Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Biomed Sci ; 24(1): 51, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28750624

ABSTRACT

BACKGROUND: Obesity and its comorbidities constitute a serious health burden worldwide. Leptin plays an important role in diet control; however, it has a stimulatory potential on cancer cell proliferation. The OB3 peptide, a synthetic peptide, was shown to be more active than leptin in regulating metabolism but with no mitogenic effects in cancer cells. METHODS: In this study, we investigated the proliferative effects, gene expressions and signaling pathways modulated by leptin and OB3 in human ovarian cancer cells. In addition, an animal study was performed. RESULTS: Leptin, but not OB3, induced the proliferation of ovarian cancer cells. Interestingly, OB3 blocked the leptin-induced proliferative effect when it was co-applied with leptin. Both leptin and OB3 activated the phosphatidylinositol-3-kinase (PI3K) signal transduction pathway. In addition, leptin stimulated the phosphorylation of signal transducer and activator of transcription-3 (STAT3) Tyr-705 as well as estrogen receptor (ER)α, and the expression of ERα-responsive genes. Interestingly, all leptin-induced signal activation and gene expressions were blocked by the co-incubation with OB3 and the inhibition of extracellular signal-regulated kinase (ERK)1/2. Coincidently, leptin, but not OB3, increased circulating levels of follicle-stimulating hormone (FSH) which is known to play important roles in the initiation and proliferation of ovarian cancer cells. CONCLUSIONS: In summary, our findings suggest that the OB3 peptide may prevent leptin-induced ovarian cancer initiation and progression by disrupting leptin-induced proliferative signals via STAT3 phosphorylation and ERα activation. Therefore, the OB3 peptide is a potential anticancer agent that might be employed to prevent leptin-induced cancers in obese people.


Subject(s)
Gene Expression Regulation, Neoplastic , Leptin/genetics , Leptin/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/physiopathology , Peptide Fragments/metabolism , Signal Transduction/genetics , Animals , Cell Proliferation/genetics , Female , Humans , Male , Mice, Inbred BALB C , Mice, Nude
2.
Mediators Inflamm ; 2016: 6953459, 2016.
Article in English | MEDLINE | ID: mdl-27504055

ABSTRACT

Periodontitis, a chronic infection by periodontopathic bacteria, induces uncontrolled inflammation, which leads to periodontal tissue destruction. 2,3,5,4'-Tetrahydroxystilbene-2-O-beta-glucoside (THSG), a polyphenol extracted from Polygoni Multiflori, reportedly has anti-inflammatory properties. In this study, we investigated the mechanisms of THSG on the Porphyromonas gingivalis-induced inflammatory responses in human gingival fibroblasts and animal modeling of ligature-induced periodontitis. Human gingival fibroblast cells were treated with lipopolysaccharide (LPS) extracted from P. gingivalis in the presence of resveratrol or THSG to analyze the expression of TNF-α, IL-1ß, and IL-6 genes. Increased AMP-activated protein kinase (AMPK) activation and SirT1 expression were induced by THSG. Treatment of THSG decreased the expression of LPS-induced inflammatory cytokines, enhanced AMPK activation, and increased the expression of SirT1. In addition, it suppressed the activation of NF-κB when cells were stimulated with P. gingivalis LPS. The anti-inflammatory effect of THSG and P. Multiflori crude extracts was reproduced in ligature-induced periodontitis animal modeling. In conclusion, THSG inhibited the inflammatory responses of P. gingivalis-stimulated human gingival fibroblasts and ameliorated ligature-induced periodontitis in animal model.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Gingiva/cytology , Glucosides/pharmacology , Glucosides/therapeutic use , Periodontitis/drug therapy , Polygonaceae/chemistry , Stilbenes/pharmacology , Stilbenes/therapeutic use , Adult , Animals , Cells, Cultured , Drugs, Chinese Herbal/chemistry , Female , Fibroblasts/drug effects , Gingiva/drug effects , Gingiva/pathology , Glucosides/chemistry , Humans , Male , Rats , Rats, Sprague-Dawley , Stilbenes/chemistry , Young Adult
3.
Am J Physiol Cell Physiol ; 307(2): C150-61, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24808494

