Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
Molecules ; 28(8)2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37110816

ABSTRACT

Procyanidins (PCs), which are organic antioxidants, suppress oxidative stress, exhibit anti-apoptotic properties, and chelate metal ions. The potential defense mechanism of PCs against cerebral ischemia/reperfusion injury (CIRI) was investigated in this study. Pre-administration for 7 days of a PC enhanced nerve function and decreased cerebellar infarct volume in a mouse middle cerebral artery embolization paradigm. In addition, mitochondrial ferroptosis was enhanced, exhibited by mitochondrial shrinkage and roundness, increased membrane density, and reduced or absent ridges. The level of Fe2+ and lipid peroxidation that cause ferroptosis was significantly reduced by PC administration. According to the Western blot findings, PCs altered the expression of proteins associated with ferroptosis, promoting the expression of GPX4 and SLC7A11 while reducing the expression of TFR1, hence inhibiting ferroptosis. Moreover, the treatment of PCs markedly elevated the expression of HO-1 and Nuclear-Nrf2. The PCs' ability to prevent ferroptosis due to CIRI was decreased by the Nrf2 inhibitor ML385. Our findings showed that the protective effect of PCs may be achieved via activation of the Nrf2/HO-1 pathway and inhibiting ferroptosis. This study provides a new perspective on the treatment of CIRI with PCs.


Subject(s)
Brain Ischemia , Ferroptosis , Proanthocyanidins , Reperfusion Injury , Animals , Mice , Proanthocyanidins/pharmacology , NF-E2-Related Factor 2 , Signal Transduction , Reperfusion Injury/drug therapy
2.
Biomed Pharmacother ; 162: 114671, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37037094

ABSTRACT

Stroke is one of the leading causes of death and long-term disability worldwide. More than 80 % of strokes are ischemic, caused by an occlusion of cerebral arteries. Without question, restoration of blood supply as soon as possible is the first therapeutic strategy. Nonetheless paradoxically, reperfusion can further aggravate the injury through a series of reactions known as cerebral ischemia-reperfusion injury (CIRI). Mitochondria play a vital role in promoting nerve survival and neurological function recovery and mitochondrial dysfunction is considered one of the characteristics of CIRI. Neurons often die due to oxidative stress and an imbalance in energy metabolism following CIRI, and there is a strong association with mitochondrial dysfunction. Altered mitochondrial dynamics is the first reaction of mitochondrial stress. Mitochondrial dynamics refers to the maintenance of the integrity, distribution, and size of mitochondria as well as their ability to resist external stimuli through a continuous cycle of mitochondrial fission and fusion. Therefore, improving mitochondrial dynamics is a vital means of treating CIRI. This review discusses the relationship between mitochondria and CIRI and emphasizes improving mitochondrial dynamics as a potential therapeutic approach to improve the prognosis of CIRI.


Subject(s)
Brain Ischemia , Reperfusion Injury , Humans , Mitochondrial Dynamics , Reperfusion Injury/metabolism , Brain Ischemia/drug therapy , Ischemia , Oxidative Stress
3.
Chem Biol Interact ; 378: 110479, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37088170

ABSTRACT

Polymyxin B (PMB) is one of the most effective drugs for the treatment of multi-resistant and pan-resistant gram-negative infections. However, it can induce acute kidney injury (AKI), the mechanism of which has not yet been fully elucidated. In this study, RNA sequencing and in vitro and in vivo experiments demonstrated that PMB induced AKI by promoting ferroptosis. Moreover, the metallothionein-1 (MT-1) level was significantly increased in the AKI group and clinical cases revealed that iron and MT-1 levels in urine were significantly higher in patients with AKI than in those without AKI. To explore the mechanism of PMB induced ferroptosis, we silenced p53 in human kidney-2 (HK2) cells according to RNA sequencing, which showed that p53 was obviously enhanced in the PMB treated group. While PMB significantly enhanced Fe2+, lipid peroxidation, malondialdehyde (MDA), transferrin receptor protein 1 (TFR1), and arachidonate 12-lpoxygenase (ALOX12), decreased the survival rate, solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), and glutathione (GSH), downregulation of p53 reversed these effects, suggesting PMB induced ferroptosis by activating p53. Studies have shown p53 can promote ferroptosis by regulating the downstream factors SLC7A11 or TFR1. Further, we verified that silencing TFR1 expression as well as overexpression of SLC7A11 inhibited ferroptosis and significantly increased the survival rate of HK2 cells. Overall, PMB induces ferroptosis in renal tubular cells by activating p53 to reduce SLC7A11 expression and elevate TFR1, leading to AKI; MT-1 and iron levels in urine were significantly increased when PMB induced ferroptosis.


