Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Br J Cancer ; 116(8): 1027-1036, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28267710

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common types of malignant tumour and has poor prognosis. Currently, systematic chemotherapy is the only approach to prolong survival. Thus the development of new treatment regimens is urgently needed to improve the therapeutic efficacy. Our study intended to assess the combination of dasatinib and irinotecan against HCC and made an effort to develop a potential medical choice for advanced HCC patients. METHODS: We used SRB colorimetric assay and clonogenic assay to assess antitumour effect in vitro and HCC xenograft model to assess antitumour effect in vivo. We applied flow cytometry and western blotting to explore the mechanism of the combined therapy. Knockdown and overexpression of PLK1 are also applied for validation. RESULTS: We confirmed that dasatinib has synergistic effect with irinotecan (or SN38) on HCC both in vitro and in vivo. The effect is due to arisen apoptosis rate of HCC cells that is accompanied by mitochondria dysfunction. The enhanced antitumour efficacy of SN38 could be explained by additional inhibition of PLK1, which is triggered by dasatinib. Unlike existed PLK1 inhibitors, dasatinib does not inhibit PLK1 activity in a direct way. Instead, we found that dasatinib reduces PLK1 level by interfering with its protein synthesis progress. We validated that this kind of downregulation of PLK1 level has a key role in the synergistic effect of the two agents. CONCLUSIONS: Dasatinib is able to reinforce the anti-HCC efficacy of irinotecan/SN38 by downregulation of PLK1 synthesis. The combination of the two agents might be a potential medical choice for HCC therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , Drug Synergism , Liver Neoplasms/drug therapy , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blotting, Western , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Dasatinib/administration & dosage , Female , Flow Cytometry , Humans , Irinotecan , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
2.
BMC Cancer ; 16: 2, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728137

ABSTRACT

BACKGROUND: Chemotherapy is the only therapy option for the majority of AML patients, however, there are several limitations for this treatment. Our aim was to find a new chemotherapy strategy that is more effective and less toxic. METHODS: MTT assays and a xenograft mouse model were employed to evaluate the synergistic activity of all-trans retinoic acid (ATRA) combined with topotecan (TPT). Drug-induced DNA damage and apoptosis were determined by flow cytometry analysis with PI and DAPI staining, the comet assay and Western blots. Short hairpin RNA (shRNA) and a RARα plasmid were used to determine whether RARα expression influenced DNA damage and apoptosis. RESULTS: We found that ATRA exhibited synergistic activity in combination with Topotecan in AML cells, and the enhanced apoptosis induced by Topotecan plus ATRA resulted from caspase pathway activation. Mechanistically, ATRA dramatically down regulated RARα protein levels and led to more DNA damage and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of Topotecan combined with ATRA was further validated in the HL60 xenograft mouse model. CONCLUSIONS: Our data demonstrated, for the first time, that the combination of TPT and ATRA showed potential benefits in AML, providing a novel insight into clinical treatment strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Synergism , Leukemia, Myeloid, Acute/drug therapy , Topotecan/administration & dosage , Tretinoin/administration & dosage , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Gene Expression Regulation, Leukemic/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , RNA, Small Interfering , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Xenograft Model Antitumor Assays
3.
Biomed Pharmacother ; 173: 116306, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401520

ABSTRACT

Clinical resistance to EGFR tyrosine kinase inhibitors in non-small-cell lung cancer (NSCLC) remains a significant challenge. Recent studies have indicated that the number of myeloid-derived suppressor cells (MDSCs) increases following gefitinib treatment, correlating with a poor patient response in NSCLC. Our study revealed that gefitinib treatment stimulates the production of CCL2, which subsequently enhances monocyte (M)-MDSC migration to tumor sites. Chidamide, a selective inhibitor of the histone deacetylase subtype, counteracted the gefitinib-induced increase in CCL2 levels in tumor cells. Additionally, chidamide down-regulated the expression of CCR2 in M-MDSCs, inhibiting their migration. Furthermore, chidamide attenuated the immunosuppressive function of M-MDSCs both alone and in combination with gefitinib. Chidamide also alleviated tumor immunosuppression by reducing the number of M-MDSCs in LLC-bearing mice, thereby enhancing the antitumor efficacy of gefitinib. In conclusion, our findings suggest that chidamide can improve gefitinib treatment outcomes, indicating that MDSCs are promising targets in NSCLC.


