Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 974: 176611, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38663540

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disorder of the gastrointestinal tract for which treatment options remain limited. In this study, we used a dual-luciferase-based screening of an FDA-approved drug library, identifying Bazedoxifene (BZA) as an inhibitor of the NF-κB pathway. We further investigated its therapeutic effects in a dextran sodium sulfate (DSS)-induced colitis model and explored its impact on gut microbiota regulation and the underlying molecular mechanisms. Our results showed that BZA significantly reduced DSS-induced colitis symptoms in mice, evidenced by decreased colon length shortening, lower histological scores, and increased expression of intestinal mucosal barrier-associated proteins, such as Claudin 1, Occludin, Zo-1, Mucin 2 (Muc2), and E-cadherin. Used independently, BZA showed therapeutic effects comparable to those of infliximab (IFX). In addition, BZA modulated the abundance of gut microbiota especially Bifidobacterium pseudolongum, and influenced microbial metabolite production. Crucially, BZA's alleviation of DSS-induced colitis in mice was linked to change in gut microbiota composition, as evidenced by in vivo gut microbiota depletion and fecal microbiota transplantation (FMT) mice model. Molecularly, BZA inhibited STAT3 and NF-κB activation in DSS-induced colitis in mice. In general, BZA significantly reduced DSS-induced colitis in mice through modulating the gut microbiota and inhibiting STAT3 and NF-κB activation, and its independent use demonstrated a therapeutic potential comparable to IFX. This study highlights gut microbiota's role in IBD drug development, offering insights for BZA's future development and its clinical applications.


Subject(s)
Colitis , Dextran Sulfate , Gastrointestinal Microbiome , NF-kappa B , STAT3 Transcription Factor , Signal Transduction , Animals , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/microbiology , Colitis/pathology , Gastrointestinal Microbiome/drug effects , Mice , Signal Transduction/drug effects , Indoles/pharmacology , Indoles/therapeutic use , Mice, Inbred C57BL , Disease Models, Animal , Colon/drug effects , Colon/pathology , Colon/metabolism , Colon/microbiology , Male , Humans
2.
Water Res ; 255: 121510, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38555780

ABSTRACT

Despite the frequent detection of fluorinated liquid-crystal monomers (FLCMs) in the environment, the level of understanding of their fate, toxicity, and transformation remains insufficient. Herein, we investigated the degradation kinetics and mechanism of an FLCM (4-cyano-3-fluorophenyl 4-ethylbenzoate, CEB-F) under ultraviolet (UV) photolysis in aquatic environment. Our findings demonstrated that the UV photolysis of CEB-F followed first-order kinetics. Photodegradation products were identified using liquid chromatography with mass spectrometry, and detailed reaction pathways were proposed. It is postulated that through the attack of reactive oxygen species, hydroxylation, and CO/C-F bond cleavage, CEB-F gradually degraded into small molecular compounds, releasing fluorine ions. Acute immobilization tests with Daphnia magna (D. magna) revealed significant acute toxicity of CEB-F, with LC50 values ranging from 1.023 to 0.0536 µM over 24 to 96 h, emphasizing the potential high risk of FLCMs in aquatic ecosystems if inadvertently discharged. Interestingly, we found that the toxicity of CEB-F photolysis reaction solutions was effectively reduced. Through catalase and acetylcholinesterase activities analysis along with molecular docking simulation, we proposed differences in the underlying toxicity mechanisms of CEB-F and its photolysis products to D. magna. These findings highlight the potential harmful effects of FLCMs on aquatic ecosystems and enrich our understanding of the photolysis behavior of FLCMs.

3.
Biol Pharm Bull ; 46(11): 1569-1575, 2023.
Article in English | MEDLINE | ID: mdl-37914359

ABSTRACT

Ovarian cancer (OC) is one of the most common and high mortality type of cancer among women worldwide. The majority of patients with OC respond to chemotherapy initially; however, most of them become resistant to chemotherapy and results in a high level of treatment failure in OC. Therefore, novel agents for the treatment of OC are urgently required. Benzimidazole anthelmintics might have the promising efficacy for cancer therapy as their selectively binding activity to ß-tubulin. Recent study has shown that one of the benzimidazole anthelmintics oxfendazole inhibited cell growth of non-small cell lung cancer cells, revealing its anti-cancer activity; however, the pharmacological action and detailed mechanism underlying the effects of oxfendazole on OC cells remain unclear. Therefore, the present study investigated the cytotoxic effects of oxfendazole on OC cells. Our results demonstrated that oxfendazole significantly decreased the viability of OC cells. Oxfendazole inhibited the proliferation, induced G2/M phase arrest and apoptotic cell death in A2780 cells. The c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK) pathway was activated and reactive oxygen species (ROS) generation was increased in OC cells treated with oxfendazole; oxfendazole-induced apoptosis was notably abrogated when co-treated with JNK inhibitor SP600125 and ROS scavenger N-acetyl-L-cysteine (NAC), indicating that JNK/MAPK pathway activation and ROS accumulation was associated with the oxfendazole-induced apoptosis of OC cells. Moreover, oxfendazole could also induce the proliferation inhibition and apoptosis of cisplatin resistant cells. Collectively, these results revealed that oxfendazole may serve as a potential therapeutic agent for the treatment of OC.