Subject(s)
Acute Kidney Injury , Ferroptosis , Humans , Polymyxin B , Tumor Suppressor Protein p53/genetics , Acute Kidney Injury/chemically induced , Glutathione , Iron , Metallothionein
4.
Neural Regen Res ; 18(7): 1512-1520, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36571356

ABSTRACT

CDGSH iron sulfur domain 2 can inhibit ferroptosis, which has been associated with cerebral ischemia/reperfusion, in individuals with head and neck cancer. Therefore, CDGSH iron sulfur domain 2 may be implicated in cerebral ischemia/reperfusion injury. To validate this hypothesis in the present study, we established mouse models of occlusion of the middle cerebral artery and HT22 cell models of oxygen-glucose deprivation and reoxygenation to mimic cerebral ischemia/reperfusion injury in vivo and in vitro, respectively. We found remarkably decreased CDGSH iron sulfur domain 2 expression in the mouse brain tissue and HT22 cells. When we used adeno-associated virus and plasmid to up-regulate CDGSH iron sulfur domain 2 expression in the brain tissue and HT22 cell models separately, mouse neurological dysfunction was greatly improved; the cerebral infarct volume was reduced; the survival rate of HT22 cells was increased; HT22 cell injury was alleviated; the expression of ferroptosis-related glutathione peroxidase 4, cystine-glutamate antiporter, and glutathione was increased; the levels of malondialdehyde, iron ions, and the expression of transferrin receptor 1 were decreased; and the expression of nuclear-factor E2-related factor 2/heme oxygenase 1 was increased. Inhibition of CDGSH iron sulfur domain 2 upregulation via the nuclear-factor E2-related factor 2 inhibitor ML385 in oxygen-glucose deprived and reoxygenated HT22 cells blocked the neuroprotective effects of CDGSH iron sulfur domain 2 up-regulation and the activation of the nuclear-factor E2-related factor 2/heme oxygenase 1 pathway. Our data indicate that the up-regulation of CDGSH iron sulfur domain 2 can attenuate cerebral ischemia/reperfusion injury, thus providing theoretical support from the perspectives of cytology and experimental zoology for the use of this protein as a therapeutic target in patients with cerebral ischemia/reperfusion injury.

5.
J Hematol Oncol ; 15(1): 174, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36482419

ABSTRACT

Many types of human cells self-destruct to maintain biological homeostasis and defend the body against pathogenic substances. This process, called regulated cell death (RCD), is important for various biological activities, including the clearance of aberrant cells. Thus, RCD pathways represented by apoptosis have increased in importance as a target for the development of cancer medications in recent years. However, because tumor cells show avoidance to apoptosis, which causes treatment resistance and recurrence, numerous studies have been devoted to alternative cancer cell mortality processes, namely necroptosis, pyroptosis, ferroptosis, and cuproptosis; these RCD modalities have been extensively studied and shown to be crucial to cancer therapy effectiveness. Furthermore, evidence suggests that tumor cells undergoing regulated death may alter the immunogenicity of the tumor microenvironment (TME) to some extent, rendering it more suitable for inhibiting cancer progression and metastasis. In addition, other types of cells and components in the TME undergo the abovementioned forms of death and induce immune attacks on tumor cells, resulting in enhanced antitumor responses. Hence, this review discusses the molecular processes and features of necroptosis, pyroptosis, ferroptosis, and cuproptosis and the effects of these novel RCD modalities on tumor cell proliferation and cancer metastasis. Importantly, it introduces the complex effects of novel forms of tumor cell death on the TME and the regulated death of other cells in the TME that affect tumor biology. It also summarizes the potential agents and nanoparticles that induce or inhibit novel RCD pathways and their therapeutic effects on cancer based on evidence from in vivo and in vitro studies and reports clinical trials in which RCD inducers have been evaluated as treatments for cancer patients. Lastly, we also summarized the impact of modulating the RCD processes on cancer drug resistance and the advantages of adding RCD modulators to cancer treatment over conventional treatments.