ABSTRACT

Thyroid hormones L-thyroxine (T4) and 3,3',5-triiodo-L-thyronine (T3) have been shown to initiate short- and long-term effects via a plasma membrane receptor site located on integrin αvß3. Also insulin-like growth factor type I (IGF-I) activity is known to be subject to regulation by this integrin. To investigate the possible cross-talk between T4 and IGF-I in rat L6 myoblasts, we have examined integrin αvß3-mediated modulatory actions of T4 on glucose uptake, measured through carrier-mediated 2-deoxy-[3H]-D-glucose uptake, and on cell proliferation stimulated by IGF-I, assessed by cell counting, [3H]-thymidine incorporation, and fluorescence-activated cell sorting analysis. IGF-I stimulated glucose transport and cell proliferation via the cell surface IGF-I receptor (IGFIR) and, downstream of the receptor, by the phosphatidylinositol 3-kinase signal transduction pathway. Addition of 0.1 nM free T4 caused little or no cell proliferation but prevented both glucose uptake and proliferative actions of IGF-I. These actions of T4 were mediated by an Arg-Gly-Asp (RGD)-sensitive pathway, suggesting the existence of crosstalk between IGFIR and the T4 receptor located near the RGD recognition site on the integrin. An RGD-sequence-containing integrin inhibitor, a monoclonal antibody to αvß3, and the T4 metabolite tetraiodothyroacetic acid all blocked the inhibition by T4 of IGF-I-stimulated glucose uptake and cell proliferation. Western blotting confirmed roles for activated phosphatidylinositol 3-kinase and extracellular regulated kinase 1/2 (ERK1/2) in the effects of IGF-I and also showed a role for ERK1/2 in the actions of T4 that modified the effects of IGF-I. We conclude that thyroid hormone inhibits IGF-I-stimulated glucose uptake and cell proliferation in L6 myoblasts.


Subject(s)
Cell Proliferation/drug effects , Glucose/metabolism , Insulin-Like Growth Factor I/metabolism , Integrin alphaVbeta3/metabolism , Myoblasts/metabolism , Thyroxine/metabolism , Animals , Biological Transport , Cell Line , Gene Expression Regulation/physiology , Insulin-Like Growth Factor I/genetics , Integrin alphaVbeta3/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Signal Transduction
4.
Histopathology ; 64(5): 713-21, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24117943

ABSTRACT

AIMS: In order to determine whether the expression of tumour-associated carbohydrate antigens (Tn/sTn) and a representative inflammation marker, nuclear factor-κB (NF-κB), is associated with the invasiveness of oral squamous cell carcinoma (OSCC), this study has attempted to investigate the correlation of the aforementioned markers with the well-established invasive pattern grading score (IPGS) and clinicopathological parameters. METHODS AND RESULTS: Specimens from 143 OSCC patients with classified clinicopathological parameters and IPGS were stained immunohistochemically using anti-Tn, sTn and NF-κB antibodies. Our results showed that the expression of both Tn and NF-κB was correlated positively with staging (P = 0.036; P = 0.015), recurrence (P < 0.001; P < 0.001) and distant metastasis (P = 0.005; P = 0.009), as well as with IPGS, while the expression of sTn was correlated inversely. In addition, poor survival was associated with overexpression of Tn and NF-κB but not with expression of sTn. CONCLUSIONS: Our results indicate that a reciprocal relationship between Tn and sTn expression may serve as a reliable indicator for OSCC prognostic evaluation. In addition, expression of Tn rather than sTn may play an important role in deeply invasive OSCC via regulation of NF-κB signalling.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , NF-kappa B/metabolism , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Signal Transduction
5.
FASEB J ; 27(8): 3209-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23640055