Subject(s)
Anthelmintics , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Ovarian Neoplasms , Humans , Female , JNK Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Ovarian Neoplasms/drug therapy , Apoptosis , Benzimidazoles/pharmacology , MAP Kinase Signaling System , Anthelmintics/pharmacology
4.
Environ Sci Technol ; 57(32): 11977-11987, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37526086

ABSTRACT

While carbon dots (CDs) have the potential to support the agricultural revolution, it remains obscure about their environmental fate and bioavailability by plants. Fungal laccase-mediated biotransformation of carbon nanomaterials has received little attention despite its known capacity to eliminate recalcitrant contaminants. Herein, we presented the initial investigation into the transformation of CDs by fungal laccase. The degradation rates of CDs were determined to be first-order in both substrate and enzyme. Computational docking studies showed that CDs preferentially bonded to the pocket of laccase on the basal plane rather than the edge through hydrogen bonds and hydrophobic interactions. Electrospray ionization-Fourier transform-ion cyclotron resonance mass spectrometry (ESI-FT-ICR MS) and other characterizations revealed that the phenolic/amino lignins and tannins portions in CDs are susceptible to laccase transformation, resulting in graphitic structure damage and smaller-sized fragments. By using the 13C stable isotope labeling technique, we quantified the uptake and translocation of 13C-CDs by mung bean plants. 13C-CDs (10 mg L-1) accumulated in the root, stem, and leaf were estimated to be 291, 239, and 152 µg g-1 at day 5. We also evidenced that laccase treatment alters the particle size and surface chemistry of CDs, which could facilitate the uptake of CDs by plants and reduce their nanotoxicity to plants.


Subject(s)
Carbon , Laccase , Laccase/chemistry , Laccase/metabolism , Biodegradation, Environmental , Mass Spectrometry , Biotransformation , Trametes/metabolism
5.
iScience ; 26(3): 106140, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36879799

ABSTRACT

Antibody-secreting B cells have long been considered the central element of gut homeostasis; however, tumor-associated B cells in human colorectal cancer (CRC) have not been well characterized. Here, we show that the clonotype, phenotype, and immunoglobulin subclasses of tumor-infiltrating B cells have changed compared to adjacent normal tissue B cells. Remarkably, the tumor-associated B cell immunoglobulin signature alteration can also be detected in the plasma of patients with CRC, suggesting that a distinct B cell response was also evoked in CRC. We compared the altered plasma immunoglobulin signature with the existing method of CRC diagnosis. Our diagnostic model exhibits improved sensitivity compared to the traditional biomarkers, CEA and CA19-9. These findings disclose the altered B cell immunoglobulin signature in human CRC and highlight the potential of using the plasma immunoglobulin signature as a non-invasive method for the assessment of CRC.

6.
Front Pharmacol ; 12: 729367, 2021.
Article in English | MEDLINE | ID: mdl-34776955

ABSTRACT

There is an urgent need for novel agents for colorectal cancer (CRC) due to the increasing number of cases and drug-resistance related to current treatments. In this study, we aim to uncover the potential of chaetocin, a natural product, as a chemotherapeutic for CRC treatment. We showed that, regardless of 5-FU-resistance, chaetocin induced proliferation inhibition by causing G2/M phase arrest and caspase-dependent apoptosis in CRC cells. Mechanically, our results indicated that chaetocin could induce reactive oxygen species (ROS) accumulation and activate c-Jun N-terminal kinase (JNK)/c-Jun pathway in CRC cells. This was confirmed by which the JNK inhibitor SP600125 partially rescued CRC cells from chaetocin induced apoptosis and the ROS scavenger N-acetyl-L-cysteine (NAC) reversed both the chaetocin induced apoptosis and the JNK/c-Jun pathway activation. Additionally, this study indicated that chaetocin could down-regulate the expression of CD47 at both mRNA and protein levels, and enhance macrophages phagocytosis of CRC cells. Chaetocin also inhibited tumor growth in CRC xenograft models. In all, our study reveals that chaetocin induces CRC cell apoptosis, irrelevant to 5-FU sensitivity, by causing ROS accumulation and activating JNK/c-Jun, and enhances macrophages phagocytosis, which suggests chaetocin as a candidate for CRC chemotherapy.

7.
Cell Death Dis ; 12(6): 576, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34088891

ABSTRACT

Cancer-secreted exosomes are critical mediators of cancer-host crosstalk. In the present study, we showed the delivery of miR-21-5p from colorectal cancer (CRC) cells to endothelial cells via exosomes increased the amount of miR-21-5p in recipient cells. MiR-21-5p suppressed Krev interaction trapped protein 1 (KRIT1) in recipient HUVECs and subsequently activated ß-catenin signaling pathway and increased their downstream targets VEGFa and Ccnd1, which consequently promoted angiogenesis and vascular permeability in CRC. A strong inverse correlation between miR-21-5p and KRIT1 expression levels was observed in CRC-adjacent vessels. Furthermore, miR-21-5p expression in circulating exosomes was markedly higher in CRC patients than in healthy donors. Thus, our data suggest that exosomal miR-21-5p is involved in angiogenesis and vascular permeability in CRC and may be used as a potential new therapeutic target.


Subject(s)
Colorectal Neoplasms/blood supply , KRIT1 Protein/metabolism , MicroRNAs/metabolism , Animals , Capillary Permeability , Cell Movement/physiology , Cell Proliferation/physiology , Chick Embryo , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Exosomes/genetics , Exosomes/metabolism , HCT116 Cells , HT29 Cells , Heterografts , Human Umbilical Vein Endothelial Cells , Humans , KRIT1 Protein/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Tumor Microenvironment
8.
Aging (Albany NY) ; 12(10): 8893-8912, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32461377

ABSTRACT

Chemoresistance remains a significant obstacle for effective adriamycin (ADR) treatment in breast cancer. Recent efforts have revealed that long noncoding RNAs (lncRNAs) play a crucial role in cancer biology, including chemoresistance. We identified the lncRNA LOC645166 was upregulated in adriamycin resistant-breast cancer cells by Microarray analysis, which was further confirmed in the tissues of nonresponsive patients by reverse transcription-quantitative polymerase chain reaction (RT-qPCR), western blotting, and immunohistochemical assays. Downregulation of lncRNA LOC645166 increased cell sensitivity to adriamycin both in vitro and in vivo. In contrast, upregulation of lncRNA LOC645166 strengthened the tolerance of breast cancer cells to adriamycin. Chromatin immunoprecipitation (ChIP) and RNA binding protein immunoprecipitation (RIP) demonstrated that lncRNA LOC645166 could increase the expression of GATA binding protein 3 (GATA3) via binding with nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), leading to the activation of STAT3 and promoting chemoresistance in breast cancer. Together, the present study suggested that lncRNA LOC645166 mediated adriamycin chemoresistance in breast cancer by regulating GATA3 via NF-κB.


Subject(s)
Breast Neoplasms , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , GATA3 Transcription Factor , RNA, Long Noncoding , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
9.
Medicine (Baltimore) ; 98(44): e17686, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31689791

ABSTRACT

RATIONALE: Ependymomas are neuroepithelial tumors that typically occur in the central nervous system. Ependymomas arising in the mediastinum are exceedingly rare, with only approximately 9 isolated cases reported in the literature to date. PATIENT CONCERNS: A 35-year-old woman was referred to our hospital with complaints of progressive back pain for 3 months. Physical examination revealed decreased breathing sounds and tenderness. Contrast-enhanced computed tomography showed a soft tissue mass with heterogeneous enhancement in the right posterior mediastinum. DIAGNOSES: The diagnosis of primary mediastinal ependymomas (PMEs) was confirmed by postoperative histopathologic examination. INTERVENTIONS AND OUTCOMES: The patient underwent surgical resection of the tumor and experienced local recurrence with neck metastasis 2 years postoperatively. She underwent reoperation for the recurrent tumors and received postoperative radiotherapy and adjuvant chemotherapy. Two years later, the patient is doing well, with no evidence of tumor progression or recurrence. LESSONS: Since PMEs are exceedingly rare, treatment options are limited. Surgical resection seems to be the mainstay of treatment. Further evidence-based studies are required to prove the benefit of radiotherapy and chemotherapy in the treatment of PMEs.