Subject(s)
Apoptosis , Neoplasms , Humans , Cell Death , Neoplasms/drug therapy , Copper , Ferroptosis , Pyroptosis , Necroptosis
6.
Signal Transduct Target Ther ; 7(1): 221, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35803926

ABSTRACT

Liquid-liquid phase separation (LLPS) is a novel principle for explaining the precise spatial and temporal regulation in living cells. LLPS compartmentalizes proteins and nucleic acids into micron-scale, liquid-like, membraneless bodies with specific functions, which were recently termed biomolecular condensates. Biomolecular condensates are executors underlying the intracellular spatiotemporal coordination of various biological activities, including chromatin organization, genomic stability, DNA damage response and repair, transcription, and signal transduction. Dysregulation of these cellular processes is a key event in the initiation and/or evolution of cancer, and emerging evidence has linked the formation and regulation of LLPS to malignant transformations in tumor biology. In this review, we comprehensively summarize the detailed mechanisms of biomolecular condensate formation and biophysical function and review the recent major advances toward elucidating the multiple mechanisms involved in cancer cell pathology driven by aberrant LLPS. In addition, we discuss the therapeutic perspectives of LLPS in cancer research and the most recently developed drug candidates targeting LLPS modulation that can be used to combat tumorigenesis.


Subject(s)
Neoplasms , Proteins , Biology , Humans , Neoplasms/genetics , Neoplasms/metabolism
7.
Cell Cycle ; 21(15): 1651-1661, 2022 08.
Article in English | MEDLINE | ID: mdl-35373707

ABSTRACT

Pannexin1 (Panx-1) is a gap junction channel protein that mediates the release of intracellular ATP during autophagy, and thus plays an important role in tumor cell apoptosis and chemo-resistance. However, the role of Panx-1 in cisplatin-resistance of testicular cancer cells remains unclear. We found that cisplatin-resistant I-10 testicular cancer cell lines (I-10/CDDP) autophagy-associated proteins (p62, p-mTOR, mTOR and LC3) exhibited high levels of autophagy in their expression, while LC3-II expression was more significantly in the presence of lysosomal degradation blocked by chloroquine (CQ). Xenograft models using I-10/CDDP cells with knockdown ATG5 and ATG7 were established in mouse models and showed blockade of autophagic flux and inhibition of tumor growth. In addition, inhibition of Panx-1 by carbenoxolone (CBX) and probenecid (PBN), as well as shRNA-mediated knockdown promoted autophagy in the I-10/CDDP cells, which was accompanied by a decrease in the levels of extracellular ATP. In contrast, overexpression of Panx-1 decreased autophagy of I-10/CDDP cells and increased extracellular ATP levels. To further determine the effect of panx-1-mediated ATP release on the autophagy of I-10/CDDP cells, apyrase was used to hydrolyze the extracellular ATP. Apyrase promoted autophagy in I-10/CDDP cells city by decreasing extracellular ATP, regardless of Panx-1 expression. This study demonstrated for the first time that Panx-1-mediated ATP release inhibits autophagy of I-10/CDDP cells, which provides a potential therapeutic strategy for cisplatin-resistant testicular cancer.


Subject(s)
Antineoplastic Agents , Cisplatin , Connexins , Nerve Tissue Proteins , Testicular Neoplasms , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Apyrase/pharmacology , Autophagy , Cell Line, Tumor , Cisplatin/pharmacology , Connexins/genetics , Drug Resistance, Neoplasm , Humans , Male , Mice , Neoplasms, Germ Cell and Embryonal , Nerve Tissue Proteins/genetics , TOR Serine-Threonine Kinases/metabolism , Testicular Neoplasms/drug therapy , Testicular Neoplasms/genetics
8.
Aging Dis ; 12(8): 2052-2068, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34881085

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease which significantly influences the life quality of patients. The protein α-synuclein plays an important driving role in PD occurrence and development. Braak's hypothesis suggests that α-synuclein is produced in intestine, and then spreads into the central nervous system through the vagus nerve. The abnormal expression of α-synuclein has been found in inflammatory bowel disease (IBD). Intestinal inflammation and intestinal dysbiosis have been involved in the occurrence and development of PD. The present review aimed to summarize recent advancements in studies focusing on intestinal inflammation and PD, especially the mechanisms through which link intestinal inflammation and PD. The intestinal dysfunctions such as constipation have been introduced as non-motor manifestations of PD. The possible linkages between IBD and PD, including genetic overlaps, inflammatory responses, intestinal permeability, and intestinal dysbiosis, are mainly discussed. Although it is not confirmed whether PD starts from intestine, intestinal dysfunction may affect intestinal microenvironment to influence central nervous system, including the α-synuclein pathologies and systematic inflammation. It is expected to develop some new strategies in the diagnosis and treatment of PD from the aspect of intestine. It may also become an exciting direction to find better ways to regulate the composition of gut microorganism to treat PD.

9.
Pharmacol Ther ; 226: 107859, 2021 10.
Article in English | MEDLINE | ID: mdl-33895184

ABSTRACT

Inflammatory bowel disease (IBD), which includes Crohn's disease and ulcerative colitis, is characterized by persistent inflammation in a hereditarily susceptible host. In addition to gastrointestinal symptoms, patients with IBD frequently suffer from extra-intestinal complications such as fibrosis, stenosis or cancer. Mounting evidence supports the targeting of cytokines for effective treatment of IBD. Cytokines can be included in a newly proposed classification "soluble ligands" that has become the third major target of human protein therapeutic drugs after enzymes and receptors. Soluble ligands have potential significance for research and development of anti-IBD drugs. Compared with traditional drug targets for IBD treatment, such as receptors, at least three factors contribute to the increasing importance of soluble ligands as drug targets. Firstly, cytokines are the main soluble ligands and targeting of them has demonstrated efficacy in patients with IBD. Secondly, soluble ligands are more accessible than receptors, which are embedded in the cell membrane and have complex tertiary membrane structures. Lastly, certain potential target proteins that are present in membrane-bound forms can become soluble following cleavage, providing further opportunities for intervention in the treatment of IBD. In this review, 49 drugs targeting 25 distinct ligands have been evaluated, including consideration of the characteristics of the ligands and drugs in respect of IBD treatment. In addition to approved drugs targeting soluble ligands, we have also assessed drugs that are in preclinical research and drugs inhibiting ligand-receptor binding. Some new types of targetable soluble ligands/proteins, such as epoxide hydrolase and p-selectin glycoprotein ligand-1, are also introduced. Targeting soluble ligands not only opens a new field of anti-IBD drug development, but the circulating soluble ligands also provide diagnostic insights for early prediction of treatment response. In conclusion, soluble ligands serve as the third-largest protein target class in medicine, with much potential for the drugs targeting them.


Subject(s)
Drug Delivery Systems , Inflammatory Bowel Diseases , Ligands , Cytokines , Humans , Inflammatory Bowel Diseases/drug therapy
10.
Oxid Med Cell Longev ; 2021: 9979707, 2021.
Article in English | MEDLINE | ID: mdl-35069968

ABSTRACT

Colorectal cancer (CRC) is a highly malignant tumor associated with poor prognosis, yet the molecular mechanisms are not fully understood. In this study, we showed that LYAR, a nucleolar protein, is expressed at a higher level in CRC tissue than in adjacent normal tissue and that LYAR expression is closely associated with distant CRC metastasis. LYAR not only significantly promotes the migration and invasion of CRC cells in vitro, but knockdown (KD) of LYAR in CRC cells also inhibits xenograft tumor metastasis in vivo. Microarray analysis of LYAR KD cells combined with a chromatin immunoprecipitation (ChIP) assay, gene reporter assay, and rescue experiment indicated that FSCN1 (encoding fascin actin-bundling protein 1 (Fascin-1)) serves as a novel key regulator of LYAR-promoted migration and invasion of CRC cells. Knockdown of FSCN1 significantly inhibits subcutaneous tumorigenesis of CRC cells and leads to the downregulation of FASN and SCD, genes encoding key enzymes in fatty acid synthesis. In summary, this study reveals a novel mechanism by which LYAR promotes tumor cell migration and invasion by upregulating FSCN1 expression and affecting fatty acid metabolism in CRC.


Subject(s)
Carrier Proteins/metabolism , Colorectal Neoplasms/genetics , DNA-Binding Proteins/metabolism , Fatty Acids/metabolism , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Transfection , Up-Regulation
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(4): 463-468, 2020 Apr 30.
Article in Chinese | MEDLINE | ID: mdl-32895131

ABSTRACT

OBJECTIVE: To investigate the protective effects of dexmedetomidine (DEX) against cerebral ischemia/reperfusion (I/R) injury in mice and its relation with mitochondrial fusion and fission. METHODS: Male ICR mice were randomly divided into sham-operated group, I/R group, I/R+DEX group and I/R+DEX+dorsomorphin group. Mouse models of cerebral I/R injury were established by modified thread occlusion of the middle cerebral artery. DEX (50 µg/kg) was injected intraperitoneally at 30 min before cerebral ischemia, which lasted for 1 h followed by reperfusion for 24 h. The neurobehavioral deficits of the mice were evaluated based on Longa's scores. The volume of cerebral infarction was detected by TTC staining. The changes in mitochondrial morphology of the brain cells were observed with transmission electron microscopy. Western blotting was performed to detect the expressions of phosphorylated AMP-activated protein kinase (p-AMPK), mitochondrial fusion protein (Mfn2) and mitochondrial fission protein (p-Drp1) in the brain tissues. RESULTS: DEX pretreatment significantly reduced the neurobehavioral score and the percent volume of cerebral infarction in mice with cerebral I/R injury. Treatment with dorsomorphin (an AMPK inhibitor) in addition to DEX significantly increased the neurobehavioral score and the percent volume of cerebral infarction in the mouse models. Transmission electron microscopy showed that DEX obviously reduced mitochondrial damage caused by cerebral I/R injury and restored mitochondrial morphology of the brain cells, and such effects were abolished by dorsomorphin treatment. Western blotting showed that DEX pretreatment significantly increased the expressions of p-AMPK and Mfn2 protein and decreased the expression of p-Drp1 protein in the brain tissue of the mice, and these changes were also reversed by dorsomorphin treatment. CONCLUSIONS: Preconditioning with DEX produces protective effects against cerebral I/R injury in mice possibly by activating AMPK signaling to regulate mitochondrial fusion and fission in the brain cells.


Subject(s)
Brain Ischemia , Reperfusion Injury , Animals , Dexmedetomidine , Male , Mice , Mice, Inbred ICR , Mitochondrial Dynamics
12.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(8): 1090-1096, 2020 Aug 30.
Article in Chinese | MEDLINE | ID: mdl-32895173

ABSTRACT

OBJECTIVE: To investigate the effect of down-regulation of pannexin 2 (Panx-2) channels on cisplatin-induced apoptosis in I-10 cells. METHODS: The expression of Panx-2 protein in testicular cancer cells was detected with Western blotting. The testicular cancer cell line I-10 was transfected with two short hairpin RNA (shRNA1 and shRNA2) via Lipofectamine2000, the empty vector (NC group) or Lipofectamine2000 (blank control group), and the changes in the expression of Panx-2 was detected with Western blotting. The effects of transfection with a Panx-2 inhibitor on surviving fraction of the cells treated with cisplatin (16 µmol/L) for 24 h, 48 h and 72 h was assessed with MTT assay, and the clonogenic capacity of the cells was evaluated with colony-forming assay. At 8 h after incubation with 16 µmol/L cisplatin, AnnexinV/PI double staining was used to detect the early apoptosis of the cells. After 24 h of treatment with 16 µmol/L cisplatin, the cells were examined for expressions of caspase-3, Bcl-2 and Bax using Western blotting. RESULTS: The expression of Panx-2 was significantly increased in cisplatin-resistant I-10/DDP (P < 0.001) cells and Tcam-2/DDP (P < 0.01) cells as compared with I-10 cells and Tcam-2 cells. Transfection of I-10 cells with shRNA1 and shRNA2 resulted in significantly decreased Panx-2 expression (P < 0.05) and significantly reduced cell surviving fraction (P < 0.001). In the presence of cisplatin, the cells in NC group showed a higher clonogenic efficiency than those in shRNA1 and shRNA2 groups (P < 0.001). The early-stage apoptosis rate of the cells in shRNA1 and shRNA2 groups were significantly higher than that in NC group (P < 0.01). Panx-2 knockdown in I-10 cells significantly increased caspase-3 and Bax expressions (P < 0.05) and significantly decreased the expression of Bcl-2 (P < 0.01). CONCLUSIONS: Down-regulation of Panx-2 channel enhances cisplatin-induced apoptosis in cultured testicular cancer cells.


Subject(s)
Testicular Neoplasms , Antineoplastic Agents , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cisplatin , Connexins , Down-Regulation , Drug Resistance, Neoplasm , Humans , Male
13.
Anticancer Drugs ; 31(2): 123-130, 2020 02.
Article in English | MEDLINE | ID: mdl-31815763

ABSTRACT

Cisplatin (CDDP) is the most effective chemotherapeutic drug against lung carcinoma. However, the emergence of resistant clones has severely limited its clinical application. We found that the cisplatin-resistant lung carcinoma cell line A549/CDDP had increased levels of the phosphorylated gap junction protein Cx43 and SRC tyrosine kinase, and low levels of total Cx43 protein and reduced gap junction formation. The SRC kinase inhibitor PP2 increased the expression of total Cx43 protein and enhanced cisplatin sensitivity, indicating that activated SRC kinase induces chemoresistance by decrease total Cx43 level. Furthermore, Cx43 gene silencing in the drug-resistant cell lines abrogated the sensitizing effect of PP2. Taken together, targeting SRC kinase by PP2 reverses cisplatin resistance by upregulating Cx43 protein levels, indicating a novel pathway of cisplatin resistance that may be amenable to therapeutic intervention.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis , Cell Proliferation , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins pp60(c-src)/genetics , Tumor Cells, Cultured
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(9): 1089-1093, 2019 Sep 30.
Article in Chinese | MEDLINE | ID: mdl-31640960

ABSTRACT

OBJECTIVE: To investigate the effect of connexin43 (Cx43) protein on autophagy in cisplatin (DDP)-resistant testicular cancer I-10 cells. METHODS: The expression of Cx43 proteins in testicular cancer I-10 cells and I-10/DDP cells were detected with Western blotting. I-10/DDP cells were transfected with a full- length mouse Cx43 vector (mCx43) via Lipofectamine2000, the empty vector or Lipofectamine2000 (blank control group), and the changes in the expressions of LC3 and p62 proteins were determined with Western blotting. mCherry-GFP-LC3B transfection and transmission electron microscopy were used to analyze the changes in autophagy of the cells with Cx43 overexpression. RESULTS: Cx43 was significantly decreased in I-10/DDP cells compared with I-10 cells (P < 0.01). Transfection of the I-10/DDP cells with mCx43 vector resulted in significantly increased Cx43 expression in the cells (P < 0.01) and caused significantly decreased expression of LC3-Ⅱ (P < 0.01) and increased expression of p62 (P < 0.05) as compared with the negative control cells. Both transmission electron microscopy and mCherry-GFP-LC3B transfection showed that the number of autophagosomes was obviously reduced in mCx43-transfected cells as compared with the negative control cells. CONCLUSIONS: Cx43 inhibits autophagy in cisplatin-resistant testicular cancer I-10 /DDP cells.


Subject(s)
Autophagy , Connexin 43/metabolism , Drug Resistance, Neoplasm , Testicular Neoplasms/pathology , Animals , Cell Line, Tumor , Cisplatin , Male , Mice , Testicular Neoplasms/metabolism
15.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(7): 797-803, 2019 Jul 30.
Article in Chinese | MEDLINE | ID: mdl-31340912

ABSTRACT

OBJECTIVE: To investigate the effect of SRC kinase inhibitor PP2 on the invasion and metastasis of lung cancer A549 cells and explore its molecular mechanism. METHODS: MTT assay was used to evaluate the inhibitory effect of PP2 on the proliferation of A549 cells. Cell scratch and Transwell assays were performed to assess the invasion and metastatic capacity of A549 cells after treatment with 1, 2, 4, 8, and 16 µmol/L PP2 for 24 h. Western blotting was used to detect the expressions of connexin43 (Cx43) and MMP-2 in the cells after small interfering RNA (siRNA)-mediated silencing or overexpression of Cx43; the changes in the cell invasion and metastasis in response to PP2 treatment after Cx43 silencing or overexpression were investigated. RESULTS: MTT assay showed that treatment with PP2 at 2, 4, 8, 16, and 32 µmol/L significantly inhibited the proliferation of A549 cells in a concentration-dependent manner. Treatments with PP2 at 1, 2, 4, 8, and 16 µmol/L for 24 h also concentration-dependently lowered the invasion and metastatic abilities of the cells (P < 0.05). At 4 and 8 µmol/L, PP2 significantly increased the expression level of Cx43 protein and decreased the expression level of MMP-2 protein. Overexpression of Cx43 significantly enhanced the inhibitory effect of PP2 on the cell invasion and metastasis, and Cx43 silencing significantly attenuated the inhibitory effect of PP2 (P < 0.05). CONCLUSIONS: PP2 treatment can suppress the invasion and metastasis of A549 cells in vitro possibly by modulating the expression of Cx43.


Subject(s)
Lung Neoplasms , A549 Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation , Connexin 43 , Humans , Neoplasm Invasiveness , Protein Kinase Inhibitors , src-Family Kinases
16.
Biomed Pharmacother ; 117: 109090, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31202174

ABSTRACT

Pannexin (Panx) plays a crucial role in several cellular processes such as immune cell death, cell proliferation, invasion, and migration, apoptosis, and autophagy. However, the role of Panx in regulating cell migration and invasion in testicular cancer remains to be elucidated. In the present study, we determined the correlation between Panx-1 channel function and migration and invasion in I-10 testicular cancer cells. Transwell and wound healing assays showed that inhibition of Panx-1 by carbenoxolone (CBX) and probenecid (PBN) attenuated the migration and invasion of testicular cancer cells in vitro. Moreover, knockdown of Panx-1 with short hairpin RNA (shRNA) remarkably decreased the migration and invasion ability of I-10 cells. In shRNA-transfected cells, extracellular ATP (released through Panx channel) was also found to be decreased. Similarly, overexpression of Panx-1 with mPanx-1 increased the migration and invasion ability of I-10 cells. Moreover, we found that in mPanx-1-transfected cells treated with U0126 (inhibitor of p-ERK1/2), the migration and invasion of I-10 cells were remarkably attenuated. Overall, increased Panx-1 promotes migration and invasion in testicular cancer cells, and the effect is probably be related with ERK1/2 kinase activity. Thus, Panx-1 can serve as a potential therapeutic target for the treatment of testicular cancer.


Subject(s)
Cell Movement/genetics , Connexins/genetics , MAP Kinase Signaling System/genetics , Neoplasm Invasiveness/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Nerve Tissue Proteins/genetics , Testicular Neoplasms/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carbenoxolone/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Neoplasms, Germ Cell and Embryonal/drug therapy , Probenecid/pharmacology , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Testicular Neoplasms/drug therapy
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 49(5): 700-705, 2018 Sep.
Article in Chinese | MEDLINE | ID: mdl-30378329

ABSTRACT

OBJECTIVE: To investigate the role of SRC kinase inhibitor PP2 in drug resistance to adriamycin (ADM) in breast cancer cells and invasion, metastasis of cells. METHODS: MTT assay was used to detect the inhibitory effect of ADM on MCF-7 and MCF-7/ADM cells. The 50% inhibitory concentration (IC50) and resistance index (RI) of cells were calculated. The expression of MDR1, connexin 43 (Cx43) and SRC proteins in breast cancer cells were detected by Western blot assay. Transwell experiment and cell scratch test were used to determine the invasion and migration of cells respectively [MCF-7, MCF-7/ADM, PP2 (1, 2, 4 µmol/L)]. Standard colony formation assay was used to detect the cytotoxicity effect of 4 µmol/L PP2 pretreatment on ADM. RESULTS: ADM inhibited the proliferation of MCF-7 more than MCF-7/ADM cells (P<0.01). The IC50 of MCF-7/ADM cells was 24.55 µmol/L, the IC50 of MCF-7/ADM cells was 770.57 µmol/L, the RI was 31. Compared with MCF-7 cells, expressions of the multidrug resistance proteins MDR1 and SRC were significantly increased (P<0.01). The invasion and migration ability of the MCF-7/ADM cells was stronger than that of the sensitive cells (P<0.01). When MCF-7/ADM was exposed to SRC inhibitor PP2, the invasion and metastasis ability of cells were inhibited (P<0.01) and the rate of colony formation was decreased, that is, more sensitivity to ADM (P<0.01). CONCLUSION: The resistance of MCF-7 to ADM is accompanied by increased expression of SRC. SRC inhibitor PP2 can reduce the cell resistance, ability of invasion and metastasis.


Subject(s)
Breast Neoplasms/enzymology , Doxorubicin/metabolism , Drug Resistance, Neoplasm , Pyrimidines/pharmacology , src-Family Kinases/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Connexin 43/metabolism , Humans , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Metastasis
18.
Biomed Pharmacother ; 98: 826-833, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29571253

ABSTRACT

The effect of gap junction intercellular communication composed of connexin on cancer invasion/metastasis has been thoroughly explored; however, its effect on testicular cancer resistanced to chemotherapy is still unclear. In this study, we found that the capability of invasion and migration of I-10/DDP (cisplatin (DDP)-resistance) cells were elevated. Furthermore, the expression of Cx43 and the function of gap junction (GJ) in I-10/DDP cells were decreased compared with parental I-10 cells. Pharmacological inhibition of GJs by oleamide (Olea) enhanced invasion and migration. However, enhancement of GJs by retinoic acid (RA) decreased invasion and migration of I-10/DDP cells. To further clarify the invasion/migration inhibited effect of GJ in the testicular cancer resistanced to DDP, GJ function was modulated by overexpression and knockdown of Cx43 expression. Overexpression of Cx43 reduced invasion and migration of I-10/DDP cells. Conversely, knockdown of Cx43 expression increased invasion and migration of I-10/DDP cells. In summary, GJ composed of Cx43 inhibits I-10/DDP cells invasion and migration, and it may become the potential therapeutic target for testicular cancer chemotherapy.


Subject(s)
Cisplatin/pharmacology , Connexin 43/metabolism , Drug Resistance, Neoplasm , Gap Junctions/metabolism , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Animals , Cell Communication/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm/drug effects , Gap Junctions/drug effects , Gene Knockdown Techniques , Gene Silencing/drug effects , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis
19.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(7): 780-786, 2018 Jul 30.
Article in Chinese | MEDLINE | ID: mdl-33168517

ABSTRACT

OBJECTIVE: To observe the effect of functional modulation of gap junctions (GJ) on the antitumor effect of adriamycin in breast cancer cells positive for estrogen receptor (ER). METHODS: The inhibitory effect of 0 to 24.0 µmol/L adriamycin on the surviving fraction of ER-positive human breast cancer MCF-7 cells and ER-negative MDA-MB-231 cells was assessed with MTT assay; Western blotting and immunofluorescence assay were used to detect the expressions of Cx43 total protein and membrane protein in the cells. A parachute assay was used to evaluate the function of the GJ in MCF-7 cells. The cytotoxic effect of adriamycin was observed in the cells treated with retinoic acid (RA) for enhancing GJ function, in cells treated with oleamide and 18-α- glycyrrhizic acid (18-α-ga) for inhibiting GJ function, and also in cells transfected with Cx43siRNA for Cx43 knockdown. RESULTS: ER-positive MCF-7 cells expressed a significantly higher level of Cx43 with stronger GJ function than ER-negative MDA- MB-231 cells. Adriamycin significantly inhibited the proliferation of MCF-7 cells (P < 0.01), and RA treatment further increased the cytotoxicity of adriamycin (P < 0.01) while oleamide and 18-α-GA obviously attenuated the cytotoxicity of adriamycin (P < 0.01). In the cells with Cx43 knockdown, the expressions of total Cx43 protein and Cx43 on the membrane were significantly reduced, the function of GJ was attenuated, and the cytotoxicity of adriamycin was significantly decreased (P < 0.01). CONCLUSIONS: ER-positive breast cancer cells have stronger Cx43 expressions and GJ function than the ERnegative cells. The cytotoxicity of adriamycin against the breast cancer cells can be strengthened by enhancing GJ function and attenuated by inhibiting GJ function. Cx43 silencing inhibits the function of GJ to lower the cytotoxicity of adriamycin in human breast cancer MCF-7 cells.

20.
Zhonghua Nan Ke Xue ; 24(9): 776-781, 2018 Sep.
Article in Chinese | MEDLINE | ID: mdl-32212454

ABSTRACT

OBJECTIVE: To investigate the expressions of the pannexin (Panx) proteins in I-10 Leydig tumor cells and TM3 Leydig cells and their regulatory effect on the Panx channel function in mice. METHODS: The expressions of the Panx-1 and Panx-2 proteins in the mouse Leydig tumor cells were determined by Western blot. The I-10 Leydig tumor cells were treated with carbenoxolone (CBX) at 100 µmol/L or probenecid (PBN) at 200 µmol/L, the fluorescence resonance energy transfer (FRET) detected by time-lapse fluorescence imaging, and the extracellular adenosine 5'-triphosphate (eATP) level measured with the commercial detection kit. Molecular biological methods were used to interfere with shRNA and overexpress mPanx-1 the Panx-1 gene and regulate the expression and function of the Panx-1 protein. RESULTS: The expressions of Panx-1 (ï¼»289.5 ± 55.8ï¼½%) and Panx-2 (ï¼»264.5 ± 24.6ï¼½%) were significantly increased in the I-10 Leydig tumor cells as compared with those in the normal TM3 Leydig cells (both P < 0.05). FRET was remarkably reduced after treated with CBX (ï¼»87.5 ± 17.7ï¼½%) and PBN (ï¼»89.3 ± 14.3ï¼½%) in comparison with that in the control group (both P < 0.01). At 8, 16 and 24 hours, the eATP level was decreased by (57.3 ± 7.2)%, (56.4 ± 9.6)% and (63.4 ± 6.4)% in the CBX group (P < 0.01) and (61.7 ± 2.5)%, (35.8 ± 1.6)% and (13.5 ± 8.3)% in the PBN group (P < 0.01). Molecular biological treatment down-regulated the expression of Panx-1 by (38.3 ± 5.2)% and (31.8 ± 5.1)% in the shRNA1 and shRNA2 groups, respectively (both P < 0.01), but up-regulated that of Panx-1 by (128.4 ± 7.5)% in the mPanx-1 group (P < 0.01) as compared with the negative control. FRET was reduced by (72.4 ± 39.4)% in the shRNA group (P < 0.01) and the eATP level by (14.7 ± 0.1)%, (13.7 ± 0.3)% and (13.1 ± 0.3)% at 8, 16 and 24 hours, respectively (P < 0.01) while FRET elevated by (122.5 ± 17.1)% in the mPanx-1 group (P < 0.01) and the eATP level by (886.1 ± 82.1)%, (885.8 ± 83.3)% and (841.5 ± 21.8)% at 8, 16 and 24 hours, respectively (P < 0.01). CONCLUSIONS: The expressions of Panx-1 and Panx-2 are increased in I-10 mouse Leydig tumor cells, and inhibiting the Panx channel with CBX, PBN and shRNA reduces FRET and the eATP level in the I-10 cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...