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










Publication year range
1.
Heliyon ; 10(8): e29005, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38628708

ABSTRACT

The main challenge in treating stomach adenocarcinoma (STAD) is chemotherapy resistance, which is characterized by changes in the immune microenvironment. Disulfidptosis, a novel form of programmed cell death, is involved in STAD but its mechanism is not fully understood. Long non-coding RNAs (LncRNAs) may play a role in regulating disulfidptosis and influencing the immune microenvironment and chemotherapy resistance in STAD. This study aims to establish disulfidptosis-related lncRNA (DRL) features and explore their significance in the immune microenvironment and chemotherapy resistance in STAD patients. By analyzing RNA sequencing and clinical data from STAD patients and extracting disulfidptosis-related genes, we identified DRLs through co-expression, single-factor and multi-factor Cox regression, and Lasso regression analyses. We also investigated differences in the immune microenvironment, immune function, immune checkpoint gene expression, and chemotherapy resistance between different risk groups using various algorithms. A prognostic risk model consisting of 2 DRLs was constructed, with a strong predictive value for patient survival, outperforming other clinical-pathological factors in predicting 3-year and 5-year survival. Immune-related analysis revealed a strong positive correlation between T cell CD4+ cells and risk score across all algorithms, and higher expression of immune checkpoint genes in the high-risk group. In addition, high-risk patients showed better sensitivity to Erlotinib, Oxaliplatin, and Gefitinib. Furthermore, three novel molecular subtypes of STAD were identified based on the 2-DRLs features, with evaluation of the immune microenvironment and chemotherapy drug sensitivity for each subgroup, which holds significant implications for achieving precise treatment in STAD. Overall, our 2-DRLs prognostic model demonstrates high predictive value for patient survival in STAD, potentially providing new targets for individualized immune and chemical therapy.

2.
World J Gastroenterol ; 30(8): 919-942, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38516243

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common types of tumors. The influence of lipid metabolism disruption on the development of HCC has been demonstrated in published studies. AIM: To establish an HCC prognostic model for lipid metabolism-related long non-coding RNAs (LMR-lncRNAs) and conduct in-depth research on the specific role of novel LMR-lncRNAs in HCC. METHODS: Correlation and differential expression analyses of The Cancer Genome Atlas data were used to identify differentially expressed LMR-lncRNAs. Quantitative real-time polymerase chain reaction analysis was used to evaluate the expression of LMR-lncRNAs. Nile red staining was employed to observe intracellular lipid levels. The interaction between RP11-817I4.1, miR-3120-3p, and ATP citrate lyase (ACLY) was validated through the performance of dual-luciferase reporter gene and RIP assays. RESULTS: Three LMR-lncRNAs (negative regulator of antiviral response, RNA transmembrane and coiled-coil domain family 1 antisense RNA 1, and RP11-817I4.1) were identified as predictive markers for HCC patients and were utilized in the construction of risk models. Additionally, proliferation, migration, and invasion were reduced by RP11-817I4.1 knockdown. An increase in lipid levels in HCC cells was significantly induced by RP11-817I4.1 through the miR-3120-3p/ACLY axis. CONCLUSION: LMR-lncRNAs have the capacity to predict the clinical characteristics and prognoses of HCC patients, and the discovery of a novel LMR-lncRNAs, RP11-817I4.1, revealed its role in promoting lipid accumulation, thereby accelerating the onset and progression of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lipid Metabolism/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Fatty Acids , Lipids , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Cell Line, Tumor
3.
J Pharm Pharmacol ; 76(3): 224-235, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38244223