Subject(s)
Aminopyridines , Benzamides , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Myeloid-Derived Suppressor Cells , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/pathology , Gefitinib/pharmacology , Gefitinib/therapeutic use , Myeloid-Derived Suppressor Cells/metabolism , Lung Neoplasms/pathology , Cell Line, Tumor , Immunosuppressive Agents/therapeutic use , Treatment Outcome , Drug Resistance, Neoplasm
4.
Spectrochim Acta A Mol Biomol Spectrosc ; 317: 124434, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38735113

ABSTRACT

Probing intracellular organelles with fluorescent dyes offers opportunities to understand the structures and functions of these cellular compartments, which is attracting increasing interests. Normally, the design principle varies for different organelle targets as they possess distinct structural and functional profiles against each other. Therefore, developing a probe with dual intracellular targets is of great challenge. In this work, a new sort of donor-π-bridge-acceptor (D-π-A) type coumaranone dyes (CMO-1/2/3/4) have been prepared. Four fluorescent probes (TPP@CMO-1/2/3/4) were then synthesized by linking these coumaranone dyes with an amphiphilic cation triphenylphosphonium (TPP). Interestingly, both TPP@CMO-1 and TPP@CMO-2 exhibited dual color emission upon targeting to two different organelles, respectively. The green emission is well localized in mitochondria, while, the red emission realizes nucleoli imaging. RNA is the target of TPP@CMOs, which was confirmed by spectroscopic analysis and computational calculation. More importantly, the number and morphology changes of nucleoli under drug stress have been successfully evaluated using TPP@CMO-1.


Subject(s)
Cell Nucleolus , Fluorescent Dyes , Mitochondria , Organophosphorus Compounds , Organophosphorus Compounds/chemistry , Mitochondria/metabolism , Mitochondria/drug effects , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Humans , Cell Nucleolus/metabolism , HeLa Cells , Spectrometry, Fluorescence , Color
5.
J Cancer Res Clin Oncol ; 149(12): 10781-10796, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37314513

ABSTRACT

BACKGROUND: Ultra-Violet Radiation (UVR) is the most significant exogenous contributor to skin aging. UVB causes the senescence of melanocytes, which results in a permanent arrest in the proliferative process. Senescence is also regarded as a physiological tumor-suppressing mechanism of normal cells. However, the mechanism of the relationship between melanocyte senescence and melanoma was not sufficiently clarified. METHODS: Melanocytes and melanoma cells were irradiated with UVB for the indicated time. The miRNA expression profile of melanocytes were obtained by miRNA sequencing and confirmed by real-time PCR. Cell count kit-8 assays, cell cycle assays were also employed to explore the effect of miR-656-3p and LMNB2 on senescence. Dual-luciferase reporter assays were applied to determine the miRNA targets. Finally, a xenograft model and a photoaging model of mice were conducted to verified the function of miR-656-3p in vivo. RESULTS: Melanoma cells did not alter into a senescence stage and the expressions of miR-656-3p had no significant changes under the same intensity of UVB radiation. miR-656-3p appeared to be upregulated in melanocytes rather than melanoma cells after UVB radiation. miR-656-3p could promote the photoaging of human primary melanocytes by targeting LMNB2. Finally, overexpression of miR-656-3p significantly induced senescence and inhibited the growth of melanomas in vitro and in vivo. CONCLUSION: Our work not only demonstrated the mechanism by which miR-656-3p induced the senescence of melanocytes but also proposed a treatment strategy for melanomas by using miR-656-3p to induce senescence.