ABSTRACT

Thyroid hormone induces tumor cell and blood vessel cell proliferation via a cell surface receptor on heterodimeric integrin αvß3. We investigated the role of thyroid hormone-induced internalization of nuclear integrin αv monomer. Physiological concentration of thyroxine (free T4, 10(-10) M), but not 3,5,3'-triiodo-l-thyronine (T3), induced cellular internalization and nuclear translocation of integrin αv monomer in human non-small-cell lung cancer (H522) and ovarian carcinoma (OVCAR-3) cells. T4 did not complex with integrin αv monomer during its internalization. The αv monomer was phosphorylated by activated ERK1/2 when it heterodimerized with integrin ß3 in vitro. Nuclear αv complexed with transcriptional coactivator proteins, p300 and STAT1, and with corepressor proteins, NCoR and SMRT. Nuclear αv monomer in T4-exposed cells, but not integrin ß3, bound to promoters of specific genes that have important roles in cancer cells, including estrogen receptor-α, cyclooxygenase-2, hypoxia-inducible factor-1α, and thyroid hormone receptor ß1 in chromatin immunoprecipitation assay. In summary, monomeric αv is a novel coactivator regulated from the cell surface by thyroid hormone for the expression of genes involved in tumorigenesis and angiogenesis. This study also offers a mechanism for modulation of gene expression by thyroid hormone that is adjunctive to the nuclear hormone receptor (TR)-T3 pathway.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Integrin alpha5/metabolism , Promoter Regions, Genetic/genetics , Thyroid Hormones/pharmacology , Active Transport, Cell Nucleus/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Endocytosis/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Humans , Immunoblotting , Integrin alpha5/chemistry , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Integrin beta3/chemistry , Integrin beta3/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Protein Binding , Protein Multimerization , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , Thyroxine/pharmacology , Triiodothyronine/pharmacology , p300-CBP Transcription Factors/metabolism
6.
Endocr Relat Cancer ; 25(5): 533-545, 2018 05.
Article in English | MEDLINE | ID: mdl-29555649

ABSTRACT

Thyroid hormone, l-thyroxine (T4), has been shown to promote ovarian cancer cell proliferation via a receptor on plasma membrane integrin αvß3 and to induce the activation of ERK1/2 and expression of programmed death-ligand 1 (PD-L1) in cancer cells. In contrast, resveratrol binds to integrin αvß3 at a discrete site and induces p53-dependent antiproliferation in malignant neoplastic cells. The mechanism of resveratrol action requires nuclear accumulation of inducible cyclooxygenase (COX)-2 and its complexation with phosphorylated ERK1/2. In this study, we examined the mechanism by which T4 impairs resveratrol-induced antiproliferation in human ovarian cancer cells and found that T4 inhibited resveratrol-induced nuclear accumulation of COX-2. Furthermore, T4 increased expression and cytoplasmic accumulation of PD-L1, which in turn acted to retain inducible COX-2 in the cytoplasm. Knockdown of PD-L1 by small hairpin RNA (shRNA) relieved the inhibitory effect of T4 on resveratrol-induced nuclear accumulation of COX-2- and COX-2/p53-dependent gene expression. Thus, T4 inhibits COX-2-dependent apoptosis in ovarian cancer cells by retaining inducible COX-2 with PD-L1 in the cytoplasm. These findings provide new insights into the antagonizing effect of T4 on resveratrol's anticancer properties.


Subject(s)
Apoptosis/drug effects , B7-H1 Antigen/metabolism , Ovarian Neoplasms/drug therapy , Resveratrol/metabolism , Thyroxine/therapeutic use , Female , Humans , Ovarian Neoplasms/pathology , Thyroxine/pharmacology , Transfection
7.
Ann N Y Acad Sci ; 1403(1): 92-100, 2017 09.
Article in English | MEDLINE | ID: mdl-28759712

