivirus containing 272 bp pSur was tested, it drove high apoptin expression in tumor cells (SW480, Hela and MCF-7) and weak expression in primary bone marrow mesenchymal stem cells. Xenograft to nude mice using infected Sw480 cells showed that lentiviruses possessing 272 bp and 990 bp pSur were able to significantly induce tumor cell death, focal necrosis, and tumor growth lag. CONCLUSIONS: The data indicated that pSur-apoptin expression cassette in lentivirus vector ensures specific suppression of tumor cells, and may be applicable to monitor malignant transformation of transplanted cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...