6.
NPJ Vaccines ; 8(1): 74, 2023 May 24.
Article in English | MEDLINE | ID: mdl-37225729

ABSTRACT

ZF2001, a protein subunit vaccine against coronavirus disease 2019 (COVID-19), contains recombinant tandem repeat of dimeric receptor-binding domain (RBD) protein of the SARS-CoV-2 spike protein with an aluminium-based adjuvant. During the development of this vaccine, two nonclinical studies were conducted to evaluate female fertility, embryo-fetal development, and postnatal developmental toxicity in Sprague‒Dawley rats according to the ICH S5 (R3) guideline. In Study 1 (embryo-fetal developmental toxicity, EFD), 144 virgin female rats were randomly assigned into four groups and received three doses of vaccine (25 µg or 50 µg RBD protein/dose, containing the aluminium-based adjuvant), the aluminium-based adjuvant or a sodium chloride injection administered intramuscularly on days 21 and 7 prior to mating and on gestation day (GD) 6. In Study 2 (pre- and postnatal developmental toxicity, PPND), ZF2001 at a dose of 25 µg RBD protein/dose or sodium chloride injection was administered intramuscularly to female rats (n = 28 per group) 7 days prior to mating and on GD 6, GD 20 and postnatal day (PND) 10. There were no obvious adverse effects in dams, except for local injection site reactions related to the aluminium-based adjuvant (yellow nodular deposits in the interstitial muscle fibres). There were also no effects of ZF2001 on the mating performance, fertility or reproductive performance of parental females, embryo-fetal development, postnatal survival, growth, physical development, reflex ontogeny, behavioural and neurofunctional development, or reproductive performance of the offspring. The strong immune responses associated with binding and neutralising antibodies were both confirmed in dams and fetuses or offspring in these two studies. These results would support clinical trials or the use of ZF2001 in maternal immunisation campaigns, including those involving women with childbearing potential, regardless of pregnancy status.

7.
Expert Opin Drug Metab Toxicol ; 19(11): 857-866, 2023.
Article in English | MEDLINE | ID: mdl-37921457

ABSTRACT

BACKGROUND: Stem cell-based therapies have demonstrated great potential in several clinical trials. However, safety data on stem cell application remain inadequate. This study evaluated the toxicity of human umbilical cord mesenchymal stem cells (hUC-MSCs) in NOD/Shi-scid/IL-2 Rγnull (NOG) mice. RESEARCH DESIGN AND METHODS: Mice were administered hUC-MSCs intravenously at doses of 3.5 × 106 cells/kg and 3.5 × 107 cells/kg. Toxicity was assessed by clinical observation, behavioral evaluation, pathology, organ weight, and histopathology. We determined the distribution of hUC-MSCs using a validated qPCR method and colonization using immunohistochemistry. RESULTS: No significant abnormal effects on clinical responses, body weight, or food intake were observed in the mice, except for two in the high-dose group that died during the last administration. Mouse activity in the high-dose group decreased 6 h after the first administration. Terminal examination revealed dose-dependent changes in hematology. The mice in the high-dose group displayed pulmonary artery wall plaques and mild alveolar wall microthrombi. hUC-MSCs colonized primarily the lung tissues and were largely distributed there 24 h after the final administration. CONCLUSIONS: The no observed adverse effect level for intravenous administration of hUC-MSCs in NOG mice over a period of 3 w was 3.5 × 106 cells/kg.


Subject(s)
Mesenchymal Stem Cells , Umbilical Cord , Humans , Mice , Animals , Injections, Intravenous , Mice, Inbred NOD , Lung , Mesenchymal Stem Cells/physiology
8.
Front Cell Dev Biol ; 11: 1273723, 2023.
Article in English | MEDLINE | ID: mdl-38020919