ABSTRACT

Nonpeptide hormones, such as thyroid hormone, dihydrotestosterone, and estrogen, have been shown to stimulate cancer proliferation via different mechanisms. Aside from their cytosolic or membrane-bound receptors, there are receptors on integrin αv ß3 for nonpeptide hormones. Interaction between hormones and integrin αv ß3 can induce signal transduction and eventually stimulate cancer cell proliferation. Resveratrol induces inducible COX-2-dependent antiproliferation via integrin αv ß3 . Resveratrol and hormone-induced signals are both transduced by activated extracellular-regulated kinases 1 and 2 (ERK1/2); however, hormones promote cell proliferation, while resveratrol induces antiproliferation in cancer cells. Hormones inhibit resveratrol-stimulated phosphorylation of p53 on Ser15, resveratrol-induced nuclear COX-2 accumulation, and formation of p53-COX-2 nuclear complexes. Subsequently, hormones impair resveratrol-induced COX-2-/p53-dependent gene expression. The inhibitory effects of hormones on resveratrol action can be blocked by different antagonists of specific nonpeptide hormone receptors but not integrin αv ß3 blockers. Results suggest that nonpeptide hormones inhibit resveratrol-induced antiproliferation in cancer cells downstream of the interaction between ligand and receptor and ERK1/2 activation to interfere with nuclear COX-2 accumulation. Thus, the surface receptor sites for resveratrol and nonpeptide hormones are distinct and can induce discrete ERK1/2-dependent downstream antiproliferation biological activities. It also indicates the complex pathways by which antiproliferation is induced by resveratrol in various physiological hormonal environments. .


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Estrogens/pharmacology , Signal Transduction/drug effects , Stilbenes/pharmacology , Thyroid Hormones/pharmacology , Animals , Phosphorylation/drug effects , Resveratrol
8.
Oncotarget ; 8(15): 24237-24249, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-27458161

ABSTRACT

Ovarian cancer is the leading cause of death in gynecological diseases. Thyroid hormone promotes proliferation of ovarian cancer cells via cell surface receptor integrin αvß3 that activates extracellular regulated kinase (ERK1/2). However, the mechanisms are still not fully understood. Thyroxine (T4) at a physiologic total hormone concentration (10-7 M) significantly increased proliferating cell nuclear antigen (PCNA) abundance in these cell lines, as did 3, 5, 3'-triiodo-L-thyronine (T3) at a supraphysiologic concentration. Thyroid hormone (T4 and T3) treatment of human ovarian cancer cells resulted in enhanced activation of the Ras/MAPK(ERK1/2) signal transduction pathway. An MEK inhibitor (PD98059) blocked hormone-induced cell proliferation but not ER phosphorylation. Knock-down of either integrin αv or ß3 by RNAi blocked thyroid hormone-induced phosphorylation of ERK1/2. We also found that thyroid hormone causes elevated phosphorylation and nuclear enrichment of estrogen receptor α (ERα). Confocal microscopy indicated that both T4 and estradiol (E2) caused nuclear translocation of integrin αv and phosphorylation of ERα. The specific ERα antagonist (ICI 182,780; fulvestrant) blocked T4-induced ERK1/2 activation, ERα phosphorylation, PCNA expression and proliferation. The nuclear co-localization of integrin αv and phosphorylated ERα was inhibited by ICI. ICI time-course studies indicated that mechanisms involved in T4- and E2-induced nuclear co-localization of phosphorylated ERα and integrin αv are dissimilar. Chromatin immunoprecipitation results showed that T4-induced binding of integrin αv monomer to ERα promoter and this was reduced by ICI. In summary, thyroid hormone stimulates proliferation of ovarian cancer cells via crosstalk between integrin αv and ERα, mimicking functions of E2.


Subject(s)
Estrogen Receptor alpha/metabolism , Integrin alphaVbeta3/metabolism , Ovarian Neoplasms/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Ovarian Neoplasms/pathology , Protein Binding
9.
Oncotarget ; 7(19): 27641-54, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27050378

ABSTRACT

Obesity results in increased secretion of cytokines from adipose tissue and is a risk factor for various cancers. Leptin is largely produced by adipose tissue and cancer cells. It induces cell proliferation and may serve to induce various cancers. OB3-leptin peptide (OB3) is a new class of functional leptin peptide. However, its mitogenic effect has not been determined. In the present study, because of a close link between leptin and the hypothalamic-pituitary-thyroid axis, OB3 was compared with leptin in different thyroid cancer cells for gene expression, proliferation and invasion. Neither agent stimulated cell proliferation. Leptin stimulated cell invasion, but reduced adhesion in anaplastic thyroid cancer cells. Activated ERK1/2 and STAT3 contributed to leptin-induced invasion. In contrast, OB3 did not affect expression of genes involved in proliferation and invasion. In vivo studies in the mouse showed that leptin, but not OB3, significantly increased circulating levels of thyrotropin (TSH), a growth factor for thyroid cancer. In summary, OB3 is a derivative of leptin that importantly lacks the mitogenic effects of leptin on thyroid cancer cells.


Subject(s)
Leptin/pharmacology , Peptide Fragments/pharmacology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Animals , Carbohydrate Metabolism/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Humans , Leptin/metabolism , Leptin/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Peptide Fragments/metabolism , Random Allocation , Signal Transduction , Thyroid Neoplasms/blood , Thyroid Neoplasms/genetics , Thyrotropin/blood
10.
Taiwan J Obstet Gynecol ; 53(4): 542-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25510698

ABSTRACT

OBJECTIVE: This study was conducted to explore the association between sexual orientations and polycystic ovary syndrome (PCOS)-related parameters. MATERIALS AND METHODS: A cross-sectional study with participants recruited from the regular outpatient clinic at the Department of Obstetrics and Gynecology at Taipei Medical University Hospital, Taipei, Taiwan between July 2012 and December 2013 was carried out. A total of 97 women met the criterion of having been diagnosed with PCOS. Among these 97 women, 89 were heterosexuals and eight were self-identified as lesbians. At the same time, 78 women without PCOS were enrolled to serve as the control group. Participants were given a standard questionnaire and had blood withdrawn for biochemical analysis of androgen parameters--including total testosterone, androstenedione, sex hormone binding globulin, free androgen index, 17ß-estradiol (E2), luteinizing hormone, and follicular-stimulating hormone. The biochemical data were measured to compare the PCOS clinical parameters present in people of different sexual orientations (lesbians and heterosexuals). RESULTS: The women with PCOS, regardless of sexual orientation, had higher percentages and serum levels of hyperandrogenism-related clinical parameters than the women without PCOS [acne (87.5% and 60.7% vs. 23.1%), p < 0.001]; hirsutism (62.5% and 57.3% vs. 15.4%, p ≤ 0.001)]; and biochemical parameters (total T, p < 0.05 or p < 0.001, and luteinizing hormone/follicular-stimulating hormone ratio, p ≤ 0.001]. The sexual orientation of women with PCOS affected their body mass index (BMI), because lesbians with PCOS possessed higher BMI than heterosexual women with PCOS (26.5 ± 1.9 vs. 22.5 ± 0.55; p < 0.05). However, hyperandrogenism-related clinical and biochemical parameters were not significantly different statistically between women with PCOS but of different sexual orientations. CONCLUSION: Our preliminary data showed that sexual orientation influenced the BMI of women with PCOS, but did not affect hyperandrogenism-related clinical or biochemical characteristics. This observation requires further confirmation.


Subject(s)
Heterosexuality/statistics & numerical data , Homosexuality, Female/statistics & numerical data , Polycystic Ovary Syndrome/psychology , Adult , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Hyperandrogenism/blood , Hyperandrogenism/diagnosis , Hyperandrogenism/etiology , Hyperandrogenism/psychology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/complications , Surveys and Questionnaires , Taiwan
11.
Oncotarget ; 5(24): 12891-907, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25436977

ABSTRACT

We have used a perfusion bellows cell culture system to investigate resveratrolinduced anti-proliferation/apoptosis in a human estrogen receptor (ER)-negative breast cancer cell line (MDA-MB-231). Using an injection system to perfuse media with stilbene, we showed resveratrol (0.5 - 100 µM) to decrease cell proliferation in a concentration-dependent manner. Comparison of influx and medium efflux resveratrol concentrations revealed rapid disappearance of the stilbene, consistent with cell uptake and metabolism of the agent reported by others. Exposure of cells to 10 µM resveratrol for 4 h daily × 6 d inhibited cell proliferation by more than 60%. Variable extracellular acid-alkaline conditions (pH 6.8 - 8.6) affected basal cell proliferation rate, but did not alter anti-proliferation induced by resveratrol. Resveratrol-induced gene expression, including transcription of the most up-regulated genes and pro-apoptotic p53-dependent genes, was not affected by culture pH changes. The microarray findings in the context of induction of anti-proliferation with brief daily exposure of cells to resveratrol-and rapid disappearance of the compound in the perfusion system-are consistent with existence of an accessible initiation site for resveratrol actions on tumor cells, e.g., the cell surface receptor for resveratrol described on integrin αvß3.