Subject(s)
Ependymoma/diagnosis , Mediastinal Neoplasms/diagnosis , Adult , Diagnosis, Differential , Ependymoma/diagnostic imaging , Ependymoma/therapy , Female , Humans , Mediastinal Neoplasms/diagnostic imaging , Mediastinal Neoplasms/therapy , Neoplasm Recurrence, Local , Tomography, X-Ray Computed
10.
Cell Death Dis ; 10(11): 809, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649256

ABSTRACT

Novel drugs are urgently needed for gastric cancer (GC) treatment. The thioredoxin-thioredoxin reductase (TRX-TRXR) system has been found to play a critical role in GC tumorigenesis and progression. Thus, agents that target the TRX-TRXR system may be highly efficacious as GC treatments. In this study, we showed that chaetocin, a natural product isolated from the Chaetomium species of fungi, inhibited proliferation, induced G2/M phase arrest and caspase-dependent apoptosis in both in vitro and in vivo models (cell xenografts and patient-derived xenografts) of GC. Chaetocin inactivated TRXR-1, resulting in the accumulation of reactive oxygen species (ROS) in GC cells; overexpression of TRX-1 as well as cotreatment of GC cells with the ROS scavenger N-acetyl-L-cysteine attenuated chaetocin-induced apoptosis; chaetocin-induced apoptosis was significantly increased when GC cells were cotreated with auranofin. Moreover, chaetocin was shown to inactivate the PI3K/AKT pathway by inducing ROS generation; AKT-1 overexpression also attenuated chaetocin-induced apoptosis. Taken together, these results reveal that chaetocin induces the excessive accumulation of ROS via inhibition of TRXR-1. This is followed by PI3K/AKT pathway inactivation, which ultimately inhibits proliferation and induces caspase-dependent apoptosis in GC cells. Chaetocin therefore may be a potential agent for GC treatment.


Subject(s)
Cell Death/drug effects , Stomach Neoplasms/drug therapy , Thioredoxin Reductase 1/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Auranofin/pharmacology , Cell Line, Tumor , Heterografts , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Reactive Oxygen Species/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Thioredoxin Reductase 1/genetics , Thioredoxins/genetics
11.
Oncol Lett ; 18(2): 1915-1921, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31423261

ABSTRACT

Ovarian cancer (OC) is one of the most common types of cancer among women worldwide. The majority of patients with OC respond to current chemotherapy approaches initially; however, patients are likely to experience cancer recurrence and become resistant to the chemotherapy. Therefore, novel agents for the treatment of OC are urgently required. Chaetocin, a natural product isolated from Chaetomium fungi, has been reported to exhibit anticancer activity against various types of cancer; however, the pharmacological action and detailed mechanism underlying the effects of chaetocin on OC cells remain unclear. Therefore, the present study investigated the cytotoxic effects of chaetocin on OC cells. A Cell Counting kit-8 assay was used to study cell viability, a colony formation assay was used to assess cell proliferation, flow cytometry was used to detect apoptosis, cell cycle and reactive oxygen species (ROS) generation, and western blotting was used to determine the protein levels of poly (ADP-ribose) polymerase, caspase-3 and cleaved-caspase-3. The results demonstrated that chaetocin significantly decreased the viability of OC cells. Chaetocin inhibited the proliferation and induced G2/M phase arrest of the OVCAR-3 OC cell line. Additionally, chaetocin induced apoptotic cell death in OVCAR-3 cells via the caspase pathway. It was observed that chaetocin induced the accumulation of ROS in OVCAR-3 cells. Treatment with the ROS scavenger N-acetyl-L-cysteine reversed the apoptotic effects and activation of the caspase pathway induced by chaetocin. Collectively, these results revealed that chaetocin suppressed the proliferation and promoted the caspase-dependent apoptosis of OC cells by increasing the levels of ROS. Therefore, chaetocin may serve as a potential therapeutic agent for the treatment of OC.

12.
Biochem Biophys Res Commun ; 498(4): 1002-1008, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29550489

ABSTRACT

Long non-coding RNA (lncRNA) is an important member of non-coding RNA family and emerging evidence has indicated that it plays a pivotal role in many physiological and pathological processes. The lncRNA X inactive specific transcript (XIST) is a potential tumour suppressor in some types of cancers. However, the expression and function of XIST in breast cancer remain largely unclear. The objective of this study was to evaluate the expression and biological role of XIST in breast cancer. The results showed that XIST was significantly down-regulated in breast cancer tissues and cell lines. Further functional analysis indicated that overexpression of XIST remarkably inhibited breast cancer cell growth, migration, and invasion. The results of luciferase reporter assays verified that miR-155 was a direct target of XIST in breast cancer. Moreover, caudal-type homeobox 1 (CDX1) was identified as a direct target of miR-155 and miR-155/CDX1 rescued the effects of XIST in breast cancer cells. Taken together, our results suggest that XIST is down-regulated in breast cancer and suppresses breast cancer cell growth, migration, and invasion via the miR-155/CDX1 axis.


Subject(s)
Breast Neoplasms/genetics , Homeodomain Proteins/physiology , MicroRNAs/physiology , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/physiology , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , RNA, Long Noncoding/analysis , Tumor Cells, Cultured
16.
Stem Cells Dev ; 21(14): 2703-17, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22468883

ABSTRACT

Bone marrow stem cells (BMSCs) have been shown to improve neurological function recovery in cerebral ischemia. Hypoxia-inducible factor-1 (HIF-1) α-AA is a more stable mutant form of HIF-1α, which is a crucial oxygen-sensitive regulator. To investigate the protective effects of HIF-1α-AA-modified BMSCs on neuron survival in cerebral ischemia models, we co-cultured HIF-1α-AA-modified BMSCs with neuron-like cells (PC12 cells) and observed a significant increase in the release of vascular endothelial growth factor (VEGF) from BMSCs, the decreased PC12 cell apoptosis, and the upregulation of Survivin expression reduced by hypoxia in PC12 cells compared to enhanced green fluorescent protein (EGFP) BMSCs. In addition, to explore whether VEGF secreted by HIF-1α-AA-modified BMSCs plays an important role in preventing hypoxia-induced apoptosis and the possible mechanism involved, exogenous VEGF were applied and the similar protective effects on PC12 cells were observed in vitro. Furthermore, hypoxia reduced the expression of phosphorylated Akt and phosphorylated FoxO1, whereas the administration of VEGF reversed these changes. Transfection of FoxO1 H215R, a DNA-binding mutant, abrogated the inhibitory ability on Survivin promoter activity, whereas FoxO1 AAA, the active form of FoxO1, presented further repression on Survivin promoter, indicating that FoxO1 directly binds on Survivin promoter as a transcriptional repressor and that phosphorylation status of FoxO1 affects its inhibition on the Survivin promoter. Transplantation of HIF-1α-AA-modified BMSCs after cerebral ischemia in vivo sufficiently reduced neurons apoptosis, decreased cerebral infarction volume, and induced a significant improvement on the modified neurological severity score compared to the EGFP BMSCs group. In conclusion, HIF-1α-AA-modified MSCs showed an obvious protective effect on neuron-like cells or neuron after ischemia in vitro and in vivo, at least in part, through the VEGF/PI3K/Akt/FoxO1 pathway.


Subject(s)
Apoptosis , Bone Marrow Cells/metabolism , Cerebral Infarction/therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Stem Cells/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation/methods , Cell Hypoxia , Cerebral Infarction/metabolism , Cobalt/adverse effects , Coculture Techniques , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Green Fluorescent Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lentivirus/metabolism , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Rats , Rats, Sprague-Dawley , Signal Transduction , Stem Cells/cytology , Survivin , Transfection , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
17.
Cell Biol Int ; 33(3): 411-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19356708

ABSTRACT

Reactive oxygen species (ROS) play essential roles in apoptosis and in the regulation of several transcription factors under both physiological and pathological conditions. However, the effects of ROS on MSCs are not well known, and therefore we have investigated the effects of preconditioning with hydrogen peroxide (H(2)O(2)) on the level of expression of the chemokine receptor, CXCR4, stromal cell-derived factor-1alpha (SDF-1alpha)-dependent migration and apoptosis in MSCs. Preconditioning with 20microM H(2)O(2) significantly increased the level of expression of CXCR4 mRNA and protein, and MSCs migration toward SDF-1alpha; increased expression of CXCR4 and SDF-1alpha-induced MSCs migration was attenuated by extracellular signal-regulated kinase (ERK) inhibitor PD98059. Preconditioning with 20microM H(2)O(2) significantly protected MSCs against apoptosis induced by 500microM H(2)O(2). These results suggest that preconditioning with H(2)O(2) can increase MSCs migration toward SDF-1alpha and protect MSCs against apoptosis.


Subject(s)
Apoptosis , Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Reactive Oxygen Species/metabolism , Animals , Cell Movement , Chemokine CXCL12/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hydrogen Peroxide/metabolism , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...