ABSTRACT

Human umbilical cord mesenchymal stem cells (hUC-MSCs) are proposed for the treatment of acute lung injury and atopic dermatitis. To advance hUC-MSC entry into clinical trials, the effects of hUC-MSCs on the general toxicity, immune perturbation and toxicokinetic study of hUC-MSCs in cynomolgus monkeys were assessed. hUC-MSCs were administered to cynomolgus monkeys by intravenous infusion of 3.0 × 106 or 3.0 × 107cells/kg or by subcutaneous injection of 3.0 × 107cells/kg twice a week for 3 weeks followed by withdrawal and observation for 6 weeks. Toxicity was assessed by clinical observation, clinical pathology, ophthalmology, immunotoxicology and histopathology. Moreover, toxicokinetic study was performed using a validated qPCR method after the first and last dose. After 3rd or 4th dosing, one or three the monkeys in the intravenous high-dose group exhibited transient coma, which was eliminated by slow-speed infusion after 5th or 6th dosing. In all dose groups, hUC-MSCs significantly increased NEUT levels and decreased LYMPH and CD3+ levels, which are related to the immunosuppressive effect of hUC-MSCs. Subcutaneous nodules and granulomatous foci were found at the site of administration in all monkeys in the subcutaneous injection group. Other than above abnormalities, no obvious systemic toxicity was observed in any group. The hUC-MSCs was detectable in blood only within 1 h after intravenous and subcutaneous administration. The present study declared the preliminary safety of hUC-MSCs, but close monitoring of hUC-MSCs for adverse effects, such as coma induced by intravenous infusion, is warranted in future clinical trials.

9.
J Zhejiang Univ Sci B ; 23(8): 699-704, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35953762

ABSTRACT

Keloids are a common type of pathological scar as a result of skin healing, which are extremely difficult to prevent and treat without recurrence. The pathological mechanism of keloids is the excessive proliferation of fibroblasts, which synthesize more extracellular matrices (ECMs), including type I/III collagen (COL-1/3), mucopolysaccharides, connective tissue growth factor (CTGF, also known as cellular communication network factor 2 (CCN2)), and fibronectin (FN) in scar tissue, mostly through the abnormal activation of transforming growth factor-|ß (TGF-|ß)/Smads pathway (Finnson et al., 2013; Song et al., 2018). Genetic factors, including race and skin tone, are considered to contribute to keloid formation. The reported incidence of keloids in black people is as high as 16%, whereas white people are less affected. The prevalence ratio of colored people to white people is 5:1||-||15:1 (Rockwell et al., 1989; LaRanger et al., 2019). In addition, keloids have not been reported in albinism patients of any race, and those with darker skin in the same race are more likely to develop this disease (LaRanger et al., 2019). Skin melanocyte activity is significantly different among people with different skin tones. The more active the melanocyte function, the more melanin is produced and the darker the skin. Similarly, in the same individual, the incidence of keloids increases during periods when melanocytes are active, such as adolescence and pregnancy. Keloids rarely appear in areas where melanocytes synthesize less melanin, such as in the palms and soles. Thus, the formation of keloids seems to be closely related to melanocyte activity.


Subject(s)
Exosomes , Keloid , Adolescent , Cells, Cultured , Exosomes/metabolism , Fibroblasts/metabolism , Humans , Keloid/pathology , Melanins/metabolism , Melanocytes/metabolism , Melanocytes/pathology , Pilot Projects , Skin/metabolism , Transforming Growth Factor beta/metabolism
10.
J Ethnopharmacol ; 283: 114670, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34653522

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Fritillariae thunbergii Miq (FTM)exhibit versatile biological activities including the significant antitussive and expectorant activities. As a herbal medicine, the therapeutic effects of FTM may be expressed by multi-components which have complicated integration effects on multi-targets. With the time going, the different processing methods of FTM has been changed a lot. Thus,the study described the effect of processing methods to FTM and its quality. MATERIAL AND METHOD: Studies were undertaken by using UHPLC-LTQ Orbitrap MS and pharmacodynamic models. All reagents were involved of analytical grade. While a HPLC-ELSD's method has been developed and validated, a certified Quality System is conformed to ICH requirements. The experimental animals followed the animal welfare guidelines. AIM OF THE STUDY: We aimed to found the differences after the different processing methods of FTM, and to demonstrate the changes could be selected as quality control indicators, and established a method for simultaneous determination of these for quality control. RESULTS: we have previously found two new steroidal alkaloids: zhebeininoside and imperialine-3-ß-D-glucoside from the different processing methods of FTM, which is the difference between the different processing methods of FTM, mainly on the steroidal alkaloids. The activity analysis of zhebeininoside, imperialine-3-ß-D-glucoside, verticine and verticinone showed that the mouse model of cough expectorant has antitussive effect. The positive drug selected was dextromethorphan syrup. The positive group showed biological activity, but the blank group showed nothing. The model group showed illness which means that the model was effective. There are two ways of the mechanism of action of the expectorant action which can make sputum thin, reduce its viscosity, and be easy to cough up, or can accelerate the movement of mucous cilia in the respiratory tract and promote the discharge of sputum. In our study, the content of phenol red was significantly reduced in the administration group. CONCLUSIONS: To sum up, our results suggest that zhebeininoside and other three components cloud be selected as quality control indicators, and a method for simultaneous determination of zhebeininoside and other three components was established for quality control.