Subject(s)
Breast Neoplasms/drug therapy , Stilbenes/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Humans , Integrin alphaVbeta3/metabolism , Resveratrol , Signal Transduction
13.
PLoS One ; 6(4): e18988, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21526151

ABSTRACT

The ubiquitin-selective chaperone Cdc48, a member of the AAA (ATPase Associated with various cellular Activities) ATPase superfamily, is involved in many processes, including endoplasmic reticulum-associated degradation (ERAD), ubiquitin- and proteasome-mediated protein degradation, and mitosis. Although Cdc48 was originally isolated as a cell cycle mutant in the budding yeast Saccharomyces cerevisiae, its cell cycle functions have not been well appreciated. We found that temperature-sensitive cdc48-3 mutant is largely arrested at mitosis at 37°C, whereas the mutant is also delayed in G1 progression at 38.5°C. Reporter assays show that the promoter activity of G1 cyclin CLN1, but not CLN2, is reduced in cdc48-3 at 38.5°C. The cofactor npl4-1 and ufd1-2 mutants also exhibit G1 delay and reduced CLN1 promoter activity at 38.5°C, suggesting that Npl4-Ufd1 complex mediates the function of Cdc48 at G1. The G1 delay of cdc48-3 at 38.5°C is a consequence of cell wall defect that over-activates Mpk1, a MAPK family member important for cell wall integrity in response to stress conditions including heat shock. cdc48-3 is hypersensitive to cell wall perturbing agents and is synthetic-sick with mutations in the cell wall integrity signaling pathway. Our results suggest that the cell wall defect in cdc48-3 is exacerbated by heat shock, which sustains Mpk1 activity to block G1 progression. Thus, Cdc48-Npl4-Ufd1 is important for the maintenance of cell wall integrity in order for normal cell growth and division.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Cell Wall/metabolism , G1 Phase , Heat-Shock Response , Nucleocytoplasmic Transport Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Vesicular Transport Proteins/metabolism , Cyclins/metabolism , Endoplasmic Reticulum/metabolism , Enzyme Activation , Osmolar Concentration , Phosphorylation , Promoter Regions, Genetic/genetics , Saccharomyces cerevisiae/enzymology , Stress, Physiological , Temperature , Valosin Containing Protein
14.
J Virol ; 78(1): 378-88, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14671119

ABSTRACT

Dengue virus (DV) is a flavivirus and infects mammalian cells through mosquito vectors. This study investigates the roles of domain III of DV type 2 envelope protein (EIII) in DV binding to the host cell. Recombinant EIII interferes with DV infection to BHK21 and C6/36 cells by blocking dengue virion adsorption to these cells. Inhibition of EIII on BHK21 cells was broad with no serotype specificity; however, inhibition of EIII on C6/36 cells was relatively serotype specific. Soluble heparin completely blocks binding of EIII to BHK21 cells, suggesting that domain III binds mainly to cell surface heparan sulfates. This suggestion is supported by the observation that EIII binds very weakly to gro2C and sog9 mutant mammalian cell lines that lack heparan sulfate. In contrast, heparin does not block binding of EIII to mosquito cells. Furthermore, a synthetic peptide that includes amino acids (aa) 380 to 389 of EIII, IGVEPGQLKL, inhibits binding of EIII to C6/36 but not BHK21 cells. This peptide corresponds to a lateral loop region on domain III of E protein, indicating a possible role of this loop in binding to mosquito cells. In summary, these results suggest that EIII plays an important role in binding of DV type 2 to host cells. In addition, EIII interacts with heparan sulfates when binding to BHK21 cells, and a loop region containing aa 380 to 389 of EIII may participate in DV type 2 binding to C6/36 cells.


Subject(s)
Dengue Virus/pathogenicity , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cricetinae , Culicidae/virology , Dengue Virus/chemistry , Dengue Virus/classification , Models, Molecular , Receptors, Virus/metabolism , Serotyping , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL