Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 12(8): 775, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34362880

ABSTRACT

The cysteine protease, caspase-8, undergoes dimerization, processing, and activation following stimulation of cells with death ligands such as TRAIL, and mediates TRAIL induction of the extrinsic apoptosis pathway. In addition, caspase-8 mediates TRAIL-induced activation of NF-κB and upregulation of immunosuppressive chemokines/cytokines, via a mechanism independent of caspase-8 catalytic activity. The gene encoding procaspase-8 is mutated in 10% of human head and neck squamous cell carcinomas (HNSCCs). Despite a paucity of experimental evidence, HNSCC-associated caspase-8 mutations are commonly assumed to be loss of function. To investigate their functional properties and phenotypic effects, 18 HNSCC-associated caspase-8 mutants were expressed in doxycycline-inducible fashion in cell line models wherein the endogenous wild-type caspase-8 was deleted. We observed that 5/8 mutants in the amino-terminal prodomain, but 0/10 mutants in the carboxyl-terminal catalytic region, retained an ability to mediate TRAIL-induced apoptosis. Caspase-8 proteins with mutations in the prodomain were defective in dimerization, whereas all ten of the catalytic region mutants efficiently dimerized, revealing an inverse relationship between dimerization and apoptosis induction for the mutant proteins. Roughly half (3/8) of the prodomain mutants and 9/10 of the catalytic region mutants retained the ability to mediate TRAIL induction of immunosuppressive CXCL1, IL-6, or IL-8. Doxycycline-induced expression of wild-type caspase-8 or a representative mutant led to an increased percentage of T and NKT cells in syngeneic HNSCC xenograft tumors. These findings demonstrate that HNSCC-associated caspase-8 mutants retain properties that may influence TRAIL-mediated apoptosis and cytokine induction, as well as the composition of the tumor microenvironment.


Subject(s)
Apoptosis/genetics , Caspase 8/genetics , Cytokines/metabolism , Head and Neck Neoplasms/genetics , Mutation/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Apoptosis/drug effects , Female , HeLa Cells , Humans , Immunosuppression Therapy , Mice, Inbred C57BL , Protein Multimerization , Tumor Microenvironment/drug effects
2.
Mol Cancer Res ; 19(1): 14-24, 2021 01.
Article in English | MEDLINE | ID: mdl-32883697

ABSTRACT

Genetic alterations of CYLD lysine 63 deubiquitinase (CYLD), a tumor-suppressor gene encoding a deubiquitinase (DUB) enzyme, are associated with the formation of tumors in CYLD cutaneous syndrome. Genome sequencing efforts have revealed somatic CYLD alterations in multiple human cancers. Moreover, in cancers commonly associated with human papillomavirus (HPV) infection (e.g., head and neck squamous cell carcinoma), CYLD alterations are preferentially observed in the HPV-positive versus HPV-negative form of the disease. The CYLD enzyme cleaves K63-linked polyubiquitin from substrate proteins, resulting in the disassembly of key protein complexes and the inactivation of growth-promoting signaling pathways, including pathways mediated by NF-κB, Wnt/ß-catenin, and c-Jun N-terminal kinases. Loss-of-function CYLD alterations lead to aberrant activation of these signaling pathways, promoting tumorigenesis and malignant transformation. This review summarizes the association and potential role of CYLD somatic mutations in HPV-positive cancers, with particular emphasis on the role of these alterations in tumorigenesis, invasion, and metastasis. Potential therapeutic strategies for patients whose tumors harbor CYLD alterations are also discussed. IMPLICATIONS: Alterations in CYLD gene are associated with HPV-associated cancers, contribute to NF-κB activation, and are implicated in invasion and metastasis.


Subject(s)
Deubiquitinating Enzyme CYLD/genetics , Head and Neck Neoplasms/virology , Papillomaviridae/pathogenicity , Carcinogenesis , Disease Progression , Humans , Neoplasm Metastasis
3.
Theranostics ; 10(24): 10957-10972, 2020.
Article in English | MEDLINE | ID: mdl-33042264

ABSTRACT

Rationale: RNA helicase DDX5 is downregulated during hepatitis B virus (HBV) replication, and poor prognosis HBV-related hepatocellular carcinoma (HCC). The aim of this study is to determine the mechanism and significance of DDX5 downregulation for HBV-driven HCC, and identify biologics to prevent DDX5 downregulation. Methods: Molecular approaches including immunoblotting, qRT-PCR, luciferase transfections, hepatosphere assays, Assay for Transposase-Accessible Chromatin sequencing (ATAC-seq), and RNA-seq were used with cellular models of HBV replication, HBV infection, and HBV-related liver tumors, as well as bioinformatic analyses of liver cancer cells from two independent cohorts. Results: We demonstrate that HBV infection induces expression of the proto-oncogenic miR17~92 and miR106b~25 clusters which target the downregulation of DDX5. Increased expression of these miRNAs is also detected in HBV-driven HCCs exhibiting reduced DDX5 mRNA. Stable DDX5 knockdown (DDX5KD) in HBV replicating hepatocytes increased viral replication, and resulted in hepatosphere formation, drug resistance, Wnt activation, and pluripotency gene expression. ATAC-seq of DDX5KD compared to DDX5 wild-type (WT) cells identified accessible chromatin regions enriched in regulation of Wnt signaling genes. RNA-seq analysis comparing WT versus DDX5KD cells identified enhanced expression of multiple genes involved in Wnt pathway. Additionally, expression of Disheveled, DVL1, a key regulator of Wnt pathway activation, was significantly higher in liver cancer cells with low DDX5 expression, from two independent cohorts. Importantly, inhibitors (antagomirs) to miR17~92 and miR106b~25 restored DDX5 levels, reduced DVL1 expression, and suppressed both Wnt activation and viral replication. Conclusion: DDX5 is a negative regulator of Wnt signaling and hepatocyte reprogramming in HCCs. Restoration of DDX5 levels by miR17~92 / miR106b~25 antagomirs in HBV-infected patients can be explored as both antitumor and antiviral strategy.


Subject(s)
Antagomirs/pharmacology , Carcinoma, Hepatocellular/drug therapy , DEAD-box RNA Helicases/genetics , Hepatitis B, Chronic/drug therapy , Liver Neoplasms/drug therapy , Wnt Signaling Pathway/genetics , Antagomirs/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , DEAD-box RNA Helicases/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Hepatocytes , Humans , Liver/pathology , Liver/virology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA-Seq , Virus Replication/drug effects , Virus Replication/genetics , Wnt Signaling Pathway/drug effects
4.
Cancers (Basel) ; 12(2)2020 02 11.
Article in English | MEDLINE | ID: mdl-32053966

ABSTRACT

The mechanisms that drive ductal carcinoma in situ (DCIS) progression to invasive cancer are not clear. Studying DCIS progression in humans is challenging and not ethical, thus necessitating the characterization of an animal model that faithfully resembles human disease. We have characterized a canine model of spontaneous mammary DCIS and invasive cancer that shares histologic, molecular, and diagnostic imaging characteristics with DCIS and invasive cancer in women. The purpose of the study was to identify markers and altered signaling pathways that lead to invasive cancer and shed light on early molecular events in breast cancer progression and development. Transcriptomic studies along the continuum of cancer progression in the mammary gland from healthy, through atypical ductal hyperplasia (ADH), DCIS, and invasive carcinoma were performed using the canine model. Gene expression profiles of preinvasive DCIS lesions closely resemble those of invasive carcinoma. However, certain genes, such as SFRP2, FZD2, STK31, and LALBA, were over-expressed in DCIS compared to invasive cancer. The over-representation of myoepithelial markers, epithelial-mesenchymal transition (EMT), canonical Wnt signaling components, and other pathways induced by Wnt family members distinguishes DCIS from invasive. The information gained may help in stratifying DCIS as well as identify actionable targets for primary and tertiary prevention or targeted therapy.

6.
Sci Rep ; 7: 40325, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28067315

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are a type of rare sarcomas with a poor prognosis due to its highly invasive nature and limited treatment options. Currently there is no targeted-cancer therapy for this type of malignancy. Thus, it is important to identify more cancer driver genes that may serve as targets of cancer therapy. Through comparative oncogenomics, we have found that KANK1 was a candidate tumor suppressor gene (TSG) for human MPNSTs. Although KANK1 is known as a cytoskeleton regulator, its tumorigenic function in MPNSTs remains largely unknown. In this study, we report that restoration of KANK1 in human MPNST cells inhibits cell growth both in human cell culture and xenograft mice by increasing apoptosis. Consistently, knockdown of KANK1 in neurofibroma cells promoted cell growth. Using RNA-seq analysis, we identified CXXC5 and other apoptosis-related genes, and demonstrated that CXXC5 is regulated by KANK1. Knockdown of CXXC5 was found to diminish KANK1-induced apoptosis in MPNST cells. Thus, KANK1 inhibits MPNST cell growth though CXXC5 mediated apoptosis. Our results suggest that KANK1 may function as a tumor suppressor in human MPNSTs, and thus it may be useful for targeted therapy.


Subject(s)
Apoptosis , Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cytoskeletal Proteins , DNA-Binding Proteins , Gene Dosage , Gene Knockdown Techniques , Humans , Mice, Inbred NOD , Mice, SCID , Transcription Factors , Xenograft Model Antitumor Assays , Zebrafish
7.
Sci Rep ; 6: 27816, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27292017

ABSTRACT

Gene co-option, usually after gene duplication, in the evolution of development is found to contribute to vertebrate morphological innovations, including the endothelium-based vascular system. Recently, a zebrafish kank gene was found expressed in the vascular vessel primordium, suggesting KANK genes are a component of the developmental tool kit for the vertebrate vascular system. However, how the KANK gene family is involved in vascular vessel development during evolution remains largely unknown. First, we analyzed the molecular evolution of the KANK genes in metazoan, and found that KANK1, KANK2, KANK3 and KANK4 emerged in the lineage of vertebrate, consistent with the two rounds of vertebrate whole-genome duplications (WGD). Moreover, KANK genes were further duplicated in teleosts through the bony-fish specific WGD, while only kank1 and kank4 duplicates were retained in some of the examined fish species. We also found all zebrafish kank genes, except kank1b, are primarily expressed during embryonic vascular development. Compared to invertebrate KANK gene expression in the central nervous system, the vascular expression of zebrafish kank genes suggested KANK genes were co-opted for vertebrate vascular development. Given the cellular roles of KANK genes, our results suggest that this co-option may facilitate the evolutionary origin of vertebrate vascular vessels.


Subject(s)
Evolution, Molecular , Tumor Suppressor Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Bayes Theorem , Blood Vessels/growth & development , Blood Vessels/metabolism , Chromosomes/genetics , Embryo, Nonmammalian/metabolism , Gene Duplication , Gene Expression Regulation, Developmental , In Situ Hybridization , Phylogeny , Tumor Suppressor Proteins/classification , Tumor Suppressor Proteins/metabolism , Zebrafish/growth & development , Zebrafish Proteins/classification , Zebrafish Proteins/metabolism
8.
Molecules ; 21(2): 140, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26821007

ABSTRACT

Miracle fruit (Synsepalum dulcificum) belongs to the Sapotaceae family. It can change flavors on taste buds, transforming acidic tastes to sweet. We evaluated various miracle fruit extracts, including water, butanol, ethyl acetate (EA), and hexane fractions, to determine its antioxidant effects. These extracts isolated from miracle fruit exerted potential for reduction of uric acid and inhibited xanthine oxidase activity in vitro and in monosodiumurate (MSU)-treated RAW264.7 macrophages. Moreover, we also found that the butanol extracts of miracle fruit attenuated oxonic acid potassium salt-induced hyperuricaemia in ICR mice by lowering serum uric acid levels and activating hepatic xanthine oxidase. These effects were equal to those of allopurinol, suggesting that the butanol extract of miracle fruit could be developed as a novel anti-hyperuricaemia agent or health food.


Subject(s)
Antioxidants/administration & dosage , Butanols/administration & dosage , Hyperuricemia/drug therapy , Plant Extracts/analysis , Synsepalum/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Butanols/chemistry , Butanols/pharmacology , Disease Models, Animal , Hyperuricemia/blood , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred ICR , Plant Extracts/administration & dosage , Plant Extracts/chemistry , RAW 264.7 Cells , Uric Acid/blood , Xanthine Oxidase/metabolism
9.
Mar Drugs ; 14(1): 2, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26703631

ABSTRACT

Pardaxin (H-GFFALIPKIISSPLFKTLLSAVGSALSSSGGQE-OH), a 33-amino-acid polypeptide, is an antimicrobial peptide (AMP) isolated from the marine fish species Pardachirus marmoratus. Pardaxin shows antibacterial and antitumor activities. However, pardaxin-induced inhibition of oral cancer and the mechanism of tumor reduction in buccal pouch carcinogenesis after pardaxin painting remain undetermined. Additionally, the toxic effects of pardaxin on normal tissue remain unclear. The present study investigated the anticancer activity of pardaxin in oral squamous cell carcinoma (OSCC) cells in the hamster buccal pouch model with or without 7,12-dimethylbenz[a]anthracene (DMBA) pretreatment. This is the first study to confirm the effects of pardaxin on normal tissue and its nontoxic effects in vivo. Cell viability assays and colony formation tests in OSCC cell lines (SCC-4) demonstrated that pardaxin reduced cell viability in a dose-dependent manner. Immunofluorescence staining of cleaved caspase-3 in SCC-4 cells revealed that expression of activated caspase-3 in SCC-4 cells significantly increased after 24-h treatment with pardaxin. Additionally, a cell cycle analysis indicated that pardaxin treatment resulted in the cell cycle arrest of SCC-4 cells in the G2/M phase, thereby limiting cell proliferation. Furthermore, pardaxin treatment substantially alleviated carcinogenesis in the DMBA-induced hamster buccal pouch model by lowering prostaglandin E2 levels. These results suggest that pardaxin is a potential marine drug for adjuvant chemotherapy for human OSCC and oral cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Fish Venoms/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor/drug effects , Cricetinae , Disease Models, Animal , Fish Venoms/chemistry , Fish Venoms/therapeutic use , Humans , In Vitro Techniques , Mice , Mouth Neoplasms/drug therapy
10.
Oncol Rep ; 33(5): 2454-60, 2015 May.
Article in English | MEDLINE | ID: mdl-25777981

ABSTRACT

PLU-1/JARID1B (jumonji AT rich interactive domain 1B) is one of the testis cancer antigens and functions as a histone demethylase in the regulation of various human types of cancers. However, its functions in head and neck squamous cell carcinoma (HNSCC) are rarely reported. The aim of the present study was to examine PLU-1/JARID1B expression levels in HNSCCs and to investigate its role in cancer cell proliferation. In the present study, we found that PLU-1/JARID1B mRNA was upregulated in all tested HNSCC cell lines. Immunohistochemical staining showed that PLU-1/JARID1B protein expression was detected in 87.8% (87/99) of the HNSCC cases. A positive association was observed between high PLU-1/JARID1B expression and higher Ki-67 labeling in the HNSCC samples (Pearson r=0.6514, P=0.0003). Stable PLU-1/JARID1B knockdown by PLU-1-shRNAs in the HNSCC cell lines suppressed cell growth both in the in vitro and in vivo studies. Moreover, PLU-1/JARID1B knockdown resulted in G1 arrest and early apoptosis by suppressing Bcl-2 family members in the HNSCCs. These data indicate that PLU-1/JARID1B is overexpressed in HNSCCs and is associated with tumor proliferation and apoptosis. Therefore, PLU-1/JARID1B represents a candidate proliferation biomarker for HNSCC diagnosis and treatment.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Jumonji Domain-Containing Histone Demethylases/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , G1 Phase Cell Cycle Checkpoints/genetics , Humans , Mice , Mice, Nude , Proto-Oncogene Proteins c-bcl-2/genetics , Squamous Cell Carcinoma of Head and Neck
11.
Int Rev Cell Mol Biol ; 314: 149-70, 2015.
Article in English | MEDLINE | ID: mdl-25619717

ABSTRACT

Aneuploidy is one of the most common genetic alterations in cancer cell genomes. It greatly contributes to the heterogeneity of cancer cell genomes, and its roles in tumorigenesis are attracting more and more attentions. Zebrafish is emerging as a new genetic model for many human diseases including cancer. The zebrafish cancer model has shown an equivalent degree of aneuploidy as found in corresponding human cancers, thus it provides a great tool for us to study cancer aneuploidy and, in general, cancer biology. Here, we discuss some new advances of aneuploidy and the potential usages of this cancer model system.


Subject(s)
Aneuploidy , Fish Diseases/metabolism , Genome , Neoplasms, Experimental/genetics , Zebrafish/genetics , Animals , Fish Diseases/genetics , Fish Diseases/pathology , Humans , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Zebrafish/metabolism
12.
PLoS One ; 8(5): e63887, 2013.
Article in English | MEDLINE | ID: mdl-23717505

ABSTRACT

Tripartite motif-containing 24 (TRIM24), a member of the transcriptional intermediary factor 1 family, functions as a co-regulator that positively or negatively modulates the transcriptional activities of several nuclear receptors. The aim of this study was to investigate TRIM24 expression and its clinical significance in head and neck squamous cell carcinoma. The expression levels of TRIM24 variants were examined in head and neck squamous cell carcinoma (HNSCC) samples and cell lines by real-time PCR and WB. The expression levels of TRIM24 measured in 91 locally advanced HNSCC tumors were measured by immunohistochemistry and correlated with clinical and pathological parameters. The functional role of TRIM24 in HNSCC was further investigated by silencing its expression in HNSCC cell lines. TRIM24 variants were up-regulated in 56 HNSCC samples (P<.001) and 9 HNSCC cell lines (P<.05). TRIM24 protein was overexpressed in 6 of 8 HNSCC cell lines and in 2 of 3 HNSCC samples. Furthermore, 54.95% (50/91) of HNSCC samples exhibited remarkably elevated expression of TRIM24 by immunohistochemistry. Univariate analysis revealed that high TRIM24 expression was associated with worse overall survival (P = .020). In multivariate analysis, TRIM24 expression was identified as an independent predictor of overall survival (P = .030), after adjusting for other clinicopathological parameters. Upon TRIM24 silencing, the proliferation of HNSCC cells was notably inhibited due to the induction of apoptosis. These results suggest that aberrant TRIM24 expression may play an important role in the development of HNSCC and is a promising prognostic indicator for patients with locally advanced HNSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carrier Proteins/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Prognosis , Receptors, Cytoplasmic and Nuclear/genetics , Squamous Cell Carcinoma of Head and Neck , Up-Regulation/genetics
13.
Cancer ; 118(11): 2858-71, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-21989926

ABSTRACT

BACKGROUND: The authors previously observed that enhancer of zeste homolog 2 (EZH2) overexpression was associated significantly with the development of oral cancer. In the current study, they investigated whether EZH2 can function as a prognostic predictor for patients with head and neck squamous cell carcinoma (HNSCC). METHODS: Expression levels of EZH2 in HNSCC cells were detected using reverse transcriptase-polymerase chain reaction (PCR) and Western blot analyses. In addition, the effects of EZH2 ablation on the proliferation and invasion of HNSCC cells were investigated through small interfering RNA (siRNA)-mediated knockdown. Real-time PCR and immunohistochemistry were used to evaluate EZH2 and cyclin D1 expression in 46 HNSCC samples, and the expression levels also were re-evaluated in 124 independent samples by immunohistochemistry. RESULTS: EZH2 expression was elevated remarkably in HNSCC specimens and cell lines. Upon EZH2 silencing, the proliferation and invasion of HNSCC cells were remarkably suppressed. EZH2 expression frequently was correlated with cyclin D1 expression (P = .034) and tumor differentiation (P = .020). In addition, both EZH2 messenger RNA levels and EZH2 protein levels were strongly associated with signs of histologic severity (P = .012 and P = .032, respectively). Univariate analysis revealed that high EZH2 expression was associated with worse overall survival (P = .001) and disease-free survival (P = .002). The combined expression of EZH2 and cyclin D1 had superior prognostic ability for patients with HNSCC than the expression of either marker alone. In multivariate analysis, EZH2 expression was identified as an independent predictor of overall and disease-free survival. CONCLUSIONS: The current results indicated that EZH2 is an independent prognostic indicator for patients with HNSCC. In addition, an analysis of the combined expression of EZH2 and cyclin D1 can serve as a more powerful prognostic predictor for patients with HNSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cyclin D1/metabolism , DNA-Binding Proteins/genetics , Head and Neck Neoplasms/genetics , Transcription Factors/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , Female , Gene Silencing , Head and Neck Neoplasms/metabolism , Humans , Male , Middle Aged , Polycomb Repressive Complex 2 , Prognosis , Squamous Cell Carcinoma of Head and Neck , Transfection , Up-Regulation
14.
Guang Pu Xue Yu Guang Pu Fen Xi ; 28(3): 642-4, 2008 Mar.
Article in Chinese | MEDLINE | ID: mdl-18536432

ABSTRACT

Hot water extracting and ethanol precipitating method was employed to isolate polysaccharides. RCP (Rhodiola crenulata polysaccharide) was fractionally precipitated with EtOH. RCP3 (Rhodiola crenulata polysaccharide 3) was one of the three fractions. RCPS was obtained after RCP3 was purified by deproteination; decolourization and gel chromatography on Sephadex G-100. The homogeneity and molecular masses of RCPS were proved by HLGPC. The amount of total carbohydrates of RCPS was measured with phenol-sulfuric acid method. IR spectrometry and UV-spectrophotometer were used to determine the characteristic absorption of RCPS. The monosaccharides contained in the RCPS were analyzed by GC. The amount of total carbohydrates in RCPS is 99.11%. The molecular weight was 27 876. IR spectrometry analysis indicated that RCPS showed typical signals of acid polysaccharide, including signals at 3 424.83, 2 934.10, 1 742.11, 1 438.96, 1 261.40, 1 103.54 and 832.86 cm(-1); UV-spectrophotometer analysis indicated that RCPS showed a signal of polysaccharide at 195 nm and no signals of protein, nucleic acid at 260 and 280 nm. The monosaccharide constituents of RCPS were Rha, Ara, Xyl, Man, Glu, Gal and GalA, and their molar proportions were 1 : 2.96 : 0.21 : 0.26 : 0.08 : 0.58 and 0.15, respectively.


Subject(s)
Polysaccharides/analysis , Rhodiola/chemistry , Chromatography, Gel , Molecular Weight , Polysaccharides/isolation & purification , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet
15.
Zhonghua Nan Ke Xue ; 8(2): 117-9, 2002.
Article in Chinese | MEDLINE | ID: mdl-12479025

ABSTRACT

OBJECTIVES: To evaluate the effects of antidepressant Seroxat on premature ejaculation. METHODS: After having taken Seroxat 20 mg every noon for four weeks, the patients were asked to fill the investigating questionaire about the effects and side effects before and after the treatment. RESULTS: After the treatment, 43 cases in our study had increased their ejaculation latency time, enhanced the quality of their sexy lives and their wives', and had significant difference compared with pre-treatment(P < 0.001). They had good effects on improving premature ejaculation after taking Seroxat (11.26 +/- 5.79) days; and after having stopped taking seroxat for (20.94 +/- 8.04) days, the situation of premature ejaculation in 32 cases were as same as that of before. There were seven cases whose sexuality and oomph increased, and two cases whose sexuality were decreased. A few patients had constipation, dry in mouth, insomnia and itch in skin, after taking the drugs. CONCLUSIONS: Antidepressant Seroxat has rather good effects on premature ejaculation and should be used and studied further.


Subject(s)
Antidepressive Agents/therapeutic use , Paroxetine/therapeutic use , Sexual Dysfunction, Physiological/drug therapy , Adult , Antidepressive Agents/adverse effects , Ejaculation/drug effects , Humans , Male , Middle Aged , Paroxetine/adverse effects , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...