Subject(s)
Antitussive Agents , Cevanes , Cough , Drugs, Chinese Herbal , Fritillaria , Animals , Mice , Ammonia/toxicity , Antitussive Agents/chemistry , Antitussive Agents/standards , Antitussive Agents/therapeutic use , Cevanes/chemistry , Cough/chemically induced , Cough/drug therapy , Dextromethorphan/therapeutic use , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/standards , Drugs, Chinese Herbal/therapeutic use , Fritillaria/chemistry , Phytotherapy , Plant Stems/chemistry , Quality Control , Random Allocation
11.
Autophagy ; 17(10): 3221-3237, 2021 10.
Article in English | MEDLINE | ID: mdl-33315519

ABSTRACT

Liver dysfunction is an outstanding dose-limiting toxicity of gefitinib, an EGFR (epidermal growth factor receptor)-tyrosine kinase inhibitor (TKI), in the treatment of EGFR mutation-positive non-small cell lung cancer (NSCLC). We aimed to elucidate the mechanisms underlying gefitinib-induced hepatotoxicity, and provide potentially effective intervention strategy. We discovered that gefitinib could sequentially activate macroautophagy/autophagy and apoptosis in hepatocytes. The inhibition of autophagy alleviated gefitinib-induced apoptosis, whereas the suppression of apoptosis failed to lessen gefitinib-induced autophagy. Moreover, liver-specific Atg7+/- heterozygous mice showed less severe liver injury than vehicle, suggesting that autophagy is involved in the gefitinib-promoted hepatotoxicity. Mechanistically, gefitinib selectively degrades the important anti-apoptosis factor COX6A1 (cytochrome c oxidase subunit 6A1) in the autophagy-lysosome pathway. The gefitinib-induced COX6A1 reduction impairs mitochondrial respiratory chain complex IV (RCC IV) function, which in turn activates apoptosis, hence causing liver injury. Notably, this autophagy-promoted apoptosis is dependent on PLK1 (polo like kinase 1). Both AAV8-mediated Plk1 knockdown and PLK1 inhibitor BI-2536 could mitigate the gefitinib-induced hepatotoxicity in vivo by abrogating the autophagic degradation of the COX6A1 protein. In addition, PLK1 inhibition could not compromise the anti-cancer activity of gefitinib. In conclusion, our findings reveal the gefitinib-hepatotoxicity pathway, wherein autophagy promotes apoptosis through COX6A1 degradation, and highlight pharmacological inhibition of PLK1 as an attractive therapeutic approach toward improving the safety of gefitinib-based cancer therapy.Abbreviations: 3-MA: 3-methyladenine; AAV8: adeno-associated virus serotype 8; ATG5: autophagy related 5; ATG7: autophagy related 7; B2M: beta-2-microglobulin; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CHX: cycloheximide; COX6A1: cytochrome c oxidase subunit 6A1; c-PARP: cleaved poly(ADP-ribose) polymerase; CQ: chloroquine; GOT1/AST: glutamic-oxaloacetic transaminase 1, soluble; GPT/ALT: glutamic pyruvic transaminase, soluble; HBSS: Hanks´ balanced salt solution; H&E: hematoxylin and eosin; MAP1LC3/LC3: microtubule associated proteins 1 light chain 3; PLK1: polo like kinase 1; RCC IV: respiratory chain complex IV; ROS: reactive oxygen species; TUBB8: tubulin beta 8 class VIII.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Chemical and Drug Induced Liver Injury , Lung Neoplasms , Animals , Apoptosis , Autophagy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle Proteins , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/pharmacology , Gefitinib/pharmacology , Lung Neoplasms/metabolism , Mice , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Polo-Like Kinase 1
12.
PLoS One ; 15(8): e0237023, 2020.
Article in English | MEDLINE | ID: mdl-32785244