ABSTRACT

OBJECTIVES: Colorectal cancer (CRC) is the fourth most commonly diagnosed cancer worldwide. Solanine is a phytochemical extracted from traditional Chinese medicine with widely reported anticancer effects. Here, we investigated the potential role of solanine in regulating ferroptosis in CRC cells and scrutinized the molecular mechanism. METHODS: Cell growth and cytotoxicity were examined using CCK-8 proliferation assay and lactate dehydrogenase assay. Oxidative stress was determined by measuring glutathione (GSH), malondialdehyde, and reactive oxygen species (ROS) levels. Subcellular changes in mitochondria were examined by transmission electron microscopy. Gene and protein expression levels were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Protein-protein interaction was determined by co-immunoprecipitation. KEY FINDINGS: Solanine arrested cell proliferation in CRC cells and induced typical ferroptotic changes. Solanine treatment promoted ROS production, lipid peroxidation, and cell membrane disruption, while the cellular level of antioxidant GSH was reduced upon solanine treatment. ALOX12B was identified as a molecular mediator of solanine to promote ferroptosis. Solanine treatment upregulated ALOX12B levels and silencing ALOX12B could suppress solanine-induced ferroptosis. Further, ADCY4 was found to physically associate with ALOX12B and maintain ALOX12B protein stability. Silencing ADCY4 destabilized ALOX12B and attenuated solanine-induced ferroptosis. CONCLUSIONS: Our data demonstrated the ferroptosis-inducing effect of solanine in CRC cells, and revealed ALOX12B/ADCY4 molecular axis as the ferroptosis mediator of solanine. Solanine may synergize with existing ferroptosis inducer as an anticancer strategy in CRC, which warrants further validation in animal experiments.


Subject(s)
Colorectal Neoplasms , Ferroptosis , Solanine , Animals , Reactive Oxygen Species , Cell Membrane , Glutathione , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics
6.
Front Mol Biosci ; 10: 1298077, 2023.
Article in English | MEDLINE | ID: mdl-38106991

ABSTRACT

Introduction: The involvement of endoplasmic reticulum (ER) stress in cancer biology is increasingly recognized, yet its role in pancreatic cancer (PC) remains unclear. This study aims to elucidate the impact of ER stress on prognosis and biological characteristics in PC patients. Methods: A bioinformatic analysis was conducted using RNA-seq data and clinicopathological information from PC patients in the TCGA and ICGC databases. The ER stress-associated gene sets were extracted from MSigDB. ER stress-associated genes closely linked with overall survival (OS) of PC patients were identified via log-rank test and univariate Cox analysis, and further narrowed by LASSO method. A risk signature associated with ER stress was formulated using multivariate Cox regression and assessed through Kaplan-Meier curves, receiver operating characteristic (ROC) analyses, and Harrell's concordance index. External validation was performed with the ICGC cohort. The single-sample gene-set enrichment analysis (ssGSEA) algorithm appraised the immune cell infiltration landscape. Results: Worse OS in PC patients with high-risk signature score was observed. Multivariate analysis underscored our ER stress-associated signature as a valuable and independent predictor of prognosis. Importantly, these results based on TCGA were further validated in ICGC dataset. In addition, our risk signature was closely associated with homeostasis, protein secretion, and immune regulation in PC patients. In particular, PC microenvironment in the high-risk cluster exhibited a more immunosuppressive status. At last, we established a nomogram model by incorporating the risk signature and clinicopathological parameters, which behaves better in predicting prognosis of PC patients. Discussion: This comprehensive molecular analysis presents a new predictive model for the prognosis of PC patients, highlighting ER stress as a potential therapeutic target. Besides, the findings indicate that ER stress can have effect modulating PC immune responses.

8.
Obes Surg ; 33(3): 897-910, 2023 03.
Article in English | MEDLINE | ID: mdl-36701012

ABSTRACT

PURPOSE: This study reviews the prevalence of iron deficiency (ID) in bariatric surgery candidates and the long-term outcomes of the prevalence of ID after bariatric surgery. MATERIALS AND METHODS: A systematic literature search and meta-analysis were performed in PubMed for articles published by August 31, 2022, including these search terms: bariatric surgery, metabolic surgery, weight loss surgery, obesity surgery, sleeve gastrectomy, gastric banding, gastric bypass, duodenal switch, duodenojejunal bypass, iron, iron deficiency, sideropenia, and hypoferritinemia. Fifty-seven studies examining a total of 26,328 patients with morbidly obese were included in this meta-analysis finally. RESULTS: The results showed a prevalence of 17% of ID in bariatric surgery candidates and a prevalence of 14%, 17%, 26%, 34%, 23%, 38%, and 23% of ID at 1-, 2-, 3-, 4-, 5-, 8-, and 10-year follow-up after bariatric surgery, respectively. Additionally, the results showed a prevalence of 15%, 19%, 35%, 38%, 29%, 30%, and 23% of ID at 1-, 2-, 3-, 4-, 5-, 8-, and 10-year follow-up after Roux-en-Y gastric bypass, respectively; a prevalence of 12%, 12%, 15%, 31%, and 17% of ID at 1-, 2-, 3-, 4-, and 5-year follow-up after sleeve gastrectomy, respectively; and a prevalence of 19% of ID at 1-year follow-up after anastomosis gastric bypass. CONCLUSION: As a result, preoperative evaluation and correction of ID may lead to better outcomes for bariatric surgery candidates. ID is also common in patients after bariatric procedures, especially RYGB. Long-term, even lifelong, medical and nutritional monitoring and tailored interventions are critical.


Subject(s)
Bariatric Surgery , Gastric Bypass , Iron Deficiencies , Obesity, Morbid , Humans , Obesity, Morbid/surgery , Weight Loss , Bariatric Surgery/methods , Gastric Bypass/methods , Gastrectomy/methods , Retrospective Studies , Treatment Outcome
9.
Anticancer Drugs ; 32(2): 127-137, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33417325

ABSTRACT

Gastric cancer is one of the most common malignant tumors. MicroRNA-196b (miR-196b) has been demonstrated to play important roles in human cancers. However, its functions in gastric cancer progression were still largely unknown. In this study, the expression of miR-196b was determined by quantitative real-time PCR. Esophageal cancer-related gene 4 (ECRG4) level was examined by western blot assay and immunohistochemistry staining assay. Cell proliferation was evaluated by Cell Counting Kit-8 (CCK-8) assay and colony formation assay. Cell migration and invasion were analyzed by transwell assay. The association between miR-196b and ECRG4 was analyzed by dual-luciferase reporter assay. The functional role of miR-196b in vivo was analyzed by murine xenograft assay. As a result, we found the expression of miR-196b was elevated and the protein expression of ECRG4 was reduced in gastric cancer tissues and cells. MiR-196b inhibition suppressed gastric cancer cell proliferation, migration and invasion. ECRG4 was a target of miR-196b and its protein expression was negatively regulated by miR-196b. Moreover, ECRG4 overexpression showed similar effects with miR-196b inhibition on the malignant behaviors of GC cells and ECRG4 knockdown reversed the effects of miR-196b inhibition on gastric cancer cell proliferation, migration and invasion. In addition, miR-196b inhibition suppressed tumor volume and weight in vivo. In conclusion, downregulation of miR-196b inhibited gastric cancer progression by modulating ECRG4 expression, indicating that miR-196b might be a potential therapeutic target for gastric cancer.


Subject(s)
MicroRNAs/biosynthesis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/biosynthesis , Aged , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Real-Time Polymerase Chain Reaction , Up-Regulation
10.
Artif Cells Nanomed Biotechnol ; 47(1): 4120-4130, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31701767

ABSTRACT

Background: Gastric cancer (GC) is a global leading source of cancer-associated deaths. Circular RNAs (circRNAs) are a new type of non-coding RNA and promising biomarkers for diagnosis of multiple diseases such as cancer.Methods: Circ-PRMT5 expression was validated in 90 GC patient tissues and 6 different GC cells by qRT-PCR. Sublocalization of circ-PRMT5 in GC cells was determined in isolated nuclear and cytoplasmic RNAs. CircInteractome and miRanda were used to predict binding sites between circ-PRMT5 with micRNAs, and micRNAs with target mRNA. The correlation between genes was determined by the Pearson correlation analysis. The molecular mechanism was demonstrated by RNA in vivo precipitation, point mutation, luciferase activity and rescue experiments.Results: Circ-PRMT5 expression was significantly higher in GC than in adjacent normal tissues, and GC patients with circ-PRMT5 high expression had shorter survival times. Functionally, circ-PRMT5 silence inhibited GC cell growth and invasion. Mechanism analysis showed that circ-PRMT5 sponged miR-145/miR-1304 to upregulate MYC expression and GC development.Conclusion: Our findings demonstrated that circ-PRMT5 function as an oncogene in GC patients by targeting miR-145/miR-1304/MYC axis. High circ-PRMT5 expression may provide a poor prognostic indicator of survival in GC patients and targeting circ-PRMT5/miR-145/miR-1304/MYC axis may be a novel therapeutic strategy for GC.


Subject(s)
Disease Progression , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Circular/genetics , Stomach Neoplasms/pathology , Up-Regulation/genetics , Apoptosis/genetics , Base Sequence , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics
12.
Oncol Lett ; 10(6): 3505-3509, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26788158

ABSTRACT

The aim of this study was to investigate the effects of small interfering RNA (siRNA) targeting human growth hormone receptor (hGHR) combined with 5-fluorouracil (5-FU) on the hepatic metastasis of colon cancer. The animal model of liver metastases using human SW480 colon cancer cells was established on BALB/c mice and the siRNA interfering plasmid targeting hGHR gene was constructed. The tumor-bearing mice were randomly divided into the saline control, plasmid, growth hormone (GH), 5-FU, 5-FU+plasmid and 5-FU+plasmid+GH groups. The liver metastasis in each group was observed. All the animals showed liver metastases and using siRNA-interfering plasmid treatment the incidence of liver metastases was significantly reduced in the tumor groups compared to the saline or GH group. The combined treatment of interfering plasmid and 5-FU slightly decreased the incidence of liver metastases in the tumor groups compared to the plasmid alone or 5-FU alone treatment, although the findings were not statistically significant. On the basis of the combination of interfering plasmid and 5-FU, the additional GH did not increase the incidence of liver metastases (P>0.05), but improved the weight loss of the mice (P<0.05) induced by the inhibition of GHR and toxicity of 5-FU. The present results showed that siRNA targeting hGHR is able to reduce the incidence of liver metastases of human SW480 colon cancer cells in mice. Thus, GHR may be important in tumor metastasis.

13.
Article in Chinese | MEDLINE | ID: mdl-20695382

ABSTRACT

OBJECTIVE: To establish a new induction method from human embryonic stem cells (hESCs) differentiating into hepatocyte-like cells using an adherent culture system with single-step induction. METHODS: Undifferentiated hESCs were cultured on Matrigel-coated culture plates for 4 days, hepatic differentiation was initiated at 60%-70% confluence by adding Activin A for 5 days. Then the induction medium was replaced by hepatocyte induction medium (HIM) supplemented with fibroblast growth factor 1 (FGF-1) and bone morphogenetic protein 4 (BMP-4) for another 6 days. Finally, the cells were treated with HIM adding hepatocyte growth factor (HGF) and Oncostatin M (OSM) for 5-7 days. The characteristics of differentiated cells were determined by morphology, immunofluorescence staining, RT-PCR, and Periodic acid-Schiff (PAS) test. RESULTS: Differentiated cells treated with Activin A, FGF-1, BMP-4, HGF, and OSM sequentially were morphologically larger and became spherical, oval or polygon. Some cells had 2 or 3 nuclei, suggesting that the cells have a hepatocyte-like morphology. Differentiated cells at first induction stage could be stained positive by SOX17 and Forkhead (FOX)A2 after induction by Activin A. Then they turned to be a fetoprotein (AFP) and al antitrypsin positive cells at second induction stage after induction by FGF-1 and BMP-4. Finally, the differentiated cells treated with HGF and OSM showed PAS positive for glycogen detection. The differentiated cells at various stages also expressed at early (SOX17, FOXA2, and GATA-4), middle (AFP, albumin, and cytokeratin 18), and mature (alcohol dehydrogenase 1C and Cytochrome P4501B1) stage hepatic genes, respectively. CONCLUSION: Using a simple-step induction method and by supplied with cytokines consequently, hESCs can be induced to differentiate into hepatocyte-like cells. The differentiation method can provide seed cells for hepatic tissue engineering or cell-therapy.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Cell Culture Techniques , Cells, Cultured , Humans , Tissue Engineering/methods
14.
Eur J Pharmacol ; 645(1-3): 23-31, 2010 Oct 25.
Article in English | MEDLINE | ID: mdl-20655899

ABSTRACT

Promoting human embryonic stem cell (hESC)-derived-neural progenitor survival in the pro-apoptotic niche is pivotal for stem cell replacement therapy. The present study was designed to investigate the protective effect of hepatocyte growth factor (HGF) on hESC-derived neural progenitor injured by hydrogen peroxide (H(2)O(2)) exposure. Treatment of hESC-derived neural progenitor cells with HGF prior to H(2)O(2) exposure conferred protective effect against oxidative stress-induced apoptosis. HGF treatment increased both phosphoinositide 3-kinase (PI3K)/Akt and extracellular signal-regulated kinase1/2 (ERK1/2) phosphorylation. However, selective inhibition of each pathway supported that the activation of PI3K/AKT, but not ERK1/2, provides survival advantage to the neural progenitor cells. Further investigation indicated that HGF pretreatment could attenuate the decrease of the expression of Bcl-2 protein induced by H(2)O(2), whereas the level of Bax was not affected. Additionally, we observed that H(2)O(2)-induced decrease of mitochondrial transmembrane potential, release of cytochrome c and increase of caspase-3 activation were alleviated by HGF pretreatment. These effects of HGF could be reversed by inhibition of the PI3K/Akt and ERKs pathways, indicating PI3K/Akt and ERKs signaling might be involved in HGF-mediated regulation of mitochondrial apoptotic pathway mediated by H(2)O(2). The neuroprotective effect of HGF might potentially be useful in stem cell-based therapies for neurodegenerative disorders.


Subject(s)
Apoptosis/drug effects , Embryonic Stem Cells/drug effects , Hepatocyte Growth Factor/pharmacology , Hydrogen Peroxide/pharmacology , Neural Stem Cells/drug effects , Neuroprotective Agents/pharmacology , Caspase 3/metabolism , Cells, Cultured , Cytochromes c/metabolism , Embryonic Stem Cells/cytology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neural Stem Cells/cytology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism
15.
Chin Med J (Engl) ; 123(2): 142-5, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20137360

ABSTRACT

BACKGROUND: Abdominal pain is a common symptom among patients with acute appendicitis, yet these patients have long been denied relief from suffering because of widespread misconceptions associated with the use of opioids. We determined whether morphine hydrochloride masked the physical signs in adults with acute appendicitis and assessed the efficacy of morphine in relieving abdominal pain. METHODS: A prospective, double-blind, placebo controlled, clinical trial was conducted with 106 adult patients between 16 and 70 years old with acute appendicitis. Patients were randomly divided into a morphine group (n=54) or a normal saline group (n=52). All patients presented with acute abdominal pain with onset within 3 days. The morphine group received hypodermic injection of morphine (0.15 mg/kg; maximum 20 mg) and the control group members were given an equivalent volume of normal saline solution. The clinical symptoms, physical signs, and patients' cooperation during physical examination were assessed before and after 30 minutes of morphine or normal saline administration. RESULTS: Abdominal pain was significantly relieved and the patients' cooperation was improved in the morphine group after 30 minutes treatment compared with the control group and before morphine administration (P<0.05). The physical signs were unaffected by either treatment (P>0.05). CONCLUSIONS: Morphine relieved abdominal pain and improved the patients' cooperation for treatment and care. Furthermore, the morphine did not mask the physical signs of acute appendicitis.


Subject(s)
Abdominal Pain/drug therapy , Appendicitis/drug therapy , Morphine/therapeutic use , Abdominal Pain/pathology , Adolescent , Adult , Aged , Analgesics, Opioid/therapeutic use , Appendicitis/pathology , Female , Humans , Male , Middle Aged , Young Adult
17.
Sheng Li Xue Bao ; 61(3): 247-54, 2009 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-19536437

ABSTRACT

Hepatocyte growth factor (HGF) pretreatment could protect multiple cell types from apoptosis induced by various damages including oxidative stress. The present study was designed to investigate the protective effect of HGF on rat cortical neurons against apoptosis induced by hydrogen peroxide (H2O2) in culture, and then to explore whether HGF could influence the mitochondrial pathway of apoptosis. Primary rat cortical neurons were isolated from Sprague-Dawley rats and cultured in serum free medium containing 2% B27 and Neurobasal-A. To mimic the oxidative stress damage, cortical neurons were exposed to 100 mumol/L H2O2 for 4 h. To explore the effects of HGF on the neurons subjected to H2O2 injury, cells were pretreated with HGF 15, 30, 60 ng/mL for 24 h, respectively, and then exposed to 100 mumol/L H2O2 for 4 h. The cell viability was measured by MTT colorimetric assay and cell injury was evaluated by lactate dehydrogenase (LDH) leakage rate. Apoptotic cells were detected by Hoechst 33258 staining and Annexin V-FITC/PI double labeled flow cytometry. The caspase-3 activity was assessed by colorimetry. The alteration of transmembrane potential of mitochondria was determined by confocal laser scanning microscopy. The expression of cytochrome C protein was measured by Western blot analysis. The results showed that H2O2 treatment significantly decreased the cell viability, increased LDH leakage rate and the percentage of apoptotic cells. Pretreatment of HGF at different concentrations (15-60 ng/mL) could remarkably increase the cell viability of neurons. Compared with that of H2O2 group (53.4%+/-7.4%), the cell viabilities of neurons treated with 15, 30, and 60 ng/mL HGF significantly increased to (69.3+/-6.4)%, (77.5+/-6.1)% and (82.9+/-9.3)% (P<0.05), respectively. HGF preincubation also evidently decreased the LDH leakage rate in cortical neurons damaged by H2O2. The results of Hoechst staining revealed that HGF pretreatment could significantly reduce the apoptotic rate of neurons. The apoptotic rate of H2O2 group was (62.8+/-7.1)%, while that of HGF groups decreased significantly to (34.8+/-8.4)%, (23.5+/-3.2)% and (18.6+/-4.5)% (P<0.05), respectively. The data from caspase-3 activity assay indicated that HGF preconditioning could also remarkably decrease the caspase-3 activity of neurons. In addition, in the presence of various concentrations of HGF, the decrease of transmembrane potential of mitochondria in neurons caused by H2O2 injury could be reversed. Moreover, as detected by Western blot analysis, HGF downregulated the expression of cytochrome C protein in neurons. These results suggest that HGF has a protective effect on rat cortical neurons against apoptosis induced by H2O2, which might be related to the inhibition of the mitochondrial apoptotic pathway and the suppression of the caspase-3 activity.


Subject(s)
Apoptosis , Hepatocyte Growth Factor/pharmacology , Mitochondria/physiology , Neurons/drug effects , Animals , Brain/cytology , Caspase 3/metabolism , Cell Survival , Cells, Cultured , Cytochromes c/metabolism , Hydrogen Peroxide/pharmacology , Neurons/cytology , Oxidative Stress , Rats , Rats, Sprague-Dawley
18.
Chemotherapy ; 54(6): 447-55, 2008.
Article in English | MEDLINE | ID: mdl-18832817

ABSTRACT

Human growth hormone (hGH) is a pleiotropic cytokine targeting a variety of tissues. Its protective effects on haematopoiesis during treatment with chemotherapeutic drugs were investigated. Sprague-Dawley rats were intraperitoneally administered with both carboplatin and 5-fluorouracil together with or without recombinant hGH (rhGH) at a dose of 1 IU/kg/day. Body weight, full blood count, bone marrow differential count and expression of proliferating cell nuclear antigen (PCNA) in bone marrow were measured weekly for 4 weeks in chemotherapy-treated (CT) or chemotherapy plus rhGH-treated (CT+GH) animals. During the first week, body weight, white blood cell count, haematopoietic count and reticulocyte count were decreased in the CT group as well as in the CT+GH group, but to a lesser extent in the latter group (CT vs. CT+GH group, p < 0.05). Further decreases were also prevented in the CT+GH group. Myeloid cells were extremely hypoplastic in the CT group, while in the CT+GH group, myeloid cells recovered from obviously hypoplastic to excessively hyperplastic in the first 2 weeks, and the myeloid karyocyte count increased significantly (CT+GH vs. CT group, p < 0.05). PCNA-positive cell count in the CT+GH group was also significantly higher than in the CT group (CT+GH vs. CT group, p < 0.05). Thus, rhGH showed a promising protective effect on body weight loss and haematopoietic recovery of chemotherapy-treated rats, which may be useful in clinic to reduce the side effects of chemotherapy.


Subject(s)
Carboplatin/pharmacology , Fluorouracil/pharmacology , Hematopoietic System/drug effects , Human Growth Hormone/pharmacology , Animals , Body Weight/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Humans , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley
19.
World J Gastroenterol ; 12(24): 3810-3, 2006 Jun 28.
Article in English | MEDLINE | ID: mdl-16804963

ABSTRACT

AIM: To study effects of recombinant human growth hormone (rhGH) on growth of a human gastric carcinoma cell in vivo. METHODS: Experimental mice were divided into control group, rhGH group, oxaliplatin (L-OHP) group and rhGH+L-OHP group. Cultured human gastric carcinoma cells BGC823 were inoculated into right axilla of nude mice and carcinoma xenograft model was established successfully. Inhibitory rate of xenograft tumor growth was estimated by measuring tumor volume; expression of proliferating cell nuclear antigen (PCNA), Bax and Bcl-2 proteins of xenograft tumor was detected using immunohistochemical S-P method. RESULTS: Tumor growth inhibitory rate, the positive expression rate of PCNA, Bax and Bcl-2 were 49.3%, 58.2%, 65.2% and 59.2% in rhGH+L-OHP group respectively; 46.6%, 62.5%, 59.7% and 64.7% in L-OHP group; 5.0%, 82.7%, 23.2% and 82.2% in rhGH group and 0, 77.8%, 23.5% and 80.3% in control group. There was significant difference between rhGH+L-OHP group (or L-OHP group ) and control group or rhGH group (P < 0.05), whereas there were no significant differences (P > 0.05) between L-OHP group and rhGH+L-OHP group and between rhGH group and control group. CONCLUSION: rhGH does not accelerate the proli-feration of human gastric cancer cell in vivo.


Subject(s)
Cell Proliferation/drug effects , Human Growth Hormone/pharmacology , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Human Growth Hormone/therapeutic use , Humans , Immunohistochemistry , Mice , Mice, Nude , Proliferating Cell Nuclear Antigen/analysis , Proto-Oncogene Proteins c-bcl-2/analysis , Stomach Neoplasms/chemistry , Stomach Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , bcl-2-Associated X Protein/analysis
20.
World J Gastroenterol ; 10(8): 1132-6, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15069712

ABSTRACT

AIM: To study the effects of recombinant human growth hormone (rhGH) on growth of human gastric cancer cell line in vitro. METHODS: Experiment was divided into control group, rhGH group, oxaliplatin (L-OHP) group and rhGH+L-OHP group. Cell inhibitory rate, cell cycle, cell proliferation index (PI) and DNA inhibitory rate of human gastric cancer line BGC823, at different concentrations of rhGH treatment were studied by cell culture, MTT assay and flow cytometry. RESULTS: The distinctly accelerated effects of rhGH on multiplication of BGC823 cell line were not found in vitro. There was no statistical significance between rhGH group and control group, or between rhGH+L-OHP group and L-OHP group (P>0.05). The cell growth curve did not rise. Cell inhibitory rate and cells arrested in G(0)-G(1) phase were obviously increased. Meanwhile, cells in S phase and PI were distinctly decreased and DNA inhibitory rate was obviously increased in rhGH+L-OHP group in comparison with control group and rhGH group, respectively (P<0.01). Cell inhibitory rate showed an increasing trend and PI showed a decreasing trend in rhGH+L-OHP group compared with L-OHP group. CONCLUSION: In vitro rhGH does not accelerate the multiplication of human gastric cancer cells. It may increase the therapeutic efficacy when it is used in combination with anticancer drugs.


Subject(s)
Growth Hormone/pharmacology , Human Growth Hormone/pharmacology , Stomach Neoplasms , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Coloring Agents , DNA/biosynthesis , G1 Phase/drug effects , Humans , In Vitro Techniques , Organoplatinum Compounds/pharmacology , Oxaliplatin , Resting Phase, Cell Cycle/drug effects , Tetrazolium Salts , Thiazoles
SELECTION OF CITATIONS
SEARCH DETAIL
...