Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Medicine (Baltimore) ; 100(3): e24128, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33546022

ABSTRACT

BACKGROUND: Chinese herbal medicine injections (CHMIs) are frequently used for various refractory diseases including chronic pulmonary heart disease (CPHD). However, due to the diversity of CHMIs treatments, its relative effectiveness and safety remain unclear. In our study, Bayesian network meta-analysis will be used to identify differences in efficacy and safety between diverse CHMI for CPHD. METHODS: Relevant randomized controlled trials (RCTs) and prospective controlled clinical trials published in PubMed, Google Scholar, Excerpt Medica Database, Medline, Cochrane Library, Web of Science, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database and Wanfang Database will be systematic searched to identify eligible studies from their establishment to December 2020. The methodological qualities, including the risk of bias, will be evaluated using the Cochrane risk of bias assessment tool. Stata14.2 and WinBUGS 1.4.3 software were used for data synthesis. The evidentiary grade of the results will be also evaluated using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. RESULTS: The results of this study will be published in a peer-reviewed journal, and provide reliable evidence for different CHMIs on CPHD. CONCLUSIONS: The findings will provide reference for evaluating the efficacy and safety of different CHMIs for CPHD, and provide a helpful evidence for clinicians to formulate the best adjuvant treatment strategy for CPHD patients. TRIAL REGISTRATION NUMBER: INPLASY2020120004.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Pulmonary Heart Disease/drug therapy , Bayes Theorem , Chronic Disease/drug therapy , Humans , Injections , Meta-Analysis as Topic
2.
Stem Cell Res Ther ; 11(1): 207, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32460839

ABSTRACT

The novel coronavirus disease 2019 (COVID-19) has grown to be a global public health emergency since patients were first detected in Wuhan, China. Thus far, no specific drugs or vaccines are available to cure the patients with COVID-19 infection. The immune system and inflammation are proposed to play a central role in COVID-19 pathogenesis. Mesenchymal stem cells (MSCs) have been shown to possess a comprehensive powerful immunomodulatory function. Intravenous infusion of MSCs has shown promising results in COVID-19 treatment. Here, we report a case of a severe COVID-19 patient treated with human umbilical cord Wharton's jelly-derived MSCs (hWJCs) from a healthy donor in Liaocheng People's Hospital, China, from February 24, 2020. The pulmonary function and symptoms of the patient with COVID-19 pneumonia was significantly improved in 2 days after hWJC transplantation, and recovered and discharged in 7 days after treatment. After treatment, the percentage and counts of lymphocyte subsets (CD3+, CD4+, and CD8+ T cell) were increased, and the level of IL-6, TNF-α, and C-reactive protein is significantly decreased after hWJC treatment. Thus, the intravenous transplantation of hWJCs was safe and effective for the treatment of patients with COVID-19 pneumonia, especially for the patients in a critically severe condition. This report highlights the potential of hWJC infusions as an effective treatment for COVID-19 pneumonia.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Pneumonia, Viral/therapy , Betacoronavirus/genetics , C-Reactive Protein/immunology , C-Reactive Protein/metabolism , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Coronavirus Infections/diagnosis , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Humans , Immunomodulation , Infusions, Intravenous , Interleukin-6/blood , Interleukin-6/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/virology , Male , Mesenchymal Stem Cells/immunology , Middle Aged , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/immunology , SARS-CoV-2 , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology , Umbilical Cord/cytology , Umbilical Cord/immunology , Wharton Jelly/cytology , Wharton Jelly/immunology , COVID-19 Drug Treatment
3.
Anal Chem ; 92(9): 6430-6436, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32268724

ABSTRACT

Compared with semiconductor quantum dots and organic chromophores, carbon dots (CDs)-based lysosome probe with strong emission, an intrinsic targeting ability, and easy synthesis procedure were urgently desired in visualization imaging studies. Herein, we showed that it was possible to produce CDs with the desired properties for lysosome imaging via a one-step hydrothermal treatment of commercial reagents, rose bengal (RB) and branched polyethylenimine (bPEI). The prepared bPEI-RB CDs (P-R CDs) had a high fluorescence quantum yield (FLQY) of 90.49%, a narrow emission band of 30 nm, negligible phototoxicity, and dark toxicity. Moreover, P-R CDs had an intrinsic lysosome targeting ability without any postmodification of the ligands. Long-term cell imaging displayed P-R CDs can anchor lysosomes for up to 48 h without leakage. In addition, experimental results confirmed that dehalogenation cross-linking and structural reorganization of the reactants were the main causes of the ultrahigh photoluminescence efficiency, low cytotoxicity, and passivated surface of the P-R CDs. This origin was attributed to the restricted intersystem crossing and nonradiative transition, the reduced production of singlet oxygen, and suitable -NH2 functional groups. Due to outstanding characteristics, P-R CDs may be developed for a promising tool in lysosome images. The concept of facile preparation will also pave a new avenue for developing ultrabright functional nanomaterial markers.

4.
Biosci Rep ; 40(3)2020 03 27.
Article in English | MEDLINE | ID: mdl-32124923

ABSTRACT

Ginkgo leaf extract and dipyridamole injection (GLED), a kind of Chinese herbal medicine preparation, has been considered as a promising supplementary treatment for chronic cor pulmonale (CCP). Although an analysis of the published literature has been performed, the exact effects and safety of GLED have yet to be systematically investigated. Therefore, a wide-ranging systematic search of electronic databases from which to draw conclusions was conducted. All randomized controlled trials concerning the GLED plus conventional treatments for CCP were selected in the present study. Main outcomes were treatment efficacy, blood gas and hemorrheology indexes, and adverse events. Data from 28 trials with 2457 CCP patients were analyzed. The results indicated that, compared with conventional treatments alone, the combination of conventional treatments with GLED obviously improved the markedly effective rate (RR = 1.44, 95% CI = 1.31-1.58, P < 0.00001) and total effective rate (RR = 1.28, 95% CI = 1.18-1.38, P < 0.00001). Moreover, the hemorrheology (PaO2, P < 0.00001; PaCO2, P < 0.00001; SaO2, P < 0.00001; pH value, P = 0.05) and blood gas indexes (PV, WBHSV, WBMSV, WBLSV, hematocrit and FBG, P < 0.01) of CCP patients were also significantly ameliorated after the combined therapy. The frequency of adverse events did not differ significantly between the two groups (P > 0.05). In summary, evidence from the meta-analysis suggested that the combination of conventional treatments and GLED appeared to be effective and relatively safe for CCP. Therefore, GLED mediated therapy could be recommended as an adjuvant treatment for CCP.


Subject(s)
Dipyridamole/pharmacology , Plant Extracts/pharmacology , Pulmonary Heart Disease/drug therapy , Cardiovascular Diseases/drug therapy , Chronic Disease , Dipyridamole/metabolism , Drugs, Chinese Herbal/administration & dosage , Ginkgo biloba/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Plant Extracts/metabolism , Randomized Controlled Trials as Topic , Treatment Outcome
5.
Onco Targets Ther ; 9: 4617-27, 2016.
Article in English | MEDLINE | ID: mdl-27524915

ABSTRACT

BACKGROUND: Cellular immunotherapy has been widely used in the treatment of solid tumors. However, the clinical application of cord blood-derived dendritic cells and cytokine-induced killer cells (CB-DC-CIK) for the treatment of gastric cancer has not been frequently reported. In this study, the efficacy and safety of CB-DC-CIK for the treatment of gastric cancer were evaluated both in vitro and in vivo. METHODS: The phenotypes, cytokines, and cytotoxicity of CB-DC-CIK were detected in vitro. Patients with advanced gastric cancer were divided into the following two groups: the experimental group (CB-DC-CIK combined with chemotherapy) and the control group (chemotherapy alone). The curative effects and immune function were compared between the two groups. RESULTS: First, the results showed that combination therapy significantly increased the overall disease-free survival rate (P=0.0448) compared with chemotherapy alone. The overall survival rate (P=0.0646), overall response rate (P=0.410), and disease control rate (P=0.396) were improved in the experimental group, but these changes did not reach statistical significance. Second, the percentage of T-cell subsets (CD4(+), CD3(-)CD56(+), and CD3(+)CD56(+)) and the levels of IFN-γ, TNF-α, and IL-2, which reflect immune function, were significantly increased (P<0.05) after immunotherapy. Finally, no serious side effects appeared in patients with gastric cancer after the application of cellular immunotherapy based on CB-DC-CIK. CONCLUSION: CB-DC-CIK combined with chemotherapy is effective and safe for the treatment of patients with advanced gastric cancer.

6.
Cytotherapy ; 18(9): 1162-77, 2016 09.
Article in English | MEDLINE | ID: mdl-27421742

ABSTRACT

BACKGROUND: Currently, cytokine-induced killer cells (CIK)/dendritic cell (DC)-CIK-mediated immunotherapy is widely used to treat gastric cancer. However, limited information regarding clinical trials on CIK/DC-CIK therapy is available. Therefore, systemic evaluation of the efficacy and safety of the combination therapy is necessary. METHODS: A meta-analysis involving 1735 patients with gastric cancer was conducted. Before analysis, the study quality and heterogeneity were evaluated. The effects of chemotherapy combined with CIK/DC-CIK on gastric cancer were compared with the effects observed when chemotherapy alone was used. Pooled analysis was performed using RevMan version 5.2 from random or fixed-effect models. RESULTS: Seventeen trials were included. First, the analysis showed that the combination therapy significantly increased the overall survival rate and disease-free survival rate compared with those in patients treated using chemotherapy alone. The overall response rate (P = 0.002), disease control rate (P = 0.0007), and quality of life improved rate (P = 0.0008) were significantly improved in patients who received combined treatment than in patients who received chemotherapy alone. Second, the percentage of lymphocyte subsets (CD3(+), CD4(+) and CD3(-)CD56(+), CD3(+)CD56(+); P <0.01) and the levels of interleukin-12 and interferon-γ, which reflect immune function, were significantly increased (P <0.05) after the CIK/DC-CIK therapy. Further, carbohydrate antigen tumor markers were significantly reduced compared with the pre-therapy levels. Immunotherapy with CIK/DC-CIK obviously alleviated the adverse events caused by chemotherapy. CONCLUSION: The combination of CIK/DC-CIK therapy and chemotherapy was superior in prolonging the survival time, enhancing immune function and alleviating the adverse events caused by chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell- and Tissue-Based Therapy/methods , Cytokine-Induced Killer Cells , Stomach Neoplasms/therapy , China , Combined Modality Therapy , Cytokine-Induced Killer Cells/immunology , Dendritic Cells/immunology , Disease-Free Survival , Humans , Immunotherapy/methods , Lymphocyte Subsets , Quality of Life , Stomach Neoplasms/mortality , Survival Rate , Treatment Outcome
8.
Acta Pharmacol Sin ; 32(10): 1259-65, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21909126

ABSTRACT

AIM: To investigate whether the combination of fluvastatin and losartan synergistically relieve atherosclerosis and plaque inflammation induced by a high-cholesterol diet in rabbits. METHODS: Atherosclerosis was induced with a high-cholesterol diet for 3 months in 36 New Zealand white rabbits. The animals were randomly divided into model group, fluvastatin (10 mg·kg(-1)·d(-1)) group, losartan (25 mg·kg(-1)·d(-1)) group, and fluvastatin plus losartan group. After the 16-week treatments, the blood samples the animals were collected, and the thoracic aortas were examined immunohistochemically. The mRNA and protein expression levels of monocyte chemotactic protein-1 (MCP-1) were measured using RT-PCR and Western blot. RESULTS: Compared to the treatment with losartan or fluvastatin alone, the combined treatment did not produce higher efficacy in reduction of blood cholesterol level. However, the combination did synergistically decrease the intimal and media thickness of thoracic aortas with significantly reduced macrophage infiltration and MCP-1 expression in the plaques. CONCLUSION: The combined treatment with losartan and fluvastatin significantly inhibited atherosclerotic progress and reduced inflammation associated with atherosclerotic plaques.


Subject(s)
Atherosclerosis/drug therapy , Fatty Acids, Monounsaturated/therapeutic use , Indoles/therapeutic use , Losartan/therapeutic use , Macrophages/drug effects , Plaque, Atherosclerotic/drug therapy , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Chemokine CCL2/genetics , Cholesterol/blood , Drug Synergism , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Gene Expression Regulation/drug effects , Indoles/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Losartan/pharmacology , Macrophages/pathology , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Rabbits , p38 Mitogen-Activated Protein Kinases/genetics
9.
Mol Biol Rep ; 38(2): 949-55, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20533091

ABSTRACT

Ornithine decarboxylase (ODC), the first rate-limiting enzyme of polyamine biosynthesis, was found to be associated with cell growth, proliferation and transformation. ODC gene expression in gastric cancer was increased and its level was positively correlated with the degree of malignity of gastric mucosa and development of gastric lesions. In order to evaluate the therapeutic effects of antisense RNA of ODC on gastric cancer, an antisense RNA of ODC expressing plasmid pcDNA-ODCr which delivered a 120 bp fragment complementary to the initiation codon of ODC gene was constructed and transfected to gastric cancer cells SGC7901 and MGC803. Expression of ODC in gastric cancer cells was determined by western blot. Cell proliferation was assessed by MTS assay. Cell cycle was analyzed by flow cytometry and Matrigel assay was performed to assess the ability of gastric cancer cell invasiveness. The results showed that the ODC gene expression in gastric cancer cells transfected with the pcDNA-ODCr was downregulated efficiently. Tumor cell proliferation was suppressed significantly, and cell cycle was arrested at G1 phase. Gastric cancer cells had reduced invasiveness after gene transfer. Our study suggested that antisense RNA of ODC expressing plasmid pcDNA-ODCr had antitumor activity by inhibiting the expression of ODC, and downregulation of ODC expression using a gene therapy approach might be a novel therapeutic strategy for gastric cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Ornithine Decarboxylase/biosynthesis , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Collagen/chemistry , Drug Combinations , Flow Cytometry/methods , Gene Expression Profiling , Gene Transfer Techniques , Genetic Therapy/methods , Humans , Laminin/chemistry , Oligonucleotides, Antisense/genetics , Plasmids/metabolism , Proteoglycans/chemistry , RNA/metabolism
10.
Chin Med J (Engl) ; 123(16): 2269-73, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20819678

ABSTRACT

BACKGROUND: Tumstatin is a novel endogenous angiogenesis inhibitor which is widely studied using purified protein. The current study evaluates the antiangiogenic effects of tumstatin-overexpression plasmid in vitro, reveals the mechanism underlying the vascular endothelial cell growth inhibition and searches for a novel method administering tumstatin persistently. METHODS: The eukaryotic expression plasmid pcDNA-tumstatin encoding tumstatin gene was constructed and transfected to human umbilical vein endothelial cell ECV304 and human renal carcinoma cell ACHN. Expression of tumstatin in the two cell lines was determined by RT-PCR and Western blotting. Vascular endothelial cell proliferation was assessed by CCK-8 assay and cell cycle was analyzed by flow cytometry. To investigate the mechanism by which pcDNA-tumstatin inhibited vascular endothelial cell proliferation in vitro, cyclin D1 protein was detected by Western blotting. RESULTS: DNA sequence confirmed that pcDNA-tumstatin was successfully constructed. RT-PCR and Western blotting indicated that tumstatin could express in the two cell lines effectively. After tumstatin gene transfer, ECV304 cell growth was significantly inhibited and the cell cycle was arrested in G1 phase. And Western blotting showed that pcDNA-tumstatin decreased the level of cyclin D1 protein. CONCLUSIONS: Overexpression of tumstatin mediated by pcDNA 3.1 (+) specially inhibited vascular endothelial cells by arresting vascular endothelial cell in G1 phase resulting from downregulation of cyclin D1 and administration of tumstatin using a gene therapy might be a novel strategy for cancer therapy.


Subject(s)
Autoantigens/genetics , Autoantigens/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Humans , Plasmids/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Zhonghua Xin Xue Guan Bing Za Zhi ; 37(7): 622-5, 2009 Jul.
Article in Chinese | MEDLINE | ID: mdl-19961735

ABSTRACT

OBJECTIVE: Angiotensin converting enzyme 2 (ACE2) efficiently hydrolyses the potent vasoconstrictor angiotensin II to vasodilative angiotensin (1-7). We hypothesized that ACE2 overexpression may inhibit inflammation response in atherosclerotic plaque by degrading Ang II into Ang-(1-7). METHODS: Atherosclerosis (AS) plaques were induced in the abdominal aorta of 38 rabbits by endothelial injury and atherogenic diet for 3 months. Rabbits were then underwent injection of a recombinant adenovirus (2.5 x 10(9) pfu/ml) carrying a murine ACE2 gene (Ad-ACE2) through a catheter into the abdominal aortic segments rich in plaques (n = 19) or injection of a control vector Ad-EGFP (n = 19). One month later, all rabbits were sacrificed and plaques from aortic segments were analyzed. RESULTS: ACE2 expression in aortic tissues of the Ad-ACE2 group were confirmed by immunohistochemistry. Macrophage infiltration (13.6% +/- 4.2% vs. 23.6% +/- 6.9%, P < 0.01) and MCP-1 expression (13.2% +/- 0.4% vs. 25.0% +/- 7.4%, P < 0.01) were significantly reduced in Ad-ACE2 group compared to Ad-EGFP group. CONCLUSIONS: Overexpression of ACE2 inhibited atherosclerotic plaque inflammation response in hypercholesterolemic rabbits.


Subject(s)
Atherosclerosis/genetics , Peptidyl-Dipeptidase A/genetics , Transfection , Angiotensin-Converting Enzyme 2 , Animals , Atherosclerosis/metabolism , Cells, Cultured , Diet, Atherogenic , Genetic Vectors , Rabbits
12.
Cancer Sci ; 100(11): 2126-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19686286

ABSTRACT

Spermidine/spermine N(1)-acetyltransferase (SSAT) is the rate-limiting step in polyamine catabolism. In a previous study, we constructed a recombinant adenovirus, Ad-SSAT, which can express human SSAT. In the present study, we investigated the effect of upregulated and downregulated SSAT on gastric cancer cells. We found that upregulated SSAT could inhibit the growth of MGC803 and SGC7901 cells, whereas adverse results were found with downregulated SSAT. We further analyzed cell cycle profiles and the expression levels of the major cell cycle regulatory proteins of S phase. The results showed that the growth inhibition was caused by S phase arrest. Ad-SSAT suppressed the expression of cyclin A and nuclear factor E2F1 in MGC803 and SGC7901 cells. We observed the E2F promoter activity caused by Ad-SSAT using a reporter gene assay. We also investigated the antitumorigenicity of upregulated SSAT by Ad-SSAT using a SGC7901 xenograft model in nude mice. Our results suggest that the upregulation of SSAT by Ad-SSAT infection inhibited the growth of gastric cancer in vitro and in vivo. Ad-SSAT arrested gastric cancer cells in S phase, which was mediated through downregulation of the cyclin A-E2F signaling pathway.


Subject(s)
Acetyltransferases/physiology , Adenoviridae/genetics , Stomach Neoplasms/prevention & control , Acetyltransferases/genetics , Animals , Biogenic Polyamines/analysis , Cell Line, Tumor , Cyclin A/antagonists & inhibitors , E2F1 Transcription Factor/antagonists & inhibitors , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , S Phase , Stomach Neoplasms/chemistry , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Up-Regulation
13.
Oncol Rep ; 20(5): 1229-35, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18949426

ABSTRACT

Spermidine/spermine N1-acetyltransferase (SSAT) is a key enzyme of polyamine catabolism. In a previous study, we constructed a recombinant adenovirus, Ad-SSAT, which can express human SSAT. In the present study, we investigated the effect of Ad-SSAT on the growth and cell cycle of colorectal cancer cells. We found that Ad-SSAT increased the expression of SSAT and inhibited the growth of HT-29 and Lovo cells. The growth inhibition was caused by cell cycle arrest in the S phase. Furthermore, Ad-SSAT was shown to suppress the expression of cyclin A and nuclear factor E2F-1 in HT-29 and Lovo cells. The inhibitory effect of Ad-SSAT on cyclin A promoter activity was also observed in a reporter gene assay. Our results suggest that the expression of SSAT mediated by Ad-SSAT infection inhibits the growth of colorectal cancer cells and induces cell cycle arrest at the S phase, through a mechanism involving the suppression of cyclin A and E2F-1 expression.


Subject(s)
Acetyltransferases/genetics , Cell Cycle/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Spermidine/physiology , Acetyltransferases/metabolism , Adenoviridae/genetics , Blotting, Western , Cell Line, Tumor , Cyclin A/metabolism , E2F1 Transcription Factor/metabolism , Gene Expression , Genetic Vectors , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
Acta Pharmacol Sin ; 29(5): 606-13, 2008 May.
Article in English | MEDLINE | ID: mdl-18430370

ABSTRACT

AIM: To construct a recombinant adenovirus that can express human spermidine/ spermine N1-acetyltransferase (SSAT) and detect its inhibitory effect on colorectal cancer cell growth in vitro. METHODS: A 516 bp cDNA of SSAT was amplified and cloned into a pGL3-hTERT plasmid. The pGL3-hTERT-SSAT recombinant was digested, and the small fragment was cloned into the shuttle vector pAdTrack. The pAdTrack-hTERT-SSAT plasmids were recombined with pAdEasy-1 vectors in AdEasy-1 cells. Positive clones were selected and transfected into the HEK293 packaging cells (transformed human embryonic kidney cells) after they were linearized by PacI. The process of adenovirus packaging and amplification was monitored by green fluorescent protein (GFP) expression. The SSAT protein levels were determined by Western blotting, and the intracellular polyamine content was detected by reverse-phase high performance liquid chromatography. The MTS (3-(4, 5-dimethylthiaol-2-yl)-5-(3-carboxy-methoxyphenyl)-2-(-4- sulfophenyl)-2H-tetrazolium, inner salt) and colony-forming assays were used to analyze the gene transduction efficiency and effect on the growth of HT-29 and LoVo cells. A viable cell count was used to determine the cell growth with or without exogenous polyamines. RESULTS: The GFP expression in 293 cells during virus packing and amplification was observed by fluorescence microscopy. Western blotting results demonstrated that Ad-hTERT-SSAT could increase the expression of SSAT, and consequently, spermidine and spermine were reduced to low levels. The MTS and colony-forming assay results showed that HT-29 and LoVo cell growth were significantly inhibited, and the inhibitory effect could be partially reversed by exogenous spermidine and spermine. CONCLUSION: The successfully constructed recombinant adenovirus Ad-hTERT-SSAT could accelerate polyamine catabolism and inhibit the colorectal cell growth in vitro. It also has therapeutic potential in the treatment of colorectal cancer.


Subject(s)
Acetyltransferases/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Spermidine/metabolism , Spermine/metabolism , Acetyltransferases/metabolism , Adenoviridae/genetics , Base Sequence , Cell Line , DNA, Complementary/chemistry , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/metabolism , HT29 Cells , Humans , Kidney/cytology , Molecular Sequence Data , Plasmids , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...