ABSTRACT

OBJECTIVE: Melanocytes play a central role in skin homeostasis. In this study, we focus on the function of melanocyte releasing exosomes as well as exosomal microRNAs (miRNAs) and investigate whether ultraviolet B (UVB) irradiation exerts an impact on it. MATERIALS AND METHODS: Exosomes derived from human primary melanocytes were isolated through differential centrifugation and were identified in three ways, including transmission electron microscopy observation, nanoparticle tracking analysis, and Western blot analysis. Melanocytes were irradiated with UVB for the indicated time, and then melanin production and exosome secretion were measured. The exosomal miRNA expression profile of melanocytes were obtained by miRNA sequencing and confirmed by real-time PCR. RESULTS: Exosomes derived from human primary melanocytes were verified. UVB irradiation induced melanin production and increased the exosome release by the melanocytes. In total, 15 miRNAs showed higher levels in UVB-irradiated melanocyte-derived exosomes compared with non-irradiated ones, and the top three upregulated exosomal miRNAs were miR-4488, miR-320d, and miR-7704 (fold change > 4.0). CONCLUSION: It is verified for the first time that UVB irradiation enhanced the secretion of exosomes by melanocytes and changed their exosomal miRNA profile. This findings open a new direction for investigating the communication between melanocytes and other skin cells, and the connection between UVB and skin malignant initiation.


Subject(s)
Exosomes/genetics , Melanocytes/metabolism , Melanocytes/radiation effects , Bodily Secretions/metabolism , Exosomes/metabolism , Humans , Melanins/analysis , MicroRNAs/genetics , Primary Cell Culture , Transcriptome/genetics , Ultraviolet Rays/adverse effects
14.
Glob Med Genet ; 9(4): 263-264, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36530527
15.
Environ Toxicol Pharmacol ; 49: 81-88, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27951409

ABSTRACT

Capecitabine, as the first-line treatment for multiple tumor types, has a serious drawback of hand-foot-syndrome (HFS) that limits its clinical use. However, the pathophysiology and mechanism of capecitabine-induced HFS is rarely known. Here we built the experimental mouse model of HFS induced by capecitabine at first and it was shown that 3 of 6 mice appeared HFS in the 5th day and 5 mice occurred HFS in the 30th day. The corneous layer was reduced in capecitabine-induced HFS in vivo. Moreover, we found that capecitabine could significantly induce keratinocytes cells death in vitro through activated apoptosis pathway and decreased mitochondrial membrane potential. In conclusion, these results suggested that HFS of capecitabine may be developed from reduction of corneous layer through stimulation of intracellular mitochondrial dysfunction following activation of caspase-dependent apoptosis pathway.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Capecitabine/adverse effects , Hand-Foot Syndrome/pathology , Keratinocytes/drug effects , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Hand-Foot Syndrome/physiopathology , Humans , Keratinocytes/pathology , Keratinocytes/physiology , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred ICR
16.
PLoS One ; 11(1): e0146968, 2016.
Article in English | MEDLINE | ID: mdl-26752698

ABSTRACT

Chemotherapy is the only choice for most of the advanced hepatocellular carcinoma (HCC) patients, while few agents were available, making it an urgent need to develop new chemotherapy strategies. A phase II clinical trial suggested that the efficacy of irinotecan in HCC was limited due to dose-dependent toxicities. Here, we found that gefitinib exhibited synergistic activity in combination with SN-38, an active metabolite of irinotecan, in HCC cell lines. And the enhanced apoptosis induced by gefitinib plus SN-38 was a result from caspase pathway activation. Mechanistically, gefitinib dramatically promoted the ubiquitin-proteasome-dependent degradation of Rad51 protein, suppressed the DNA repair, gave rise to more DNA damages, and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of gefitinib combined with irinotecan was further validated in a HepG2 xenograft mice model. Taken together, our data demonstrated for the first time that the combination of irinotecan and gefitinib showed potential benefit in HCC, which suggests that Rad51 is a promising target and provides a rationale for clinical trials investigating the efficacy of the combination of topoisomerase I inhibitors and gefitinib in HCC.


Subject(s)
Camptothecin/analogs & derivatives , Carcinoma, Hepatocellular/drug therapy , DNA Repair , Liver Neoplasms/drug therapy , Quinazolines/administration & dosage , Rad51 Recombinase/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis , Blotting, Western , Camptothecin/administration & dosage , Camptothecin/chemistry , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Drug Synergism , Gefitinib , Hep G2 Cells , Humans , Irinotecan , Mice , Mice, Nude , Neoplasm Transplantation , Quinazolines/chemistry , RNA, Small Interfering/metabolism , Topoisomerase I Inhibitors/administration & dosage , Topoisomerase I Inhibitors/chemistry
17.
Mol Med Rep ; 13(1): 21-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26531807

ABSTRACT

The aim of the present study was to investigate the protective effects of melatonin (MLT) on hypertension-induced renal injury and identify its mechanism of action. Twenty-four healthy male Wistar rats were divided into a sham control group (n=8), which was subjected to sham operation and received vehicle treatment (physiological saline intraperitoneally at 0.1 ml/100 g), a vehicle group (n=8), which was subjected to occlusion of the left renal artery and vehicle treatment, and the MLT group (n=8), which was subjected to occlusion of the left renal artery and treated with MLT (10 mg/kg/day). Pathological features of the renal tissues were determined using hematoxylin and eosin staining and Masson staining. Urine protein, serum creatinine (Scr), superoxide dismutase (SOD) and malondialdehyde (MDA) were determined. Immunohistochemical analysis was performed to determine the expression of heme oxygenase­1 (HO­1), intercellular adhesion molecule­1 (ICAM­1), inducible nitric oxide synthase (iNOS) and endothelial nitric oxide synthase (eNOS). Furthermore, reverse transcription polymerase chain reaction was conducted to determine the mRNA expression of HO­1, ICAM­1, eNOS and iNOS. A marked decrease in blood pressure was noticed in the MLT group at week 4 compared with that of the vehicle group (P<0.01). Furthermore, MLT treatment attenuated the infiltration of inflammatory cells and oedema/atrophy of renal tubules. MLT attenuated hypertension-induced increases in urine protein excretion, serum creatinine and MDA as well as decreases in SOD activity in renal tissues. Furthermore, MLT attenuated hypertension-induced increases in iNOS and ICAM­1 as well as decreases in eNOS and HO­1 expression at the mRNA and protein level. In conclusion, the results of the present study indicated that MLT had protective roles in hypertension­induced renal injury. Its mechanism of action is, at least in part, associated with the inhibition of oxidative stress.


Subject(s)
Hypertension, Renal/complications , Kidney Diseases/etiology , Melatonin/pharmacology , Oxidative Stress/drug effects , Animals , Blood Pressure/drug effects , Creatinine/blood , Edema/complications , Edema/pathology , Edema/physiopathology , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hypertension, Renal/blood , Hypertension, Renal/physiopathology , Immunohistochemistry , Inflammation/blood , Inflammation/complications , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/blood , Kidney Diseases/physiopathology , Male , Malondialdehyde/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Proteinuria/blood , Proteinuria/complications , Proteinuria/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Superoxide Dismutase/metabolism
18.
Toxicol Rep ; 3: 78-86, 2016.
Article in English | MEDLINE | ID: mdl-28959525

ABSTRACT

Visual impairment is a global public health problem that needs new candidate drugs. Chrysanthemum is a traditional Chinese drug, famous for its eye-protective function, with an unclear mechanism of action. To determine how chrysanthemum contributes to vision, we identified, for the first time, the component of chrysanthemum, diosmetin (DIO), which acts in protecting the injured retina in an adriamycin (ADR) improving model. We observed that DIO could attenuate the apoptosis of retinal cells in Sprague-Dawley rats and verified this effect in cultured human retinal pigment epithelium (RPE) cells, ARPE-19. Our further study on the mechanism revealed the counteractive effect of DIO on the attenuation of DNA damage and oxidative stress, which occurs in a wide range of retinal disorders. These results collectively promise the potential value of DIO as a retinal-protective agent for disorders that lead to blindness. In addition, we identified, for the first time, the component of chrysanthemum, DIO, which acts in protecting the injured retina.

19.
Oncotarget ; 6(5): 3254-67, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25226612

ABSTRACT

Adriamycin, a widely used anthracycline antibiotic in multiple chemotherapy regimens, has been challenged by the cardiotoxicity leading to fatal congestive heart failure in the worst condition. The present study demonstrated that Dihydromyricetin, a natural product extracted from ampelopsis grossedentat, exerted cardioprotective effect against the injury in Adriamycin-administrated ICR mice. Dihydromyricetin decreased ALT, LDH and CKMB levels in mice serum, causing a significant reduction in the toxic death triggered by Adriamycin. The protective effects were also indicated by the alleviation of abnormal electrocardiographic changes, the abrogation of proliferation arrest and apoptotic cell death in primary myocardial cells. Further study revealed that Dihydromyricetin-rescued loss of anti-apoptosis protein ARC provoked by Adriamycin was involved in the cardioprotection. Intriguingly, the anticancer activity of Adriamycin was not compromised upon the combination with Dihydromyricetin, as demonstrated by the enhanced anticancer effect achieved by Adriamycin plus Dihydromyricetin in human leukemia U937 cells and xenograft models, in a p53-dependent manner. These results collectively promised the potential value of Dihydromyricetin as a rational cardioprotective agent of Adriamycin, by protecting myocardial cells from apoptosis, while potentiating anticancer activities of Adriamycin, thus further increasing the therapeutic window of the latter one.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Flavonols/pharmacology , Heart Diseases/prevention & control , Lymphoma, Large B-Cell, Diffuse/drug therapy , Protective Agents/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Proliferation/drug effects , Cytoprotection , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , HL-60 Cells , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , K562 Cells , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice, Inbred BALB C , Mice, Inbred ICR , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Tumor Burden , Tumor Suppressor Protein p53/metabolism , U937 Cells , Xenograft Model Antitumor Assays
20.
Oncotarget ; 6(8): 6203-17, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25749037

ABSTRACT

Liver dysfunction is a common side effect associated with the treatment of dasatinib and its mechanism is poorly understood. Autophagy has been thought to be a potent survival or death factor for liver dysfunction, which may shed the light on a novel strategy for the intervention of hepatotoxicity caused by dasatinib. In this study, we show for the first time that autophagy is induced, which is consistent with the formation of liver damage. Autophagy inhibition exacerbated dasatinib-induced liver failure, suggesting that autophagy acted as a self-defense mechanism to promote survival. Oxidative stress has been shown to be an important stimulus for autophagy and hepatotoxicity. Interestingly, dasatinib increased the activity of p38, which is a critical modulator of the oxidative stress related to liver injury and autophagy. p38 silencing significantly blocked LC3-II induction and p62 reduction by dasatinib, which was accompanied by increased caspase-3 and PARP cleavage, indicating that autophagy alleviated dasatinib-induced hepatotoxicity via p38 signaling. Finally, the p38 agonist isoproterenol hydrochloride (ISO) alleviated dasatinib-induced liver failure by enhancing autophagy without affecting the anticancer activity of dasatinib. Thus, this study revealed that p38-activated autophagy promoted survival during liver injury, which may provide novel approaches for managing the clinical applications of dasatinib.


Subject(s)
Autophagy/physiology , Chemical and Drug Induced Liver Injury/pathology , Dasatinib/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Dasatinib/adverse effects , Drug Interactions , Female , Humans , Isoproterenol/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL